Cardiac Tissue Engineering 2014th Milica Radisic
Lauren D Black Iii download
https://ebookbell.com/product/cardiac-tissue-engineering-2014th-
milica-radisic-lauren-d-black-iii-47742462
Explore and download more ebooks at ebookbell.com
Here are some recommended products that we believe you will be
interested in. You can click the link to download.
Cardiac Tissue Engineering 2nd Kareen Lk Coulombe Lauren D Black Iii
https://ebookbell.com/product/cardiac-tissue-engineering-2nd-kareen-
lk-coulombe-lauren-d-black-iii-47937244
Cardiac Regeneration And Repair Biomaterials And Tissue Engineering
Renke Li
https://ebookbell.com/product/cardiac-regeneration-and-repair-
biomaterials-and-tissue-engineering-renke-li-4653098
Cardiac Reconstructions With Allograft Tissues 1st Edition Richard A
Hopkins Md Auth
https://ebookbell.com/product/cardiac-reconstructions-with-allograft-
tissues-1st-edition-richard-a-hopkins-md-auth-4413200
Cardiac Reconstructions With Allograft Tissues Hopkins R A
https://ebookbell.com/product/cardiac-reconstructions-with-allograft-
tissues-hopkins-r-a-1098410
Cardiac Gene Therapy Methods And Protocols 2nd Edition Kiyotake
Ishikawa
https://ebookbell.com/product/cardiac-gene-therapy-methods-and-
protocols-2nd-edition-kiyotake-ishikawa-46073868
Cardiac Arrhythmias Pacing And Sudden Death Peter Kowey Jonathan P
Piccini
https://ebookbell.com/product/cardiac-arrhythmias-pacing-and-sudden-
death-peter-kowey-jonathan-p-piccini-48356682
Cardiac Mri Certification Exam Sadeer G Alkindi Scott E Janus
https://ebookbell.com/product/cardiac-mri-certification-exam-sadeer-g-
alkindi-scott-e-janus-48401156
Cardiac Surgical Complications Strategic Analysis And Clinical Review
Sandhya K Balaram
https://ebookbell.com/product/cardiac-surgical-complications-
strategic-analysis-and-clinical-review-sandhya-k-balaram-48949000
Cardiac Ultrasound 1st Edition Antoinette Kenny Kenny Antoinette
https://ebookbell.com/product/cardiac-ultrasound-1st-edition-
antoinette-kenny-kenny-antoinette-49200226
CardiacTissue
Engineering
Milica Radisic
Lauren D. Black III Editors
Methods and Protocols
Methods in
Molecular Biology 1181
ME T H O D S I N MO L E C U L A R BI O LO G Y
Series Editor
John M. Walker
School of Life Sciences
University of Hertfordshire
Hatfield, Hertfordshire, AL10 9AB, UK
For further volumes:
http://www.springer.com/series/7651
Cardiac Tissue Engineering
Methods and Protocols
Edited by
Milica Radisic
InstituteofBiomaterialsandBiomedicalEngineering,DepartmentofChemical
EngineeringandAppliedChemistry,UniversityofToronto,Toronto,ON,Canada;
TorontoGeneralResearchInstitute,UniversityHealthNetwork,Toronto,ON,Canada
Lauren D. Black III
DepartmentofBiomedicalEngineering,TuftsUniversity,Medford,MA,USA;
Cellular,MolecularandDevelopmentalBiologyProgram, Sackler School of Graduate
Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
Videos to this book can be accessed at http:/
/www.springerimages.com/videos/978-1-4939-1046-5
ISSN 1064-3745 ISSN 1940-6029 (electronic)
ISBN 978-1-4939-1046-5 ISBN 978-1-4939-1047-2 (eBook)
DOI 10.1007
/978-1-4939-1047-2
Springer New York Heidelberg Dordrecht London
Library of Congress Control Number: 2014940960
© Springer Science+Business Media New York 2014
This work is subject to copyright. All rights are reserved by the Publisher, whether the whole or part of the material is
concerned, specifically the rights of translation, reprinting, reuse of illustrations, recitation, broadcasting, reproduction
on microfilms or in any other physical way, and transmission or information storage and retrieval, electronic adaptation,
computer software, or by similar or dissimilar methodology now known or hereafter developed. Exempted from this
legal reservation are brief excerpts in connection with reviews or scholarly analysis or material supplied specifically for
the purpose of being entered and executed on a computer system, for exclusive use by the purchaser of the work.
Duplication of this publication or parts thereof is permitted only under the provisions of the Copyright Law of the
Publisher’s location, in its current version, and permission for use must always be obtained from Springer. Permissions
for use may be obtained through RightsLink at the Copyright Clearance Center. Violations are liable to prosecution
under the respective Copyright Law.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this publication does not
imply, even in the absence of a specific statement, that such names are exempt from the relevant protective laws and
regulations and therefore free for general use.
While the advice and information in this book are believed to be true and accurate at the date of publication, neither
the authors nor the editors nor the publisher can accept any legal responsibility for any errors or omissions that may be
made. The publisher makes no warranty, express or implied, with respect to the material contained herein.
Printed on acid-free paper
Humana Press is a brand of Springer
Springer is part of Springer Science+Business Media (www.springer.com)
Editors
Milica Radisic
Institute of Biomaterials
and Biomedical Engineering
Department of Chemical Engineering
and Applied Chemistry
University of Toronto
Toronto, ON, Canada
Toronto General Research Institute
University Health Network
Toronto, ON, Canada
Lauren D. Black III
Department of Biomedical Engineering
Tufts University
Medford, MA, USA
Cellular, Molecular and Developmental
Biology Program, Sackler School
of Graduate Biomedical Sciences
Tufts University School of Medicine
Boston, MA, USA
v
Human hearts have a limited regenerative potential, motivating the development of the
alternative treatment options for the conditions that result in the loss of beating cardio-
myocytes. An example is myocardial infarction that results in a death of tens of millions of
ventricular cardiomyocytes that cannot be replaced by the body. It is estimated that five to
seven million patients live with myocardial infarction in North America alone. A majority of
these patients do not need a surgical intervention, and medical management provides satisfac-
tory results. However, over a period of 5 years, one-half of the patients who experience a
myocardial infarction will develop heart failure, ultimately requiring heart transplantation.
The long-term goal of cardiac tissue engineering is to provide a living, beating, ideally
autologous, and non-immunogenic myocardial patch that can restore the contractile function
of the failing heart. The engineered tissues could also be used for preclinical drug testing to
discover new targets for cardiac therapy and eliminate drugs, cardiac and noncardiac, with
serious side effects. It generally involves a combination of suitable cell types, human or non-
human cardiomyocytes and supporting cells, with an appropriate biomaterial made out of
either synthetic or natural components and cultivation in an environment that reproduces
some of the complexity of the native cardiac environment (e.g., electrical, mechanical stimula-
tion, passive tension, or topographical cues).
This field is still young. The term cardiac tissue engineering usually refers to engineering
of myocardial wall in vitro using living and beating cardiomyocytes. The pioneering papers
appeared in the late 1990s, and they all utilized either neonatal rat cardiomyocytes or
embryonic chick cardiomyocytes as a cell source. Since then, the field has matured signifi-
cantly to include a range of approaches that all give cardiac tissues in vitro that are capable
of developing contractile force and propagating electrical impulses. Advances in human
embryonic stem cell research and induced pluripotent stem cell technology now provide
the possibility of generating millions of bona fide human cardiomyocytes. When research in
cardiac tissue engineering started some 25 years ago, the issue of a human cell source
appeared insurmountable; however the researchers continued to make way, and there are
many reports now on the use of human pluripotent stem cells as a source of cardiomyocytes
for cardiac tissue engineering. Although early researchers thought that having purified car-
diomyocytes in three-dimensional structures would be beneficial, based on analogies with
monolayer studies where fibroblasts overgrow cardiomyocytes, there is a consensus in the
field now that a mixed cell population is optimal for maintenance of cardiac phenotype and
survival of cardiomyocytes in engineered tissues both in vitro and in vivo. The mixed popu-
lation usually contains cardiomyocytes, endothelial cells, and a stromal cell type such as
fibroblasts or mesenchymal stem cells. Also, there is a consensus that a form of physical
stimulation, either mechanical or electrical, or passive tension is required for cardiomyo-
cytes to achieve and maintain a differentiated phenotype and in vivo-like functional proper-
ties during in vitro cultivation.
This book gathers for the first time a collection of protocols on cardiac tissue engineering
from pioneering and leading researchers around the globe. Protocols related to cell prepa-
ration, biomaterial preparation, cell seeding, and cultivation in various systems are provided.
Preface
vi
Our goal is to enable adoption of these protocols in laboratories that are interested in enter-
ing the field as well as enable transfer of knowledge between laboratories that are already in
this field. We hope that these efforts will lead to standardization, definition of best practices
in cardiac tissue cultivation, and direct comparison of various production protocols using
controlled in vivo studies that would ultimately lead to translational efforts. Although bio-
material patches alone and hydrogels have been investigated in clinical studies focused on
myocardial regeneration, a cardiac patch based on living, beating human cardiomyocytes
has not yet been tested in humans. Only patches based on non-cardiomyocytes have been
tested in humans with mixed results. Bringing a new therapy to the clinic is an overwhelm-
ing task, one that we must approach in a collaborative rather than competitive spirit. We
hope that sharing of the best protocols in cardiac tissue engineering will enable this goal.
Toronto, ON, Canada Milica Radisic
Medford, MA, USA Lauren D. Black III, Ph.D.
Preface
vii
Contents
Preface. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix
1 Second Generation Codon Optimized Minicircle (CoMiC)
for Nonviral Reprogramming of Human Adult Fibroblasts . . . . . . . . . . . . . . . 1
Sebastian Diecke, Leszek Lisowski, Nigel G. Kooreman,
and Joseph C. Wu
2 Scalable Cardiac Differentiation of Human Pluripotent Stem Cells
as Microwell-Generated, Size Controlled Three-Dimensional Aggregates . . . . 15
Celine L. Bauwens and Mark D. Ungrin
3 Preparation and Characterization of Circulating Angiogenic Cells
for Tissue Engineering Applications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
Aleksandra Ostojic, Suzanne Crowe, Brian McNeill,
Marc Ruel, and Erik J. Suuronen
4 Isolation and Expansion of C-Kit-Positive Cardiac Progenitor
Cells by Magnetic Cell Sorting . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 39
Kristin M. French and Michael E. Davis
5 Synthesis of Aliphatic Polyester Hydrogel for Cardiac Tissue Engineering . . . . 51
Sanjiv Dhingra, Richard D. Weisel, and Ren-Ke Li
6 Fabrication of PEGylated Fibrinogen: A Versatile Injectable
Hydrogel Biomaterial . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 61
Iris Mironi-Harpaz, Alexandra Berdichevski, and Dror Seliktar
7 Natural Cardiac Extracellular Matrix Hydrogels for Cultivation
of Human Stem Cell-Derived Cardiomyocytes . . . . . . . . . . . . . . . . . . . . . . . . 69
Donald O. Freytes, John D. O’Neill, Yi Duan-Arnold,
Emily A. Wrona, and Gordana Vunjak-Novakovic
8 Magnetically Actuated Alginate Scaffold: A Novel Platform
for Promoting Tissue Organization and Vascularization. . . . . . . . . . . . . . . . . . 83
Yulia Sapir, Emil Ruvinov, Boris Polyak, and Smadar Cohen
9 Shrink-Induced Biomimetic Wrinkled Substrates for Functional
Cardiac Cell Alignment and Culture . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 97
Nicole Mendoza, Roger Tu, Aaron Chen, Eugene Lee,
and Michelle Khine
10 Injectable ECM Scaffolds for Cardiac Repair . . . . . . . . . . . . . . . . . . . . . . . . . . 109
Todd D. Johnson, Rebecca L. Braden, and Karen L. Christman
11 Generation of Strip-Format Fibrin-Based Engineered
Heart Tissue (EHT) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 121
Sebastian Schaaf, Alexandra Eder, Ingra Vollert,
Andrea Stöhr, Arne Hansen, and Thomas Eschenhagen
viii
12 Cell Tri-Culture for Cardiac Vascularization . . . . . . . . . . . . . . . . . . . . . . . . . . 131
Ayelet Lesman, Lior Gepstein, and Shulamit Levenberg
13 Cell Sheet Technology for Cardiac Tissue Engineering . . . . . . . . . . . . . . . . . . 139
Yuji Haraguchi, Tatsuya Shimizu, Katsuhisa Matsuura,
Hidekazu Sekine, Nobuyuki Tanaka, Kenjiro Tadakuma,
Masayuki Yamato, Makoto Kaneko, and Teruo Okano
14 Design and Fabrication of Biological Wires . . . . . . . . . . . . . . . . . . . . . . . . . . . 157
Jason W. Miklas, Sara S. Nunes, Boyang Zhang, and Milica Radisic
15 Collagen-Based Engineered Heart Muscle. . . . . . . . . . . . . . . . . . . . . . . . . . . . 167
Malte Tiburcy, Tim Meyer, Poh Loong Soong,
and Wolfram-Hubertus Zimmermann
16 Creation of a Bioreactor for the Application of Variable
Amplitude Mechanical Stimulation of Fibrin Gel-Based
Engineered Cardiac Tissue . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 177
Kathy Y. Morgan and Lauren D. Black III
17 Preparation of Acellular Myocardial Scaffolds with Well-Preserved
Cardiomyocyte Lacunae, and Method for Applying Mechanical
and Electrical Simulation to Tissue Construct . . . . . . . . . . . . . . . . . . . . . . . . . 189
Bo Wang, Lakiesha N. Williams, Amy L. de Jongh Curry,
and Jun Liao
18 Patch-Clamp Technique in ESC-Derived Cardiomyocytes . . . . . . . . . . . . . . . . 203
Jie Liu and Peter H. Backx
19 Optogenetic Control of Cardiomyocytes via Viral Delivery . . . . . . . . . . . . . . . 215
Christina M. Ambrosi and Emilia Entcheva
20 Methods for Assessing the Electromechanical Integration
of Human Pluripotent Stem Cell-Derived Cardiomyocyte Grafts. . . . . . . . . . . 229
Wei-Zhong Zhu, Dominic Filice, Nathan J. Palpant,
and Michael A. Laflamme
21 Quantifying Electrical Interactions Between Cardiomyocytes
and Other Cells in Micropatterned Cell Pairs. . . . . . . . . . . . . . . . . . . . . . . . . . 249
Hung Nguyen, Nima Badie, Luke McSpadden, Dawn Pedrotty,
and Nenad Bursac
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 263
Contents
ix
CHRISTINA M. AMBROSI, PH.D. • Department of Biomedical Engineering, Institute for
Molecular Cardiology, Stony Brook University, Stony Brook, NY, USA
PETER H. BACKX • Department of Physiology and Medicine, University of Toronto, Toronto,
ON, Canada; The Heart and Stroke/Richard Lewar Centre of Excellence, Toronto, ON,
Canada; Division of Cardiology, University Health Network, Toronto, ON, Canada
NIMA BADIE • Department of Biomedical Engineering, Duke University, Durham, NC, USA
CELINE L. BAUWENS • Centre for Commercialization of Regenerative Medicine, Toronto,
ON, Canada
ALEXANDRA BERDICHEVSKI • Faculty of Biomedical Engineering, Technion—Israel Institute
of Technology, Haifa, Israel
LAUREN D. BLACK III, PH.D. • Department of Biomedical Engineering, Tufts University,
Medford, MA, USA; Cellular, Molecular and Developmental Biology Program, Sackler
School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA,
USA
REBECCA L. BRADEN, M.S. • Department of Bioengineering, University of California San Diego,
La Jolla, CA, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
NENAD BURSAC, PH.D. • Department of Biomedical Engineering, Duke University,
Durham, NC, USA
AARON CHEN • Department of Chemical Engineering and Materials Science, University of
California, Irvine, CA, USA
KAREN L. CHRISTMAN, PH.D. • Department of Bioengineering, University of California
San Diego, La Jolla, CA, USA; Sanford Consortium for Regenerative Medicine, La Jolla,
CA, USA
SMADAR COHEN, PH.D. • Avram and Stella Goldstein-Goren Department of Biotechnology
Engineering, The Center for Regenerative Medicine and Stem Cell (RMSC) Research,
Ben-Gurion University of the Negev, Beer-Sheva, Israel; The Ilse Katz Institute for
Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
SUZANNE CROWE • Division of Cardiac Surgery, University of Ottawa Heart Institute,
Ottawa, ON, Canada
MICHAEL E. DAVIS, PH.D. • Wallace H. Coulter Department of Biomedical Engineering,
Emory University and Georgia Institute of Technology, Atlanta, GA, USA
AMY L. DE JONGH CURRY • Department of Biomedical Engineering, University of Memphis,
Memphis, TN, USA
SANJIV DHINGRA • Regenerative Medicine Program, Institute of Cardiovascular Sciences,
St. Boniface Research Centre, University of Manitoba, Winnipeg, MB, Canada
SEBASTIAN DIECKE • Lorry I. Lokey Stem Cell Research Building, Stanford University School
of Medicine, Stanford, CA, USA
YI DUAN-ARNOLD • Department of Biomedical Engineering, Columbia University,
New York, NY, USA
ALEXANDRA EDER • Department of Experimental Pharmacology and Toxicology, University
Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; DZHK (German
Centre for Cardiovascular Research), Hamburg, Germany
Contributors
x
EMILIA ENTCHEVA, PH.D. • Department of Biomedical Engineering, Institute for Molecular
Cardiology, Stony Brook University, Stony Brook, NY, USA
THOMAS ESCHENHAGEN • Department of Experimental Pharmacology and Toxicology,
University Medical CenterHamburg-Eppendorf (UKE), Hamburg, Germany;
DZHK (German Centre for Cardiovascular Research), Hamburg, Germany
DOMINIC FILICE • Department of Bioengineering, Institute for Stem Cells and Regenerative
Medicine, University of Washington, Seattle, WA, USA
KRISTIN M. FRENCH • Wallace H. Coulter Department of Biomedical Engineering,
Emory University and Georgia Institute of Technology, Atlanta, GA, USA
DONALD O. FREYTES • New York Stem Cell Foundation, New York, NY, USA
LIOR GEPSTEIN • The Sohnis Family Research Laboratory for Cardiac Electrophysiology
and Regenerative medicine, the Bruce Rappaport Faculty of Medicine, Technion—Israel
Institute of Technology, Haifa, Israel
ARNE HANSEN • Department of Experimental Pharmacology and Toxicology, University
Medical CenterHamburg-Eppendorf (UKE), Hamburg, Germany; DZHK (German
Centre for Cardiovascular Research), Hamburg, Germany
YUJI HARAGUCHI • Institute of Advanced Biomedical Engineering and Science, TWIns,
Tokyo Women’s Medical University, Shinjuku-ku, Tokyo, Japan
TODD D. JOHNSON, M.S. • Department of Bioengineering, University of California San Diego,
La Jolla, CA, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
MAKOTO KANEKO • Department of Mechanical Engineering, Graduate School of
Engineering, Osaka University, Yamadaoka, Suita, Japan
MICHELLE KHINE, PH.D. • Department of Biomedical Engineering, University of
California, Irvine, CA, USA; Department of Chemical Engineering, University of
California, Irvine, CA, USA
NIGEL G. KOOREMAN • Lorry I. Lokey Stem Cell Research Building, Stanford University
School of Medicine, Stanford, CA, USA
MICHAEL A. LAFLAMME, M.D., PH.D. • Department of Pathology, Institute for Stem Cells
and Regenerative Medicine, University of Washington, Seattle, WA, USA
EUGENE LEE • Department of Biomedical Engineering, University of California, Irvine,
CA, USA
AYELET LESMAN • Department of Chemistry and Chemical Engineering, California
Institute of Technology, Pasadena, CA, USA; Department of Biomedical Engineering,
Technion—Israel Institute of Technology, Haifa, Israel
SHULAMIT LEVENBERG • Department of Biomedical Engineering, Technion—Israel Institute
of Technology, Haifa, Israel
REN-KE LI, M.D., PH.D. • Division of Cardiovascular Surgery, Toronto General Research
Institute, University Health Network, Toronto, ON, Canada; Division of Cardiac
Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
JUN LIAO, PH.D. • Tissue Bioengineering Laboratory, Department of Biological
Engineering, Mississippi State University, Mississippi State, MS, USA
LESZEK LISOWSKI • Gene Transfer, Targeting and Therapeutics Facility, Salk Institute
for Biological Studies, San Diego, CA, USA
JIE LIU • Department of Physiology and Medicine, University of Toronto, Toronto, ON,
Canada; Materials Science, University of California, Irvine, CA, USA
KATSUHISA MATSUURA • Institute of Advanced Biomedical Engineering and Science, TWIns,
Tokyo Women’s Medical University, Shinjuku-ku, Tokyo, Japan
BRIAN MCNEILL • Division of Cardiac Surgery, University of Ottawa Heart Institute,
Ottawa, ON, Canada
Contributors
xi
LUKE MCSPADDEN • Department of Biomedical Engineering, Duke University, Durham,
NC, USA
NICOLE MENDOZA • Department of Biomedical Engineering, University of California,
Irvine, CA, USA
TIM MEYER • Institute of Pharmacology, Heart Research Center Göttingen (HRCG),
University Medical Center Göttingen, Göttingen, Germany; DZHK (German Center
for Cardiovascular Research), partner site Göttingen, Göttingen, Germany
JASON W. MIKLAS • Institute of Biomaterials and Biomedical Engineering, University
of Toronto, Toronto, ON, Canada
IRIS MIRONI-HARPAZ • Faculty of Biomedical Engineering, Technion—Israel Institute
of Technology, Haifa, Israel
KATHY Y. MORGAN • Department of Biomedical Engineering, Tufts University, Medford,
MA, USA
HUNG NGUYEN • Department of Biomedical Engineering, Duke University, Durham,
NC, USA
SARA S. NUNES • Institute of Biomaterials and Biomedical Engineering, University of
Toronto, Toronto, ON, Canada; Toronto General Research Institute, University Health
Network, Toronto, ON, Canada
TERUO OKANO • Institute of Advanced Biomedical Engineering and Science, TWIns,
Tokyo Women’s Medical University, Shinjuku-ku, Tokyo, Japan
JOHN D. O’NEILL • Department of Biomedical Engineering, Columbia University,
New York, NY, USA
ALEKSANDRA OSTOJIC • Division of Cardiac Surgery, University of Ottawa Heart Institute,
Ottawa, ON, Canada
NATHAN J. PALPANT • Department of Pathology, Institute for Stem Cells and Regenerative
Medicine, University of Washington, Seattle, WA, USA
DAWN PEDROTTY • Department of Biomedical Engineering, Duke University, Durham,
NC, USA
BORIS POLYAK, PH.D. • Department of Surgery, Drexel University College of Medicine,
Philadelphia, PA, USA; Department of Pharmacology and Physiology, Drexel University
College of Medicine, Philadelphia, PA, USA
MILICA RADISIC • Institute of Biomaterials and Biomedical Engineering, Department of
Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON,
Canada; Toronto General Research Institute, University Health Network, Toronto,
ON, Canada
MARC RUEL • Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa,
ON, Canada
EMIL RUVINOV, PH.D. • Avram and Stella Goldstein-Goren Department of Biotechnology
Engineering, Ben-Gurion University of the Negev, Beer-Sheva, Israel
YULIA SAPIR, M.SC. • Avram and Stella Goldstein-Goren Department of Biotechnology
Engineering, Ben-Gurion University of the Negev, Beer-Sheva, Israel
SEBASTIAN SCHAAF • Department of Experimental Pharmacology and Toxicology,
University Medical CenterHamburg-Eppendorf (UKE), Hamburg, Germany; DZHK
(German Centre for Cardiovascular Research), Hamburg, Germany
HIDEKAZU SEKINE • Institute of Advanced Biomedical Engineering and Science, TWIns,
Tokyo Women’s Medical University, Shinjuku-ku, Tokyo, Japan
DROR SELIKTAR • Faculty of Biomedical Engineering, Technion—Israel Institute of
Technology, Haifa, Israel; Nanoscience and Nanotechnology Initiative, National
University of Singapore, Singapore, Singapore
Contributors
xii
TATSUYA SHIMIZU • Institute of Advanced Biomedical Engineering and Science, TWIns,
Tokyo Women’s Medical University, Shinjuku-ku, Tokyo, Japan
POH LOONG SOONG • Institute of Pharmacology, Heart Research Center Göttingen
(HRCG), University Medical Center Göttingen, Göttingen, Germany; DZHK (German
Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany
ANDREA STÖHR • Department of Experimental Pharmacology and Toxicology, University
Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; DZHK (German
Centre for Cardiovascular Research), Hamburg, Germany
ERIK J. SUURONEN • Division of Cardiac Surgery, University of Ottawa Heart Institute,
Ottawa, ON, Canada
KENJIRO TADAKUMA • Department of Mechanical Engineering, Graduate School of Engineering,
Osaka University, Yamadaoka, Suita, Japan
NOBUYUKI TANAKA • Institute of Advanced Biomedical Engineering and Science, TWIns,
Tokyo Women’s Medical University, Shinjuku-ku, Tokyo, Japan
MALTE TIBURCY • Institute of Pharmacology, Heart Research Center Göttingen (HRCG),
University Medical Center Göttingen, Göttingen, Germany; DZHK (German Center for
Cardiovascular Research), partner site Göttingen, Göttingen, Germany
ROGER TU • Department of Biological Sciences, University of California, Irvine, CA, USA
MARK D. UNGRIN • Department of Comparative Biology and Experimental Medicine,
Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
INGRA VOLLERT • Department of Experimental Pharmacology and Toxicology, University
Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; DZHK (German
Centre for Cardiovascular Research), Hamburg, Germany
GORDANA VUNJAK-NOVAKOVIC • Department of Biomedical Engineering, Columbia
University, New York, NY, USA
BO WANG • Tissue Bioengineering Laboratory, Department of Biological Engineering,
Mississippi State University, Mississippi State, MS, USA; Department of Surgery,
Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine,
Northwestern University, Chicago, IL, USA
RICHARD D. WEISEL • Division of Cardiovascular Surgery, Toronto General Research
Institute, University Health Network, Toronto, ON, Canada; Department of Surgery,
Division of Cardiac Surgery, University of Toronto, Toronto, ON, Canada
LAKIESHA N. WILLIAMS • Tissue Bioengineering Laboratory, Department of Biological
Engineering, Mississippi State University, Mississippi State, MS, USA
EMILY A. WRONA • New York Stem Cell Foundation, New York, NY, USA
JOSEPH C. WU, M.D., PH.D. • Lorry I. Lokey Stem Cell Research Building, Stanford
University School of Medicine, Stanford, CA, USA
MASAYUKI YAMATO • Institute of Advanced Biomedical Engineering and Science, TWIns,
Tokyo Women’s Medical University, Shinjuku-ku, Tokyo, Japan
BOYANG ZHANG • Department of Chemical Engineering and Applied Chemistry, University
of Toronto, Toronto, ON, Canada; Institute of Biomaterials and Biomedical Engineering,
University of Toronto, Toronto, ON, Canada
WEI-ZHONG ZHU • Department of Pathology, Institute for Stem Cells and Regenerative
Medicine, University of Washington, Seattle, WA, USA
WOLFRAM-HUBERTUS ZIMMERMANN, M.D. • Institute of Pharmacology, Heart Research
Center Göttingen (HRCG), University Medical Center Göttingen, Göttingen, Germany;
DZHK (German Center for Cardiovascular Research), partner site Göttingen,
Göttingen, Germany
Contributors
1
Milica Radisic and Lauren D. Black III (eds.), Cardiac Tissue Engineering: Methods and Protocols, Methods in Molecular Biology,
vol. 1181, DOI 10.1007/978-1-4939-1047-2_1, © Springer Science+Business Media New York 2014
Chapter 1
Second Generation Codon Optimized Minicircle (CoMiC)
for Nonviral Reprogramming of Human Adult Fibroblasts
Sebastian Diecke, Leszek Lisowski, Nigel G. Kooreman,
and Joseph C. Wu
Abstract
The ability to induce pluripotency in somatic cells is one of the most important scientific achievements in
the fields of stem cell research and regenerative medicine. This technique allows researchers to obtain
pluripotent stem cells without the controversial use of embryos, providing a novel and powerful tool for
disease modeling and drug screening approaches. However, using viruses for the delivery of reprogram-
ming genes and transcription factors may result in integration into the host genome and cause random
mutations within the target cell, thus limiting the use of these cells for downstream applications. To overcome
this limitation, various non-integrating techniques, including Sendai virus, mRNA, minicircle, and plasmid-
based methods, have recently been developed. Utilizing a newly developed codon optimized 4-in-1
minicircle (CoMiC), we were able to reprogram human adult fibroblasts using chemically defined media
and without the need for feeder cells.
Key words Human induced pluripotent stem cells (hiPSC), Minicircle, Reprogramming, Disease
modeling, Chemically defined, Differentiation
1 Introduction
Somatic cells, such as human adult fibroblasts, can be reverted to an
embryonic stem cell (ESC)-like state by inducing the expression of
specific genes and transcription factors that regulate the mainte-
nance of pluripotency in embryonic stem cells [1]. These so-called
core factors of pluripotency, namely OCT4, SOX2, NANOG, and
two oncogenes (c-MYC and LIN28), initiate the resetting of the
epigenetic profile of a terminal differentiated somatic cell and acti-
vate the molecular circuitry of pluripotency [2–4]. The resultant
induced pluripotent stem cells (iPSCs) have similar morphology,
growth characteristics, and gene expression profiles as hESCs [5].
Furthermore, the capacity of iPSCs to differentiate into all other cell
types of the human body has opened up new clinical opportunities
for developing successful personalized stem cell-based therapies
2
and drug discovery approaches. Initially iPSCs were produced
using retroviral and lentiviral vectors that are known to integrate
into the chromosomes of the target cells, potentially causing dis-
ruption of gene transcription and subsequent tumor formation
[6]. Therefore, various laboratories have developed additional
non-integrating reprogramming techniques such as Sendai virus,
mRNA, and protein- or DNA-based methods [7–11].
In order to induce the transformation of a nullipotent somatic
cell into a pluripotent state, it is necessary to express the right stoi-
chiometry of the reprogramming factors within the same trans-
fected cell [12]. One elegant approach to achieve this requirement
uses polycistronic constructs in which the four reprogramming
factors are controlled by one promoter separated by a different
self-cleaving 2a peptide sequence [13]. The disadvantage of such
polycistronic plasmids is their large size, which might decrease
the transfection efficiency in the adult human fibroblast. To over-
come this problem, we cloned the polycistronic expression cassette
into a minicircle plasmid backbone. Minicircles are special episomal
DNA vectors devoid of any bacterial plasmid backbone (Fig. 1a).
Therefore minicircles are significantly smaller than standard plasmids,
minicircle after induction unpurified
minicircle after induction purified
1 kb plus ladder
2 kb
7 kb
chromosomal bacterial DNA
1
2
3
1 2 3
1 2 3 5
4
1
2
3
minicircle (Nde1 digest)
Parental minicircle backbone (HindIII digest)
Parental minicircle backbone (Nde1 digest)
4
5
minicircle (HindIII digest)
Parental minicircle backbone (Nde1/HindIII digest)
2 kb
7 kb
Before Arabinose Induction
a
b c
After Arabinose Induction
Parental 4-in-1
CoMiC
11.6 kb
4-in-1 CoMiC
7.6 kb
Fig. 1 (a) Map of parental 4-in-1 reprogramming minicircle before and after arabinose induction. (b) Control
digest to verify quality of the minicircle production. (c) Gel showing bands representing the minicircle after
induction, both before and after purification steps
Sebastian Diecke et al.
3
which in turn enhances their transfection efficiency and the survival
rate of the target cells [14]. Furthermore, it has been shown that
minicircle constructs sustain a higher expression rate of their
harboring transgenes over a longer period of time. Although the
minicircle theoretically should not integrate into the genome of
the target cell, there is still a relatively small chance of integration.
Therefore, it is important to screen the derived iPSCs for random
integration using specific PCR primers or Southern blot experi-
ments [15].
The following protocol details the entire process of repro-
gramming human adult fibroblasts into human iPSCs: from disso-
ciating fibroblasts from patient skin biopsy samples, to the
reprogramming and characterization of the newly derived iPSC
line. The time from induction of pluripotency to expansion of the
subsequent iPSC line to passage 3 is approximately 1 month
(Fig. 2b). The workflow of the reprogramming process is summa-
rized in Fig. 3. The final part of the protocol explains the pluripo-
tency of the iPSCs is confirmed using immunostaining (Fig. 4a).
However, it will also be necessary to perform further experiments,
including directed differentiation into specific germ layers and
teratoma formation assay, to verify pluripotency (Fig. 4b, c) [16].
It should also be mentioned that the minicircle reprogramming
Transfection using unpurified minicircle
a
b
Low transfection efficiency, high cell death Higher transfection efficiency, less cell death
Transfection using purified minicircle
0 3 13 16 20 day
Reprogramming timeline
Phase
Tomato
TRA-1-60
iPSC colony on Matrigel
30
Fig.2 (a) Representative bright field and fluorescent (Tomato) images demonstrating the transfection efficiency
and cell survival of unpurified and purified minicircle. (b) Timeline showing the expected phase contrast and
fluorescent images at various stages of the reprogramming process.At day 20, pluripotency was confirmed by
TRA-1-60 immunofluorescent staining
iPSC Generation Using a Codon Optimized Minicircle Plasmid
4
technique still has a very low efficiency and therefore should be
only used by researchers with iPSC experience as an alternative
method for obtaining integration-free induced pluripotent stem
cells, or if it is necessary to compare different reprogramming
techniques.
1 3 10
Fibroblast-Media E8-Media
20
12 day
+ Nab + AA
Transfection and seeding cells
onto 2 Matrigel plates
Picking of individual iPSC
clones (use 10 µM Rock
inhibitor Y27632)
7 9 11 13
5
0
-1
E7-Media
split 1.1 x106
fibroblast onto a new dish
50%
50%
Fig. 3 Timeline for reprogramming adult human fibroblasts into induced pluripotent stem cells using CoMiC
Fig. 4 Immunofluorescence staining demonstrating (a) pluripotency markers NANOG, OCT4 and TRA-1-81,
DAPI, and bright field images. (b) Immunofluorescent staining of iPSC-derived cardiomyocytes demonstrating
positive staining of cardiac markers connexin 43 and cardiac troponin as well as DAPI. (c) Representative H&E
staining from teratomas to confirm the pluripotency status of the generated iPSCs
Sebastian Diecke et al.
5
2 Materials
1. Fibroblast media: Low glucose Dulbecco’s Modified Eagle Media
(DMEM low glucose) with 20 % fetal bovine serum (FBS).
2. iPSC media: Essential 8 (combine supplement with base
media) and Essential 7 (Essential 6 combined with base media
and supplemented with 100 ng/mL FGF2).
1. Phosphate buffered saline (PBS).
2. Collagenase IV.
3. Penicillin/streptomycin.
4. Lennox L Broth (LB) base.
5. Sodium hydroxide (NaOH) solution, 1 M.
6. L-(+)-Arabinose ≥99 %, distilled water (dH2O).
7. Kanamycin sulfate, 100×.
8. Matrigel—Growth factor reduced, hESC-qualified.
9. DMEM/F12.
10. TrypLE Express, store at room temperature (RT), there is no
need to warm to 37 °C before use.
11. 0.25 % Trypsin/Ethylenediaminetetraacetic acid (EDTA).
12. Accutase.
13. Y27632 (ROCK inhibitor): 10 mM stock in Ultrapure water,
store at −20 °C.
14. Ultrapure water.
15. Sodium butyrate (NaB).
16. Antibodies (see Table 1).
17. 4 % paraformaldehyde (PFA) solution.
18. Triton-X 100.
19. Bovine serum albumin (BSA).
20. Goat Serum.
21. Tween-20.
22. Vectashield mounting medium with DAPI.
23. Qiagen Plasmid Plus Maxi Kit.
24. DNAase (2,000 U/ml).
25. Gelatin.
26. Neon Buffer R.
27. Qiagen Plasmid Plus Maxi kit.
28. 4 % Paraformaldehyde (PFA).
29. New England biolabs enzymes HindIII and Nde1.
2.1 Media
2.2 Reagents
iPSC Generation Using a Codon Optimized Minicircle Plasmid
6
Table
1
List
of
antibodies
and
dilutions
used
for
characterization
of
human
induced
pluripotent
stem
cells
and
differentiated
cardiomyocytes.
Staining
protocols
were
optimized
with
the
antibodies
from
the
listed
suppliers.
Optimization
may
be
required
if
different
antibodies
are
used
Antigen
Supplier
Product
no.
Dilution
Raised
in
Secondary
(1:400)
Supplier
Product
no.
Connexin-43
SIGMA
C6219
1:400
Rabbit
Alexa
Fluor
®
488
Goat
Anti-Rabbit
IgG
(H
+
L)
Life
Technologies
A-11037
c
Troponin-T
Thermo
Scientifi
c
MS-295-P
1:400
Mouse
Alexa
Fluor
®
488
Goat
Anti-Mouse
IgG
(H
+
L)
Life
Technologies
A-11001
NANOG
Santa
Cruz
SC-33759
1:100
Rabbit
Alexa
Fluor
®
488
Goat
Anti-Rabbit
IgG
(H
+
L)
Life
Technologies
A-11037
OCT4
Santa
Cruz
SC-9081
1:100
Rabbit
Alexa
Fluor
®
488
Goat
Anti-Rabbit
IgG
(H
+
L)
Life
Technologies
A-11037
Tra-1-81
Millipore
mab4381
1:250
Mouse
Alexa
Fluor
®
488
Goat
Anti-Mouse
IgG
(H
+
L)
Life
Technologies
A-11001
Sebastian Diecke et al.
7
1. 6-well cell culture plates (surface area=9.8 cm2
) coated with
2 mL Matrigel.
2. 12-well cell culture plates (surface area=3.8 cm2
) coated with
1 mL Matrigel.
3. 10 cm tissue culture plates (surface area=58.95 cm2
) coated
with 12 mL Matrigel.
4. 90 mm petri dishes.
5. 15 and 50 mL polypropylene conical tubes.
6. 2 mL plastic aspiration pipettes.
7. 5, 10, 25, and 50 mL plastic pipettes.
8. 250 and 500 mL polyethersulfone (PES) media filters.
9. 8-chamber slides.
10. T225 flasks.
11. Coverslips.
12. Parafilm.
13. Countess Cell Counter, slides, and trypan blue.
14. Tissue culture incubator capable of 37 °C, 5 % CO2, and 85 %
relative humidity (such as New Brunswick Galaxy 170R).
15. Dual gas tissue culture incubator capable of 37 °C, 5 % CO2,
5 % O2, and 85 % relative humidity with split inner door.
16. Centrifuge.
17. Inverted tissue culture microscope (such as Nikon Ti) with
heated stage.
18. Autoclave.
19. Orbital shaker.
20. Spectrophotometer.
21. Waterbath.
22. Neon Transfection device Life Technologies.
23. Nalgene centrifuge bottle.
3 Methods
1. Prepare 1,200 mL of LB Broth media by adding 24 g of LB
Broth Base to 1,200 mL of distilled water (hH2O) and divide
this over the three culture flasks.
2. Autoclave the flasks using the “liquid cycle.”
3. After cooling down of the flasks to room temperature, add
4 mL of Kanamycin 100× to each one of the culture flasks.
4. In the morning, pick one clone from a parental plasmid trans-
formed bacterial plate in 5 mL LB media containing kanamycin
2.3 Equipment
3.1 Minicircle
Production
iPSC Generation Using a Codon Optimized Minicircle Plasmid
8
(50 μg/mL) and grow this pre-culture until the evening
at 37 °C with shaking at 250 rpm.
5. In the evening, grow an overnight culture by combining
100 μL of the pre-culture with each of the flasks containing
media with kanamycin and culture overnight (16–18 h) at
37 °C with shaking at 250 rpm (see Note 1).
6. The optical density (OD, absorbance) of the overnight culture
should be between 3.75 and 4.25.
7. Prepare a minicircle induction mix comprising 1 volume of
fresh LB Broth (1,200 mL), 4 % 1 N NaOH (48 mL), and 1 %
L-Arabinose (12 g) and mix well.
8. Add 400 mL of the induction mix to each of the three flasks
with the overnight culture and mix well.
9. Incubate the combined mix at 32 °C with shaking at 250 rpm
for 6–8 h.
10. Harvest all the culture flasks, transfer them to centrifuge
bottle, and pellet the bacteria (6,000×g).
11. Isolate minicircles according to the Qiagen Plasmid Plus Maxi
kit instructions with certain modifications (see Notes 2 and 3).
12. Confirm the successful minicircle induction by digestion using
HindIII and Nde1. Nde1 linearizes only the parental back-
bone but not the minicircle. Therefore, you should expect one
band around 7.7 kb when digesting the minicircle with both
enzymes (Fig. 1b).
1. All of these steps should be performed in a sterile tissue culture
hood. Prepare a fibroblast digestion solution by dissolving
1 mg/mL Collagenase IV in DMEM/F12 and sterile filter it
through a 0.2 μm pore size filter.
2. Transfer the skin punch biopsy into a 15 mL conical tube con-
taining 3 mL PBS+penicillin/streptomycin (1:100), repeat
this step three times.
3. Transfer the tissue into a sterile 10 cm plate and add 1 mL
Collagenase IV. Use two scalpels to cross cut the tissue into small
pieces while keeping the tissue immersed in Collagenase IV.
4. Additionally, add 2 mL Collagenase IV solution to the minced
tissue and pipette it up and down. Transfer the Collagenase
IV/tissue suspension into a 15 mL conical tube and add 25 μL
of 0.5 M EDTA and 20 μL of DNase. Afterwards, incubate the
mixture for 3 h in a tissue culture incubator at 37 °C.
5. Following the 3-h incubation time, pipette the tissue pieces up
and down several times to obtain single-cell fibroblasts.
6. Add 12 mL fibroblast media containing 1× penicillin/
streptomycin to stop the Collagenase IV digestion.
3.2 Fibroblast
Isolation from
a Patient Skin
Punch Biopsy
Sebastian Diecke et al.
9
7. Centrifuge for 4 min at 300×g.
8. Resuspend the pellet in 2 mL fibroblast media containing 1×
penicillin/streptomycin solution, and plate it in one well of a
0.1 % gelatin-coated 6-well plate. Depending on the size of
your tissue sample, you may need to plate the cells in more
than one well of a 6-well plate. To prepare gelatin-coated
plates, gelatin stock (10 %) is dissolved in PBS, 1 ml of this
solution is pipetted into a well of a 6-well plate and incubated
for 1 h at 37 °C.
9. Incubate the cells for at least 4 days without changing the
media, until some of the fibroblasts start to attach and propa-
gate. After this, change the media every other day.
10. Once the cells become confluent, split using 0.25 % Trypsin–
EDTA for 10 min. Plate the cells onto a 0.1 % gelatin-coated
T225 flask to further expand and freeze multiple vials of fibro-
blast after the flask becomes confluent.
1. Thaw stock bottle of Matrigel at 4 °C overnight.
2. Keep Matrigel on ice, and add 300 μL Matrigel to 50 mL of
cold DMEM/F12 media. Add 2 mL to each well of a 6-well
plate. (Alternatively use one Matrigel aliquot per 20 mL cold
DMEM/F12 media and cover four 10 cm plates using 5 mL.)
3. Place the plates in the fridge and allow the Matrigel to polym-
erize overnight.
1. The target fibroblast cell line should be thawed and cultured
for at least 3 days before starting the reprogramming experi-
ment. Additionally, it is beneficial to further purify the minicir-
cle DNA using the Zymoclean Gel DNA Recovery Kit to
eliminate residual chromosomal bacterial DNA of the minicir-
cle preparation (Fig. 1c). This step will increase the cell survival
after the electroporation (Fig. 2a).
2. Plate 1.1×106
human fibroblast in a 10 cm tissue culture plate
1 day before starting the reprogramming experiment.
3. On the following day (day 0), trypsinize the cells using Trypsin
0.25 % EDTA for 10 min to obtain a single cell suspension.
4. Wash off the trypsin using 10 mL of DMEM plus 20 % FBS
and centrifuge for 3 min at 300×g.
5. Aspirate the media, and dissolve the cell pellet in 100–105 μL
Neon Buffer R (the volume depends on the amount of plasmid
you have to use to get 10–12 μg DNA). The final volume of
buffer, DNA, and cells should be around 110 μL.
6. Perform the transfection using a 100 μL Neon tip and the
following settings: 1,600 V, 10 ms, and 3 pulses (transfection
efficiency should be more than 50 %).
3.3 Prepare Matrigel
Plates
3.4 Reprogramming
of Human Fibroblasts
Using Neon
Transfection
and CoMiC
iPSC Generation Using a Codon Optimized Minicircle Plasmid
10
7. Plate the cells equally distributed onto two Matrigel-coated
plates (5.5×105
cells) in fibroblast media.
8. On the following morning (day 1), change the media to
fibroblast media and add sodium butyrate (0.2 mM) plus
50 μg/mL ascorbic acid.
9. On day 3, change the media to fibroblast media and add
sodium butyrate (0.2 mM) plus 50 μg/mL ascorbic acid.
10. On day 5, change the media to 50:50 fibroblast media:Essential
7 media and add sodium butyrate (0.2 mM) plus 50 μg/mL
ascorbic acid.
11. On days 7–13, change the media every other day until the first
pre-iPSC colonies appear.
Change the media to Essential 7 and add sodium butyrate
(0.2 mM) plus 50 μg/mL ascorbic acid.
12. Between days 14 and 20, after you recognize the first colonies,
switch to Essential 8 media and hypoxic culture conditions.
Around day 20, the iPSC colonies should be big enough for
manual picking under the microscope. Pick six individual iPSC
clones using the Vitrolife stem cell cutting tool with a 10 μL tip
and transfer them into six different wells of a Matrigel-coated
plate with Essential 8 media in presence of 10 μM ROCK
Inhibitor (Y-27632). If it is difficult to detach the iPSC colonies
from the surrounding fibroblasts, you also can scrape those
fibroblasts away to make room for the outgrowing iPSC colony.
After 2 days, the iPSC clone should be large enough to pick.
1. After 5–7 days, the individual colonies become big enough to
dissociate into single cells using 0.5 mL TrypLE for 8 min.
Detach the remaining colonies by pipetting them up and down
with a 1 mL pipette.
2. Wash the cells once with 5 mL Essential 8 media.
3. Centrifuge for 3 min at 300×g.
4. Passage the cells into one well of a Matrigel-coated 6-well plate
containing 2 mL Essential 8 media.
5. After 5–7 days, colonies will have grown out and become dense
enough to split onto a Matrigel-coated 10 cm plate. Therefore
incubate the cells with 1 mL Accutase for 8 min, wash using
9 mL Essential 8 media, and centrifuge for 3 min at 300×g.
6. Seed the cells onto a Matrigel-coated 10 cm plate in Essential
8 media in the presence of 10 μM Y27632.
7. Once the 10 cm plate become confluent, freeze down 5 vials
(passage 3 iPS cells) and continue to grow at a 1:6 split ratio,
freezing vials every 10 passages.
8. Grow cells to at least passage 5 to test for the expression of the
pluripotency marker genes and to passage 15 before testing for
efficient differentiation.
3.5 Colony
Purification
and Expansion
Sebastian Diecke et al.
11
1. Coat 8-chamber slides with Matrigel (200 μL per chamber) as
described above in Subheading 3.3.
2. Seed iPSCs (1×104
cells/chamber) or differentiated cardio-
myocytes (1.75×104
cells/chamber) as a negative control, on
8-chamber slides in a final volume of 200 μL of the appropriate
medium.
3. Place the chamber slides in 90 mm Petri dishes and store in a
37 °C incubator.
4. Allow cells to attach and grow for ~24–72 h and fix as follows
(see Note 4).
5. Aspirate medium and wash cells 3× with PBS.
6. Add to each chamber 100 μL 4 % Paraformaldehyde (PFA) in
PBS for 20 min at RT.
7. Aspirate PFA and wash 3× with 200 μL PBS.
8. Aspirate PBS and replace with 500 μL of fresh PBS.
9. Seal slides with parafilm and place at 4ºC until needed for
staining (keep for up to 7 days).
10. Incubate with 100 μL of permeabilization solution (0.2 %
Triton X-100 in PBS), for 15 min at room temperature
(see Note 5).
11. Wash twice with PBS, for 5 min at room temperature.
12. Incubate with blocking solution (2 % BSA or 4 % Goat Serum
or 2 % FBS in PBS), for 1 h at room temperature or overnight
at 4ºC.
13. Wash once in PBS, for 5 min at room temperature.
14. Incubate overnight at 4 °C with primary antibody (Table 1)
diluted appropriately in blocking solution (100 μL per
chamber).
15. Wash in 1 % goat serum, 0.1 % Tween-20 in PBS four times,
for 10 min.
16. Incubate with the appropriate secondary antibody diluted in
blocking solution, for 1 h at RT (100 μL per chamber). Wrap
in aluminum foil as soon as secondary antibody is added.
17. Wash in 1 % Goat Serum, 0.1 % Tween-20 in PBS four times,
for 10 min.
18. Remove the chambers from the slide.
19. Place a small amount of Vectashield mounting medium with
DAPI over the area where each chamber used to be.
20. Place a clean coverslip over the slide and gently blot off any
liquid remaining after the staining protocol.
21. Seal with nail varnish.
22. Store slides at 4 °C in the dark.
23. Image at least 3 h later.
3.6 Immunofluores-
cence Staining
Protocol
iPSC Generation Using a Codon Optimized Minicircle Plasmid
12
1. Takahashi K et al (2007) Induction of pluripo-
tent stem cells from adult human fibroblasts by
defined factors. Cell 131(5):861–872
2. Mitalipov S, Wolf D (2009) Totipotency, pluri-
potency and nuclear reprogramming. Adv
Biochem Eng Biotechnol 114:185–199
3. Feng B, Ng JH, Heng JC, Ng HH (2009)
Molecules that promote or enhance reprogram-
ming of somatic cells to induced pluripotent
stem cells. Cell Stem Cell 4(4):301–312
4. Maherali N et al (2007) Directly repro-
grammed fibroblasts show global epigenetic
remodeling and widespread tissue contribu-
tion. Cell Stem Cell 1(1):55–70
5. Wernig M et al (2007) In vitro reprogramming
of fibroblasts into a pluripotent ES-cell-like
state. Nature 448(7151):318–324
6. Sommer CA et al (2010) Excision of reprogram-
ming transgenes improves the differentiation
potential of iPS cells generated with a single
excisable vector. Stem Cells 28(1):64–74
7. Stadtfeld M, Hochedlinger K (2010) Induced
pluripotency: history, mechanisms, and appli-
cations. Genes Dev 24(20):2239–2263
8. Stadtfeld M, Nagaya M, Utikal J, Weir G,
Hochedlinger K (2008) Induced pluripotent
stem cells generated without viral integration.
Science 322(5903):945–949
4 Notes
1. Do not start off with too much parental plasmid, as an
overgrown culture will limit the induction efficiency.
2. To ensure a high yield, make sure that the pellet is completely
resuspended in Qiagen Maxi Kit P1 buffer and use the same
volume of P2 and S3. For example, when using 400 mL spin-
ning tubes, resuspend the pellet in 10 mL P1, P2, and S3 buf-
fer instead of 8 mL.
3. To avoid bacterial genomic DNA contamination, avoid exces-
sive shaking of the resuspended pellet when adding buffer P2.
4. Solutions should be made fresh for each experiment. Keep at
4ºC until day 2.
5. The permeabilization step is only required for the detection of
nuclear protein (e.g., OCT4, SOX2, or NANOG); when sur-
face markers are detected, skip the permeabilization step and
go straight to the primary antibody incubation.
Acknowledgements
We thank Nicholas Mordwinkin for proof-reading the manuscript.
We thank funding support from the National Institutes of Health
(NIH) R01 HL113006, NIH U01 HL099776, NIH R24
HL117756, and the American Heart Association Established
Investigator Award (JCW). This work was also supported by DFG
(German Research Foundation) (to S.D.).
References
Sebastian Diecke et al.
13
9. Warren L et al (2010) Highly efficient repro-
gramming to pluripotency and directed
differentiation of human cells with synthetic
modified mRNA. Cell Stem Cell 7(5):
618–630
10. Zhou H et al (2009) Generation of induced
pluripotent stem cells using recombinant pro-
teins. Cell Stem Cell 4(5):381–384
11. Okita K et al (2011) A more efficient method
to generate integration-free human iPS cells.
Nat Methods 8(5):409–412
12. Tiemann U et al (2011) Optimal reprogram-
ming factor stoichiometry increases colony
numbers and affects molecular characteristics
of murine induced pluripotent stem cells.
Cytometry A 79(6):426–435
13. Warlich E et al (2011) Lentiviral vector design
and imaging approaches to visualize the early
stages of cellular reprogramming. Mol Ther
19(4):782–789
14. Jia F et al (2010) A nonviral minicircle vector
for deriving human iPS cells. Nat Methods
7(3):197–199
15. Gonzalez F, Boue S, Izpisua Belmonte JC
(2011) Methods for making induced pluripo-
tent stem cells: reprogramming a la carte. Nat
Rev Genet 12(4):231–242
16. Lan F et al (2013) Abnormal calcium handling
properties underlie familial hypertrophic car-
diomyopathy pathology in patient-specific
induced pluripotent stem cells. Cell Stem Cell
12(1):101–113
iPSC Generation Using a Codon Optimized Minicircle Plasmid
15
Milica Radisic and Lauren D. Black III (eds.), Cardiac Tissue Engineering: Methods and Protocols, Methods in Molecular Biology,
vol. 1181, DOI 10.1007/978-1-4939-1047-2_2, © Springer Science+Business Media New York 2014
Chapter 2
Scalable Cardiac Differentiation of Human Pluripotent
Stem Cells as Microwell-Generated, Size Controlled
Three-Dimensional Aggregates
Celine L. Bauwens and Mark D. Ungrin
Abstract
The formation of cells into more physiologically relevant three-dimensional multicellular aggregates is an
important technique for the differentiation and manipulation of stem cells and their progeny. As industrial
and clinical applications for these cells increase, it will be necessary to execute this procedure in a readily
scalable format. We present here a method employing microwells to generate large numbers of human
pluripotent stem cell aggregates and control their subsequent differentiation towards a cardiac fate.
Key words Cardiac differentiation, Microwells, Human pluripotent stem cells, Size controlled cell
aggregates, Forced cell aggregation
1 Introduction
It has been known for a long time that cellular behavior in two-
dimensional adherent culture does not fully recapitulate three-
dimensional in vivo systems, and consequently there is a growing
trend in many fields of research to employ three-dimensional tissue
constructs. These may be divided into scaffolded constructs, where
cells are cultured on or in an exogenous supporting matrix [1], and
unscaffolded systems that consist primarily or exclusively of cells
(which may then proceed to generate their own endogenous matrices)
[2, 3]. Here, we will discuss a process for generating large numbers
of uniform, unscaffolded aggregates of human pluripotent stem
cells for differentiation towards a cardiac fate.
Conventionally, size controlled cellular aggregates have been
generated in microcentrifuge tubes [4], as hanging drops [5], by
micropatterning human embryonic stem cells (hESCs) colonies
[6] or by centrifugation into U- or V-bottom plates [7, 8]; how-
ever throughput is limited using these approaches. Microwell-
based systems are similar in concept to V-bottom plates; however
16
the smaller size of the microwells permits very large numbers of
uniform aggregates to be generated from a single culture-plate
well in a mechanically simple system [9]. This approach has been
used for a variety of applications including differentiation of plu-
ripotent stem cells to ectodermal [10], endodermal [11], mesoder-
mal [12], and extraembryonic [13] fates; chondrogenesis from
mesenchymal stem cells [14]; and generation of uniform substrates
for toxicological screening [15] and investigations of mechanobi-
ology [16].
Using this approach to control aggregate size, we consequently
determine surface area to volume ratio, which in turn modulates
extraembryonic endoderm (ExE) commitment. Cardiogenic sig-
nals from this cell lineage then specify subsequent cardiomyogen-
esis [12].
2 Materials
All reagent preparation and cell culture handling should be carried
out in a biological safety cabinet under sterile conditions.
1. Biological safety cabinet.
2. Pipette aid.
3. Serological pipettes (5–25 mL).
4. Aspirator.
5. Aspirator or Pasteur pipettes.
6. 15 and 50 mL conical tubes.
7. Fume hood.
8. 0.22 μm syringe filter.
9. 5 % CO2, 5 % O2, and humidity controlled cell culture incubator
(Subheadings 3.1 and 3.4).
10. 5 % CO2, 20 % O2, and humidity controlled cell culture
incubator (Subheading 3.5).
11. Low speed centrifuge with a swinging bucket rotor fitted with
a plate holder.
12. P2, P20, P200, and P1000 micropipettors with appropriate tips.
13. Inverted microscope with 4×, 10×, and 20× phase objectives.
14. Ultra-Low Attachment (ULA) 24 well plates.
15. 1.5 mL microcentrifuge tubes.
16. Bench-top microcentrifuge.
1. L-Ascorbic Acid: Prepare a stock solution of 5 mg/mL in 4ºC,
sterile ultrapure distilled water. Leave on ice and vortex
2.1 General
Equipment
and Supplies
2.2 Basal Medium
Celine L. Bauwens and Mark D. Ungrin
17
periodically until completely dissolved. Filter-sterilize using a
0.22 μm syringe filter, aliquot at 1 mL volume, and store the
Ascorbic Acid aliquots at −20ºC. Use a freshly thawed aliquot
each time medium is prepared.
2. Monothioglycerol: MTG should be aliquoted (1 mL) and
stored frozen (−20ºC). When aliquots are thawed, they can be
used for 3 months and then discarded. Aliquoting of MTG is
strongly recommended as it minimizes the amount of oxida-
tion due to repeated opening. On the day of media prepara-
tion, dilute 13 μL MTG in 1 mL StemPro-34. Discard unused
diluted MTG.
3. Transferrin: Transferrin (stock concentration: 30 mg/mL)
should be aliquoted (1 mL aliquots) and stored at −20ºC.
4. All cytokines are stored lyophilized at −20ºC until ready to be
aliquoted in the following buffers and at the following concen-
trations, Table 1. Continue to store unused aliquots at −20 °C.
5. 4 mM Hydrochloric acid (HCl), 0.1 % BSA Buffer: In a fume
hood, transfer 30 μL of 6.0 N HCl solution to 50 mL of ultra-
pure distilled water. Filter-sterilize the solution using a 0.22 μm
syringe filter. Add 2 mL of 25 % BSA solution to the 50 mL
HCl solution.
6. PBS, 0.1 % BSA Buffer: Add 20 μL of 25 % BSA solution for
every mL PBS.
7. Supplemented DMEM/F-12: 1 % Penicillin/Streptomycin
(Pen/Strep) and 1 % L-glutamine in DMEM/F-12.
8. hESC Wash Medium: 5 % Knockout Serum Replacement in
Supplemented DMEM/F12.
Table 1
Cytokines and buffers used for media preparation
Cytokine Buffer
Stock Conc.
(μg/mL)
Human Bone morphogenetic protein
4 (hBMP-4)
4 mM Hydrochloric acid, 0.1 % BSA 10
Human Fibroblast Growth Factor 2
(basic) (hbFGF)
Phosphate buffered saline (PBS),
0.1 % BSA
10
Human Vascular Endothelial Growth
Factor (hVEGF)
Phosphate buffered saline (PBS),
0.1 % BSA
5
Activin A Phosphate buffered saline (PBS),
0.1 % BSA
10
Human Dickkopf 1 homolog (hDkk-1) Phosphate buffered saline (PBS),
0.1 % BSA
50
Scalable Cardiac Differentiation of Human Pluripotent Stem Cells…
18
9. StemPro-34: StemPro-34 is sold as a kit with two components.
The supplement is kept at −20ºC and the liquid media at 4ºC.
When combined, the media is stable for 30 days (see Note 1).
StemPro-34 is always used with the supplement added in this
protocol.
10. Modified StemPro-34: StemPro-34 modified with the addi-
tion of 1 % Pen/Strep, 1 % L-glutamine, 0.5 % Transferrin, 1 %
Ascorbic Acid, 0.3 % MTG. This medium should be prepared
on the day of use with freshly thawed Ascorbic Acid.
1. One 6 well plate culture of feeder-free or feeder-depleted (see
Note 2) hESCs adapted to single cell passaging (e.g., enzy-
matic dissociation with TrypLE).
2. TrypLE Select.
3. hESC Wash Medium (preparation previously described in
Subheading 2.2, item 8).
4. Hemocytometer.
5. Trypan Blue.
6. Aggrewell™ 400 plates (StemCell Technologies 27845).
7. Aggrewell™ Rinsing Solution (StemCell Technologies 07010).
8. Aggregation Medium: Modified StemPro-34, 0.5 ng/mL
BMP4, 10 μM Y-27632 ROCK Inhibitor. 1 mL of medium is
required per well of a 24 well Aggrewell™ 400 plate.
9. OPTIONAL: 37 μm strainer.
1. Stage 1 Induction Medium: Modified StemPro-34, 10 ng/mL
BMP4, 5 ng/mL bFGF, 6 ng/mL Activin A. 1 mL of medium
is required per well of a 24 well Aggrewell™ 400 plate
(see Note 3).
2. Stage 2 Induction Medium: Modified StemPro-34, 10 ng/mL
VEGF, 150 ng/mL DKK1. 1 mL of medium is required per
well of a 24 well ULA plate (see Note 3).
3. Stage 3 Induction Medium: Modified StemPro-34, 10 ng/mL
VEGF, 150 ng/mL DKK1, 5 ng/mL bFGF. 1 mL of medium
is required per well of a 24 well ULA plate (see Note 3).
4. hESC Wash Medium (prepared as previously described in
Subheading 2.2, item 8).
1. 1 mg/mL Collagenase Type II in Hank’s Balanced Salt
Solution.
2. TrypLE Select.
3. hESC Wash Medium (preparation previously described in
Subheading 2.2, item 8).
2.3 hESC Aggregate
Formation
Components
2.4 Cardiac
Induction Components
2.5 Flow Cytometry
Celine L. Bauwens and Mark D. Ungrin
19
4. DNase: DNase should be diluted in ultrapure distilled water to
a concentration of 1 mg/mL, aliquoted, and stored at −20 °C.
5. 96 well assay plate (or 1.5 mL microcentrifuge tubes).
6. IntraPrep fixation and permeabilization kit.
7. Cardiac Troponin T antibody.
8. Allophycocyanin (APC)-conjugated goat-anti-mouse IgG
antibody.
9. HF buffer solution: 2 % Fetal Bovine Serum (FBS) in Hank’s
Balanced Salt Solution.
10. 5 mL round-bottom flow cytometry analysis tubes.
11. Flow cytometer with the appropriate laser and filter to excite
(i.e., 633–640 nm) and detect (i.e., 660/20 nm) APC.
3 Methods
A schematic of the process is shown in Fig. 1.
1. Add 0.5 mL Aggrewell™ Rinsing Solution to each well of the
Aggrewell™ plate used. Ensure that the solution contacts the
entire surface inside each microwell by centrifuging the plate at
840×g for 2 min. Incubate the Aggrewell™ plate containing
the Rinsing Solution for 30–60 min at room temperature.
Aspirate the solution and wash twice with 1 mL PBS per well,
centrifuging at 840×g for 2 min after each PBS addition.
2. Completely dissociate the hESCs to single cells: Aspirate the cul-
ture medium from each culture well, rinse each well with 1 mL
TrypLE Select, and aspirate the TrypLE Select. Residual TrypLE
should be sufficient to dissociate the cells. Incubate the plate at
37 °C for 3 min, then add 1 mL of hESC Wash Medium to each
well and mechanically dissociate the cells from the tissue cul-
ture surface by pipetting with a P1000 micropipettor. Transfer
the cells (in hESC Wash Medium) to a 15 mL conical tube.
3.1 hESC Aggregate
Formation
Fig. 1 Schematic of cardiac differentiation protocol. The sequence of steps in
Subheadings 3.1–3.6 is diagrammed, along with an indication of the timescale
over which they occur
Scalable Cardiac Differentiation of Human Pluripotent Stem Cells…
20
Optionally, if the dissociation appears to be incomplete and cell
clumps remain, the suspension may be passed through a strainer
at this point.
3. Perform a cell count: Take a 10 μL sample of the cell suspen-
sion collected in the previous step. Add 30 μL of Trypan Blue
and mix the suspension well by pipetting up and down. Transfer
10 μL of Trypan Blue-stained cells to each chamber of a hemo-
cytometer and visualize under the inverted microscope with
the 10× objective. Count the cells.
4. From the cell count results, calculate how many wells can be
seeded at 1.2×106
cells per well. Resuspend the dissociated
hESCs in Aggregation Medium at a density of 1.2×106
cells/mL
(see Note 4).
5. Aspirate the PBS wash solution from each well on the
Aggrewell™ Plate. Using a P1000 micropipettor, evenly dis-
tribute 1 mL of the cell suspension to each well on the
Aggrewell™ Plate. If seeding a large numbers of wells, periodi-
cally mix the container of cell suspension to prevent settling.
Centrifuge the plate at 200×g for 5 min. Visualize the plate
under the microscope to confirm cells have spun to the bottom
of each microwell (see Note 5).
6. Incubate the plate for 24 h at 37 °C in a 5 % CO2, 5 % O2
(hypoxic) incubator.
1. Prepare the required volume of Stage 1 Induction Medium
(for a 24 well plate, volume=1 mL×number of wells). Place
the medium in a 37 °C water bath for at least 15 min.
2. Remove the Aggrewell™ plate from the incubator. Inspect
the aggregates under the microscope. Compared to immedi-
ately post-centrifuge aggregation, they should appear intact
with smooth edges (more round and less square than the pre-
vious day).
3. To remove the supernatant but retain the aggregates in their
individual microwells: Use a P1000 micropipettor. For each
well on the Aggrewell™ plate, place the tip of the micropipettor
at the surface of the culture medium in the well and against the
edge of the well (keep the plate level horizontally). Slowly
remove the medium, being careful not to disturb the aggre-
gates at the bottom of the microwells. Once the medium level
has been reduced to about 1–2 mm from the surface of the
textured microwell surface, the plate can be slowly tilted to
collect medium at one side of the well (once the volume is this
low, fluid motion is greatly reduced and it is easier to avoid
aggregates getting lifted out of their individual microwells).
Slowly pipette out the remaining medium in the well.
4. To add fresh Stage 1 Induction Medium while ensuring the
aggregates remain in their individual microwells: Take up 1 mL
3.2 Cardiac
Induction Stage 1
(24±2 h Following
Aggregation)
Celine L. Bauwens and Mark D. Ungrin
21
of Stage 1 Induction Medium with a P1000 micropipettor.
Hold the pipette tip against the inside edge of the well and
very slowly dispense the medium against the inside wall of the
well. Repeat for the remaining wells.
5. Return the plate to the incubator under hypoxic conditions for
4 days.
Stage 2 Induction directs mesoderm differentiation. Therefore at
this time-point it is very important to remove all residual Activin A
from the Stage 1 Induction Medium. Activin A is a very potent
signaling molecule and at this stage of differentiation, even trace
levels would promote differentiation to the endoderm lineage at
the expense of cardiac induction. Therefore, at this stage the aggre-
gates are removed from the Aggrewell™ plates, washed well, and
transferred to bulk aggregate cultures in 24 well ULA plates.
1. Place hESC Wash Medium (volume=number of wells×2 mL)
in a 37 °C water bath for 10–15 min.
2. Prepare the necessary volume of Stage 2 Induction medium
(volume=number of wells×1 mL) and place in a 37 °C water
bath for 10–15 min.
3. Using a 5 mL serological pipette, harvest the aggregates from
each well of the Aggrewell™ plate, and collect the aggregate
suspension in a 15 mL conical tube (up to 10 wells per tube).
4. Allow the aggregates to settle for 15 min in hypoxic (5 % O2)
conditions. This step is performed to separate single cells and
cellular debris from the intact aggregates. Aspirate the super-
natant carefully and resuspend the aggregates in 10 mL hESC
Wash Medium to wash out residual inductive cytokines
(especially Activin A which is a potent signaling molecule even
at very low concentrations).
5. Centrifuge the aggregates at 50×g for 2 min and aspirate the
supernatant.
6. Resuspend the pelleted aggregates in the Induction 2 Medium
prepared in step 2 above.
7. Dispense the aggregate suspension into a 24 well ULA plate at
1 mL per well. Visualize the aggregates under the microscope.
They should appear as uniform, tight cell clusters.
8. Incubate under hypoxic conditions until day 8.
Typically, in a successful cardiac induction, spontaneously con-
tracting aggregates will be observed under the microscope between
day 8 and day 12.
1. Prepare the necessary volume of Stage 3 Induction Medium
(volume=number of wells×2 mL) and place in a 37ºC water
bath for 10–15 min.
3.3 Cardiac
Induction Stage 2
(Day 4: 96±2 h After
Aggregate Formation)
3.4 Cardiac
Induction Stage 3
(Day 8: 192±2 h After
Aggregate Formation)
Scalable Cardiac Differentiation of Human Pluripotent Stem Cells…
22
2. Use a 5 mL serological pipette to transfer the aggregates to
15 mL conical tubes, pooling up to 10 mL of aggregates per
tube. Allow 10 min for the aggregates to settle. Aspirate the
supernatant and resuspend the aggregates in Stage 3 Induction
Medium. Using a 5 mL serological pipette, redistribute the
aggregates into a 24 well ULA plate at 1 mL per well.
3. Incubate under hypoxic conditions until day 12.
1. Prepare the necessary volume of Stage 3 Induction Medium
(volume=number of wells×1 mL) and place in a 37ºC water
bath for 10–15 min.
2. Use a 5 mL serological pipette to transfer the aggregates to
15 mL conical tubes, pooling up to 10 mL of aggregates per
tube. Allow 10 min for the aggregates to settle. Aspirate the
supernatant and resuspend the aggregates in Stage 3 Induction
Medium. Using a 5 mL serological pipette, redistribute the
aggregates into a 24 well ULA plate at 1 mL per well.
3. Incubate the cells at normoxic oxygen levels for the remainder
of the culture period (37ºC, 20 % O2, 5 % CO2). After this time
point, cells are no longer cultured under hypoxic conditions.
4. Repeat this complete medium exchange (steps 1–3) at day 16.
5. Cardiac aggregates are ready for harvest on day 20.
For flow cytometry analysis of cardiac Troponin T (cTnT) expres-
sion frequency, it is recommended that 4 wells of day 20 cardiac
aggregates be harvested.
1. Using a 5 mL serological pipette, transfer 1 well of aggregates
to a 15 mL conical tube. Centrifuge the aggregates at 50×g
for 2 min and carefully aspirate the supernatant.
2. Add 1 mL of freshly dissolved 1 mg/ml collagenase Type II
solution to the aggregates, transfer the aggregate suspension
to a 1.5 mL microcentrifuge tube, and incubate the aggre-
gates in collagenase Type II overnight at room temperature.
3. The next day, using a P1000 micropipettor, gently pipette the
aggregates to dissociate them into a homogenous single cell
suspension. If the aggregates do not readily dissociate, settle
the aggregates, aspirate the supernatant, and incubate the
aggregates in 700 μL TrypLE for 1–2 min at room tempera-
ture. Gently pipette the aggregates 1–2 times with a P1000
micropipette to dissociate. Add 700 μL hESC Wash Medium
containing 14 μL of 1 mg/mL DNAse stock solution to dilute
the TrypLE. Take a 10 μL sample for cell counting, and centrifuge
the remaining suspension using a bench-top microcentri-
fuge for 2 min at 300×g.
4. Stain the 10 μL counting sample with an equal volume of
Trypan Blue and count using a hemocytometer.
3.5 Cardiac
Induction Stage 3
(Day 12 to Harvest)
3.6 Flow Cytometry
Analysis of Cardiac
Troponin T Expression
Frequency
of Aggrewell™
Culture Output
Celine L. Bauwens and Mark D. Ungrin
23
5. Remove the 1.5 mL microcentrifuge tube containing the
remaining cells from the microcentrifuge. Aspirate the super-
natant and resuspend the cells in HF at a concentration of
200,000–500,000 cells per 100 μL.
6. Transfer 100 μL per well to 2 wells of a 96 well plate (see Note 6)
per experimental condition or replicate (one well will be for
cTnT staining and the other will be the unstained control).
7. Pellet by centrifuging the plate at 300×g for 2 min in a
swinging bucket centrifuge.
8. Fix the cells in 200 μL of Intraprep Reagent 1 per well for
15 min at room temperature.
9. Centrifuge the plate at 300×g for 2 min. Carefully aspirate the
supernatant and dispose of in a paraformaldehyde waste
container.
10. Wash the fixed cells twice: Add 200 μL HF to each well.
Centrifuge the plate at 300×g for 2 min, aspirate the superna-
tant, and repeat the wash step once more. Fixed cells can be
stored for up to 1 week in HF at 4 °C.
11. Permeabilize the cells: Centrifuge the plate at 300×g for
2 min. Add 100 μL of Intraprep Reagent 2 per well and incu-
bate the plate for 5 min at room temperature. Centrifuge the
plate at 300×g for 2 min and aspirate the supernatant.
12. Prepare a staining solution of anti-cTnT (Neomarkers MS-295)
in HF at the optimal concentration for the given lot number
(determined by titration: typically ranges from 1:500 to
1:2,000 dilution).
13. Add 100 μL staining solution to one well of cells and 100 μL
of plain HF to the other well (negative control). Incubate the
cells for 30 min at 4 °C.
14. Centrifuge the cells at 300×g for 2 min. Aspirate the superna-
tant and add 200 μL HF per well. Repeat the wash step one
more time.
15. Prepare a master mix of the secondary antibody: Transfer a
volume of HF that corresponds to 100 μL for every well (con-
trol and cTnT-stained) being treated. Add 1 μL of goat anti-
mouse-APC secondary antibody per 200 μL of HF (1:200
dilution).
16. Stain the samples: Add 100 μL of staining solution to each well
(negative and anti-cTnT-stained wells). Incubate cells for 30
min at 4 °C (keep the plate covered or in the dark after adding
fluorescent secondary antibody to avoid photobleaching).
17. Centrifuge the cells at 300×g for 2 min. Aspirate the superna-
tant and add 200 μL HF per well. Repeat the wash step one
more time.
Scalable Cardiac Differentiation of Human Pluripotent Stem Cells…
24
18. Transfer the samples to 5 mL round-bottom flow cytometer
analysis tubes and analyze on a flow cytometer using the
red laser.
4 Notes
1. If 500 mL of complete StemPro-34 medium (with supple-
ment) will not be used within 30 days, it is recommended that
the basal medium and supplement be aliquoted as follows:
prepare ten 50 mL aliquots of the basal medium and store at
4 °C, prepare ten 1.3 mL aliquots of the supplement and store
at −20 °C. When preparing complete medium from the ali-
quots, completely thaw 1 aliquot of supplement in a 37 °C
water bath and then add the supplement to a 50 mL aliquot of
basal medium.
2. To prepare feeder-depleted hESCs, passage the cells onto
Growth Factor Reduced Matrigel (BD 354230) coated 6 well
plates at a split ratio that will produce 1–2×106
cells per well
after 1–2 days. Use the same medium that was being used to
maintain the hESCs on feeders.
3. Depending on the volume of Induction Medium to be pre-
pared, the volume of some of the growth factor stock solutions
to be added on a given day may be less than 1 μL. To ensure
accurate volumes of growth factor stock solutions are being
added to the medium preparation, the stock may first be
diluted 1:10 in StemPro-34 basal medium, and then the appro-
priate volume of diluted stock may be added to the medium
preparation.
4. At this seeding density, aggregates containing 1,000 cells will
be formed. This is based on our observations with cardiac
induction from the HES-2 cell line maintained on feeders
[12]. The ideal seeding density may vary and should be
optimized for the specific cell lines and culture conditions
being used.
5. If the cells do not appear to form a tight pellet at the bottom
of the microwells, try eliminating air bubbles in the microwells
by spinning some medium into the wells prior to adding the
cells: Add 0.4 mL of medium per well and centrifuge the plate
at 200×g for 5 min. Then add the appropriate number of cells
to each well in 0.6 mL medium per well.
6. If the total number of samples being prepared is small, staining
can also be performed in 1.5 mL microcentrifuge tubes. If
microcentrifuge tubes are used, supernatant is removed by
aspiration and it is recommended that minimum wash volumes
of 0.5 mL and centrifuge settings of 900×g (in a bench-top
microcentrifuge) for 3 min be used for the wash steps.
Celine L. Bauwens and Mark D. Ungrin
25
Acknowledgements
We thank Dr. Peter Zandstra, in whose laboratory this protocol
was developed, and Drs. Mark Gagliardi and Gordon Keller who
provided assistance in establishing the initial methods on which
this process was based. Protocol development was supported by an
Ontario Graduate Scholarship in Science and Technology to C.B.
and a grant from the Heart and Stroke Foundation of Ontario to
Peter Zandstra.
References
1. Wintermantel E, Mayer J, Blum J et al (1996)
Tissue engineering scaffolds using superstruc-
tures. Biomaterials 17:83–91
2. Lazar A, Peshwa MV, Wu FJ et al (1995)
Formation of porcine hepatocyte spheroids for
use in a bioartificial liver. Cell Transplant 4:
259–268
3. Sachlos E, Auguste DT (2008) Embryoid
body morphology influences diffusive trans-
port of inductive biochemicals: a strategy for
stem cell differentiation. Biomaterials 29:
4471–4480
4. Johnstone B, Hering TM, Caplan AI et al
(1998) In vitro chondrogenesis of bone
marrow-derived mesenchymal progenitor
cells. Exp Cell Res 238:265–272
5. Steinberg MS (1970) Does differential adhe-
sion govern self-assembly processes in histo-
genesis? Equilibrium configurations and the
emergence of a hierarchy among populations
of embryonic cells. J Exp Zool 173:395–433
6. Bauwens CL, Peerani R, Niebruegge S et al
(2008) Control of human embryonic stem cell
colony and aggregate size heterogeneity influ-
ences differentiation trajectories. Stem Cells
26:2300–2310
7. Koike M, Kurosawa H, Amano Y (2005) A
round-bottom 96-well polystyrene plate
coated with 2-methacryloyloxyethyl phos-
phorylcholine as an effective tool for embryoid
body formation. Cytotechnology 47:3–10
8. Ng ES, Davis RP, Azzola L et al (2005) Forced
aggregation of defined numbers of human
embryonic stem cells into embryoid bodies
fosters robust, reproducible hematopoietic dif-
ferentiation. Blood 106:1601–1603
9. Ungrin MD, Joshi C, Nica A et al (2008)
Reproducible, ultra high-throughput forma-
tion of multicellular organization from single
cell suspension-derived human embryonic
stem cell aggregates. PLoS One 3:e1565
10. Kozhich O, Hamilton R, Mallon B (2013)
Standardized generation and differentiation of
neural precursor cells from human pluripotent
stem cells. Stem Cell Rev Rep 9:531–536
11. Ungrin MD, Clarke G, Yin T et al (2012)
Rational bioprocess design for human pluripo-
tent stem cell expansion and endoderm differ-
entiation based on cellular dynamics.
Biotechnol Bioeng 109:853–866
12. Bauwens CL, Song H, Thavandiran N et al
(2011) Geometric control of cardiomyogenic
induction in human pluripotent stem cells.
Tissue Eng Part A 17:1901–1909
13. Golos TG, Giakoumopoulos M, Garthwaite
MA (2010) Embryonic stem cells as models of
trophoblast differentiation: progress, opportu-
nities, and limitations. Reproduction 140:3–9
14. Markway B, Tan G-K, Brooke G et al (2010)
Enhanced chondrogenic differentiation of
human bone marrow-derived mesenchymal
stem cells in low oxygen environment micro-
pellet cultures. Cell Transplant 19(1):29–42
15. Fey SJ, Wrzesinski K (2012) Determination of
drug toxicity using 3D spheroids constructed
from an immortal human hepatocyte cell line.
Toxicol Sci 127(2):403–411
16. Wallace L, Reichelt J (2013) Using 3D culture
to investigate the role of mechanical signaling
in keratinocyte stem cells. In: Turksen K (ed)
Skin stem cells. Humana Press, Totowa, NJ,
pp 153–164
Scalable Cardiac Differentiation of Human Pluripotent Stem Cells…
27
Milica Radisic and Lauren D. Black III (eds.), Cardiac Tissue Engineering: Methods and Protocols, Methods in Molecular Biology,
vol. 1181, DOI 10.1007/978-1-4939-1047-2_3, © Springer Science+Business Media New York 2014
Chapter 3
Preparation and Characterization of Circulating Angiogenic
Cells for Tissue Engineering Applications
Aleksandra Ostojic, Suzanne Crowe, Brian McNeill,
Marc Ruel, and Erik J. Suuronen
Abstract
Circulating angiogenic cells (CACs) are a heterogeneous cell population of bone marrow (BM) origin.
These cells are most commonly derived from the peripheral blood, bone marrow, and cord blood, and are
one of the leading candidates for promoting vascularization in tissue engineering therapies. CACs can be
isolated by culturing peripheral blood mononuclear cells (PBMCs) on fibronectin or by flow cytometry to
obtain more specific subpopulations. Here we will describe how to generate a population of CACs, and
how to characterize the cells and confirm their phenotype. Also, we will provide select methods that can
be used to assess the angiogenic and endothelial cell-like properties of the CACs.
Key words Circulating angiogenic cells (CACs), Angiogenic cell therapy, Neovascularization,
Peripheral blood mononuclear cells, Tissue engineering
1 Introduction
Endothelial progenitor cells (EPCs) were initially isolated from
peripheral blood in 1997, and identified by their co-expression
of CD34 and vascular endothelial growth factor receptor-2
(VEGFR-2) [1]. EPCs have been further shown to originate from
bone marrow (BM) and other tissues, including skeletal muscle
and adipose, dermis, and vascular tissues [2–4]. A true definition
has not yet been established for these cells, since their phenotype
and function can vary depending on their source and/or method
of selection. Regardless of origin, it is generally accepted that EPCs
play a role in contributing to neovascularization in ischemic tis-
sues, directly through participation in the generation of new blood
vessels, or indirectly through the production of various pro-
angiogenic cytokines [2, 5, 6].
Based on this knowledge, EPCs are actively being tested as
cells for therapy (±biomaterials) in multiple diseases such as diabetes,
28
wound repair, and cardiovascular disease [7]. The potential for
revascularization therapy using EPCs and biomaterials has been
demonstrated in animal models [8–11] and the feasibility and
safety of cell therapy for cardiovascular regeneration has been dem-
onstrated in clinical studies, albeit with relatively modest structural
and functional benefits [12, 13]. Furthermore, EPCs have been
used successfully as a strategy to vascularize engineered tissues
[14–16]. Together, this evidence demonstrates the promise that
EPCs have for promoting vascularization in tissue engineering and
regenerative medicine applications.
In 2008, Hirschi et al. assigned the name circulating angio-
genic cells (CACs) to a specific type of EPC, due to the fact that
they are isolated from the peripheral blood and possess angiogenic
properties [17]. The most common method for obtaining CACs
from peripheral blood is to isolate the mononuclear cells (MNCs)
and culture them on fibronectin-coated plates. After 4–7 days, the
non-adherent cells are removed and the remaining adherent cells
are considered CACs [18–20]. Starting with ~100 ml of peripheral
blood, this isolation procedure can consistently yield 25–60×106
cells, which is plenty for performing multiple experiments using
the same donor.
Within the population of CACs, CD34+
cells have been identi-
fied as one of the most pro-angiogenic subpopulations [21, 22].
CD34+
cells, or other subpopulations of interest, can be isolated
from the CACs using fluorescence-activated cell sorting (FACS).
This method labels specific cell-surface antigens to identify and
sort specific subpopulations of CACs. Common markers used to
identify the endothelial precursor/progenitor population are
CD34, CD133, and VEGFR-2 [23–26].
The outlined set of experiments describes the procedure for
isolating CACs from human peripheral blood, and how to charac-
terize the phenotype of the population with flow cytometry.
Furthermore, methods are provided to evaluate the angiogenic
properties of the cells and to confirm that they are CACs (using
DiI-acLDL and FITC-lectin staining).
2 Materials
1. Wash buffer: 1 % FBS, 0.11 % EDTA in Phosphate Buffered
Saline (PBS).
2. Histopaque 1077 (Sigma).
3. 12×9 ml Ethylenediaminetetraacetic acid (EDTA) vacutainer
tubes.
4. 50 ml conical tubes.
5. Human donors to donate peripheral blood.
2.1 Isolation
of Peripheral Blood
Mononuclear Cells
Aleksandra Ostojic et al.
29
1. Fibronectin: 20 μg/ml fibronectin in PBS. Store at 4 °C.
2. EBM medium (Lonza): 500 ml EBM supplemented with
EGM-2 components (Lonza), containing fetal bovine serum
(FBS), recombinant long R insulin-like growth factor-1 in
aqueous solution (R3
-IGF-1), endothelial growth factor vascu-
lar human recombinant (VEGF), gentamicin sulfate ampho-
tericin-B, and epidermal growth factor human recombinant in
a buffered BSA saline solution (see Note 1). Store at 4 °C.
3. 4×10 cm culture dishes.
1. PBS. Store at 4 °C.
2. EBM medium+supplements (see Subheading 2.2).
1. PBS. Store at 4 °C.
2. EBM medium+supplements (see Subheading 2.2).
3. Antibodies of interest for flow cytometry.
1. Human umbilical vein endothelial cells (HUVECs).
2. HUVEC Medium: M200 medium supplemented with LSGS
kit. Store at 4 °C.
3. ECMatrix™ (EMD Millipore; ECM625): includes 10× diluent
and matrix solution.
4. 10 cm plates.
5. PBS. Store at 4 °C.
6. CellTracker™ Orange (Invitrogen).
7. 4′,6-diamidino-2-phenylindol (DAPI): 50 μl stock (1 mg/ml)
in 950 μl PBS.
8. 96-well plate.
9. EBM medium (see Subheading 2.2).
1. 1,1′-dioctadecyl-3,3,3′,3′-tetramethylindocarbocyanine (DiI)-
labeled acetylated low-density lipoprotein (acLDL, Molecular
Probes; L-3484): stock at 1 mg/ml.
2. FITC-labeled Ulex-lectin (Sigma; L-9006): stock at 1 mg/ml.
3. PBS. Store at 4 °C.
4. Cytofix buffer (BD Bioscience).
3 Methods
Carry out all procedures at room temperature unless otherwise
specified. All reagents that are stored at 4 °C should first be warmed
to room temperature before use, unless otherwise specified.
2.2 Culture
of Peripheral Blood
Mononuclear Cells
2.3 Lifting Cells
2.4 CAC
Characterization
by Fluorescence-
Activated Cell Sorting
(FACS)
2.5 Angiogenesis
Assay
2.6 DiI and Lectin
Staining
Circulating Angiogenic Cells for Tissue Engineering
30
For ~100 ml blood (12 vials of EDTA vacutainer tubes):
1. Pour 20 ml Histopaque into 4×50 ml tubes (see Note 2).
2. Collect 12 × 9 ml blood into EDTA vacutainer tubes. Mix
gently for a minimum of 5 min on rocker before processing
(see Note 3).
3. Using a sterile pipette, slowly and carefully layer blood from
three tubes on top of the 20 ml Histopaque (see Note 4).
Repeat for each of the 4×50 ml tubes.
4. Centrifuge the tubes for 30 min at 840×g at room tempera-
ture (do not use centrifuge brakes—see Note 5).
5. Remove the tubes which now contain the separated blood
layers (Fig. 2). Aspirate off top plasma layer to within 1 cm of
buffy coat (see Note 6).
3.1 Isolation
of Mononuclear Cells
from Peripheral Blood
(Fig. 1)
Blood collection
PBMC isolation with Histopaque and differential centrifugation
4 days
Fibronectin coated plates + EBM-2
Conduct assays
Fig. 1 PBMC isolation and CAC culture protocol
plasma
Red blood cells
Buffy coat
Histopaque
Fig. 2 Following density gradient centrifugation, the peripheral blood is separated
into four layers: (1) red blood cell layer; (2) clear Histopaque layer; (3) “Buffy
coat” layer consisting of the mononuclear cells; and (4) plasma layer
Aleksandra Ostojic et al.
31
6. Using a sterile transfer pipette, remove the top 1 cm of plasma
layer and all of buffy coat and place it in clean, sterile 50 ml
tubes (should have four separate tubes). Bring final volume in
each tube to 40 ml with wash buffer.
7. Centrifuge the tubes at 300×g for 10 min (do not use centri-
fuge brakes).
8. Remove and discard the supernatant. Resuspend the cell pellets
in a few ml of wash buffer and combine the cell pellet from
each of the four tubes into a single tube. Bring the final volume
up to 20 ml using wash buffer.
9. Centrifuge the tubes at 300×g for 10 min (do not use centri-
fuge brakes).
10. Resuspend the pellet in 1 ml EBM culture medium.
1. Coat 4×10 cm plate with 3 ml of fibronectin solution
(see Note 7).
2. Incubate at room temperature for 30–60 min.
3. Aspirate the excess liquid and air dry for 15 min.
4. Rinse the plates 2× with PBS: leave PBS on for 5 min, aspirate,
and repeat.
5. Add 10 ml EBM medium with supplements, warmed to room
temperature.
6. Add ¼ of the cells to each of the four plates and culture at 5 %
CO2 at 37 °C for 4 days without changing the medium.
7. After 4 days, the adherent population is considered to be
CACs (Fig. 3).
3.2 Plating
Fibronectin
for Selection of CACs
Fig. 3 Adherent population of CACs generated from the culture of PBMCs on
fibronectin-coated plates for 4 days
Circulating Angiogenic Cells for Tissue Engineering
32
1. Aspirate the medium.
2. Lift cells using gentle pipetting (see Note 8).
3. Centrifuge at 300×g for 5 min (do not use centrifuge brakes).
4. Remove the supernatant and resuspend the CACs in desired
volume of EBM medium with supplements, warmed to room
temperature.
1. Lift and resuspend the cells as previously described.
2. Count cells with an automated system or a hemocytometer.
3. Place 5 × 105
CACs in a 1.5 ml centrifuge tube in 200 μl
of PBS.
4. Stain cells for 30 min at 4 °C with pre-conjugated antibodies.
Common antibodies used for characterization are CD34, KDR
(VEGFR-2), CD133, CD31, and L-selectin, among others
(see Table 1).
5. Wash with PBS and centrifuge at 300×g for 5 min.
6. Aspirate medium and resuspend in 500 μl EBM medium with
supplements, warmed to room temperature.
7. Place another 5×105
CACs in a 1.5 ml centrifuge tube in
200 μl of PBS and stain with appropriate IgG isotype-matched
control antibodies (see Note 9). Repeat steps 5 and 6. This
can be done concurrently with steps 3–6.
8. Analyze and quantify cells with flow cytometry (see Note 10)
[25, 27].
3.3 Lifting Cells
3.4 CAC
Characterization
by Fluorescence-
Activated Cell Sorting
(FACS)
Table 1
Common markers used to characterize CAC populations. This Table lists some of the markers that
have been previously used to characterize the phenotypes of cells in CAC populations. The listed
functions are general and not exclusive
Marker Function References
CD14 Monocyte marker [28, 29]
CD31/PECAM-1 Endothelial adhesion molecule [28–30]
CD34 Progenitor cell marker [1, 25, 29, 31, 32]
CD45 Leukocyte marker [28, 29, 33]
CD133 Progenitor cell marker [25, 29, 31, 32]
CD144/VE-cadherin Endothelial adhesion molecule [28, 29, 31]
CD146 Endothelial adhesion molecule [29, 31, 33]
c-Kit Progenitor cell marker [25, 31]
DiI-acLDL uptake Endothelial/macrophage detection [28, 31]
VEGFR-2 (KDR, Flk-1) Endothelial cell receptor [1, 25, 29, 31]
L-selectin Leukocyte adhesion molecule [25, 30]
Aleksandra Ostojic et al.
33
1. HUVECs are provided from the manufacturer in a frozen
state, and stored this way until needed. Thaw the HUVECs
at 37 °C in a water bath.
2. Transfer cells to a 15 ml tube with 4 ml pre-warmed HUVEC
medium.
3. Centrifuge at 300×g for 5 min.
4. Remove supernatant, and resuspend cells in pre-warmed
HUVEC medium.
5. Plate cells in 2× 10 cm dishes, split 50:50, which is equivalent
to ~5×105
cells/dish.
6. Incubate at 37 °C and 5 % CO2 until 70–80 % confluent
(see Note 11).
7. Change HUVEC medium the following day, and every second
day thereafter.
1. This protocol uses the ECMatrix™ angiogenesis kit (EMD
Millipore).
2. Thaw the 10× diluent and the gel solution on ice (see Note 12).
3. Mix 100 μl diluent with 900 μl of the gel solution. Aliquot
80 μl of gel mixture into individual wells of a 96-well plate
(see Note 13).
4. Allow to set at 37 °C for approximately 1 h.
5. Gels are then ready to use.
1. Remove the supernatant from the HUVEC cultures.
2. Add 1 ml of 0.25 % trypsin and incubate for 5 min at RT with
constant agitation (see Note 14).
3. Add 5 ml of EBM medium to the plate to inactivate the tryp-
sin. Gently pipette the cells to loosen them from the dish.
4. Rinse the plate with PBS and transfer contents to a 15 ml tube.
5. Centrifuge at 300×g for 5 min and then remove the
supernatant.
6. Stain the cells by resuspending them in CellTracker™ Orange
(2 μl aliquot of CellTracker™ Orange+998 μl PBS) for 30 min
at 37 °C (see Note 15).
7. Add 5 ml of PBS, and then spin the cells as in step 5.
8. Resuspend in desired volume of EBM medium with supple-
ments, warmed to room temperature.
1. After completing step 3 of Subheading 3.3, remove the
supernatant.
2. Resuspend the CACs in 4′,6-diamidino-2-phenylindole
(DAPI) for 30 min at 37 °C. DAPI solution is prepared by
mixing 50 μl of stock (1 mg/ml) with 950 μl PBS.
3.5 Angiogenesis
Assay
3.5.1 Human Umbilical
Vein Endothelial Cell
(HUVEC) Culture
3.5.2 Making the In Vitro
Angiogenesis Gels
3.5.3 Lifting HUVECs
3.5.4 Performing the
Angiogenesis Assay
Circulating Angiogenic Cells for Tissue Engineering
34
3. Add 5 ml PBS, and centrifuge at 300×g for 5 min.
4. Remove the supernatant and resuspend CACs in the desired
volume of EBM medium with supplements, warmed to room
temperature.
5. Count cells using a hemocytometer.
6. Add 10,000 HUVECs and 10,000 CACs together to each well
containing ECMatrix™ gel (see Note 16).
7. Fill the wells with 150 μl EBM medium with supplements and
incubate at 37 °C overnight (should be 18 h, and no more
than a 24 h incubation) (see Note 17).
8. Take photographs of the capillary-like structure formations
using fluorescence microscopy. Evaluate the number of CACs
(blue DAPI+
cells) that have incorporated into the tubule
structures (orange HUVECs), as well as the total length of
tubule formation (see Note 18) (Fig. 4).
1. Prepare (DiI)-labeled acetylated low-density lipoprotein
(acLDL) stain (stock at 1 mg/ml). Take 2 μl and add to 998 μl
PBS.
2. Aspirate medium from CAC cultures and rinse the plates 2×
with PBS.
3. Add 1 ml of DiI-acLDL per dish and incubate at 37 °C for 1 h.
4. Aspirate the DiI-acLDL dye and rinse the cells 2× with PBS.
5. Fix the cells with 1.0 ml of Cytofix buffer for 15 min at 37 °C.
3.6 DiI and Lectin
Staining
Fig. 4 Angiogenesis assay. HUVECs are stained with CellTracker™ Orange (red/
orange) and CACs are stained with DAPI (blue). Arrows indicate CACs that have
incorporated into the capillary-like structures
Aleksandra Ostojic et al.
35
6. During this incubation step, prepare the lectin stain. Dilute
FITC-labeled Ulex-lectin to 10 μg/ml by adding 10 μl of
stock solution (1 mg/ml) to 990 μl of PBS.
7. Rinse cells 2× with PBS and add 1 ml FITC-labeled Ulex-lectin
per dish and incubate at 37 °C for 1 h.
8. Rinse cells 2× with PBS.
9. Add 1 ml of PBS to each dish and image the cells by fluores-
cence microscopy (Fig. 5). Dual-staining cells are considered
to be CACs (see Note 19).
4 Notes
1. Use a pipette when adding the supplements to minimize vol-
ume loss during the transfer.
2. 20 ml of Histopaque is the recommended amount per 30 ml of
blood. This amount can be reduced to 10 ml of Histopaque
per 30 ml of blood to save on reagents. However, the more
Histopaque there is, the easier it is to identify the buffy coat.
3. This step is necessary to mix the blood with the EDTA and
prevent clotting. If using the blood right away, mixing can be
achieved by inverting the collection tubes multiple times, oth-
erwise leave on the rocker.
4. Cannot have layers mix because it will affect the separation
efficacy, and the buffy coat layer will not be as visible.
5. It is very important that braking is not applied during centrifu-
gation, as this may disrupt proper separation of the layers.
Fig. 5 DiI-acLDL (red) and FITC-lectin (green) staining of a CAC culture
Circulating Angiogenic Cells for Tissue Engineering
36
6. Make sure not to aspirate any cells that may be stuck to the side
of falcon tube. Gently scrape them off with a pipette before
proceeding with the transfer.
7. Cover the plate and gently swirl to spread the fibronectin.
Make sure all areas are covered; otherwise cells may not uni-
formly adhere. Fibronectin coating step can be performed
immediately prior to CAC isolation. This way the plates will be
ready right about the same time that the cells are.
8. PBS can be added and cells can be washed multiple times.
If absolutely necessary, a scraper can be used to help lift the
cells. Do not allow areas of the plate to dry during this step.
Add PBS and look under the microscope to ensure that most
of the cells have been removed.
9. If flow cytometry analysis cannot be performed immediately,
then the cells can be fixed with 4 % PFA for 20 min, washed
with PBS, and spun for 5 min at 300×g. Cells can then be left
in the fridge for an extended period of time (weeks) before
staining (with primary or IgG control antibodies).
10. Use flow cytometry for analysis using a set number of events
(cells). A recommended number of events is 100,000–200,000,
but more or less can be used depending on the desired analysis.
11. If cells are over-confluent, they may suffer contact inhibition and
lose their growth and proliferative potential, and may also become
very difficult to trypsinize. Monitor cell density and make sure to
split cells before confluence is reached (>80–90 %).
12. The gel components typically take about 2 h to thaw on ice.
When ECMatrix™ is prepared, it is imperative to keep it on ice,
otherwise solidification will occur.
13. When pipetting the gel into the wells, do not expel all the way
in order to avoid bubbles. As an alternative to 96-well plates,
Ibidi microslides can be used, which will reduce reagent
consumption.
14. Carefully tap the side of the plate to help lift the cells.
15. Depending on the setup of various microscope systems, other
cytoplasmic dyes/fluorochromes may be more suitable to
visualize the cells, such as CellTracker™ Green (Invitrogen).
16. Must have 1:1 ratio. Adding too little cells will not allow the
HUVECs to form tubule structures and adding too many cells
may result in overcrowding or tubules forming too quickly and
then regressing.
17. Allowing the angiogenesis assay to proceed for longer than 24 h
will lead to regression of the tubule structures. As an alternative
to EBM medium, the assay can be performed using M200
medium (this is the medium used for HUVEC culture).
Aleksandra Ostojic et al.
37
18. It is recommended that a minimum of three, and ideally six
images of random fields-of-view (FOV) be taken per well.
There are three suggested ways to analyze the results using
imaging analysis software (such as ImageJ; http://rsbweb.nih.
gov/ij/). (A) Count the number of DAPI+
CACs (blue) that
have incorporated into tubule structures per FOV (HUVECs,
stained with CellTracker™ Orange). (B) Quantify the total
length of tubule formation per field-of-view. This is done by
measuring the length of each tubule branch and adding them
all together. (C) Quantify the total area of tubule formation.
This is done by measuring the area of all the tubule formations
and adding them together. The results from all 3–6 FOV are
then averaged to obtain the final value per well.
19. DiI-acLDL and FITC-lectin staining is a traditional method
originally used to identify adherent endothelial progenitor cells
(EPCs) cells from the peripheral blood. However, this means
of identifying EPCs is not considered sufficient, and additional
characterization is necessary [7]. We suggest this staining sim-
ply as a means of confirming the successful generation of an
adherent CAC population.
Acknowledgements
This work was supported by the Canadian Institutes of Health
Research (to MR and EJS). BM was supported by a University of
Ottawa Cardiology Research Endowment Fellowship.
References
1. Asahara T, Murohara T, Sullivan A et al (1997)
Isolation of putative progenitor endothelial
cells for angiogenesis. Science 275:964–967
2. Asahara T, Takahashi T, Masuda H et al (1999)
VEGF contributes to postnatal neovasculariza-
tion by mobilizing bone marrow-derived endo-
thelial progenitor cells. EMBO J 18:3964–3972
3. Grenier G, Scime A, Le Grand F et al (2007)
Resident endothelial precursors in muscle,
adipose, and dermis contribute to postnatal
vasculogenesis. Stem Cells 25:3101–3110
4. Tamaki T, Akatsuka A, Ando K et al (2002)
Identification of myogenic-endothelial progen-
itor cells in the interstitial spaces of skeletal
muscle. J Cell Biol 157:571–577
5. Asahara T, Kawamoto A, Masuda H (2011)
Concise review: circulating endothelial pro-
genitor cells for vascular medicine. Stem Cells
29:1650–1655
6. Moreno PR, Sanz J, Fuster V (2009)
Promoting mechanisms of vascular health:
circulating progenitor cells, angiogenesis, and
reverse cholesterol transport. J Am Coll Cardiol
53:2315–2323
7. Fadini GP, Losordo D, Dimmeler S (2012)
Critical reevaluation of endothelial progenitor
cell phenotypes for therapeutic and diagnostic
use. Circ Res 110:624–637
8. Gilbert PM, Blau HM (2011) Engineering a
stem cell house into a home. Stem Cell Res
Ther 2:3
9. Kuraitis D, Giordano C, Ruel M et al (2012)
Exploiting extracellular matrix-stem cell interac-
tions: a review of natural materials for therapeutic
muscle regeneration. Biomaterials 33:428–443
10. Segers VF, Lee RT (2011) Biomaterials to
enhance stem cell function in the heart. Circ
Res 109:910–922
Circulating Angiogenic Cells for Tissue Engineering
38
11. Nunes SS, Song H, Chiang CK et al (2011)
Stem cell-based cardiac tissue engineering.
J Cardiovasc Transl Res 4:592–602
12. Delewi R, Andriessen A, Tijssen JG et al (2013)
Impact of intracoronary cell therapy on left ven-
tricular function in the setting of acute myocar-
dial infarction: a meta-analysis of randomised
controlled clinical trials. Heart 99:225–232
13. Jeevanantham V, Butler M, Saad A et al (2012)
Adult bone marrow cell therapy improves sur-
vival and induces long-term improvement in
cardiac parameters: a systematic review and
meta-analysis. Circulation 126:551–568
14. Amini AR, Laurencin CT, Nukavarapu SP
(2012) Differential analysis of peripheral
blood- and bone marrow-derived endothelial
progenitor cells for enhanced vascularization in
bone tissue engineering. J Orthop Res 30:
1507–1515
15. Critser PJ, Voytik-Harbin SL, Yoder MC
(2011) Isolating and defining cells to engineer
human blood vessels. Cell Prolif 44(Suppl
1):15–21
16. Zhang Z, Ito WD, Hopfner U et al (2011) The
role of single cell derived vascular resident
endothelial progenitor cells in the enhance-
ment of vascularization in scaffold-based skin
regeneration. Biomaterials 32:4109–4117
17. Hirschi KK, Ingram DA, Yoder MC (2008)
Assessing identity, phenotype, and fate of
endothelial progenitor cells. Arterioscler
Thromb Vasc Biol 28:1584–1595
18. Favre J, Terborg N, Horrevoets AJ (2013) The
diverse identity of angiogenic monocytes. Eur J
Clin Invest 43:100–107
19. Kalka C, Masuda H, Takahashi T et al (2000)
Transplantation of ex vivo expanded endothe-
lial progenitor cells for therapeutic neovascu-
larization. Proc Natl Acad Sci U S A 97:
3422–3427
20. Kuraitis D, Hou C, Zhang Y et al (2011) Ex
vivo generation of a highly potent population
of circulating angiogenic cells using a collagen
matrix. J Mol Cell Cardiol 51:187–197
21. Mackie AR, Losordo DW (2011) CD34-
positive stem cells: in the treatment of heart
and vascular disease in human beings. Tex
Heart Inst J 38:474–485
22. Yang J, Ii M, Kamei N et al (2011) CD34+
cells represent highly functional endothelial
progenitor cells in murine bone marrow. PLoS
One 6:e20219
23. Garmy-Susini B, Varner JA (2005) Circulating
endothelial progenitor cells. Br J Cancer 93:
855–858
24. Peichev M, Naiyer AJ, Pereira D et al (2000)
Expression of VEGFR-2 and AC133 by circu-
lating human CD34(+) cells identifies a popu-
lation of functional endothelial precursors.
Blood 95:952–958
25. Sofrenovic T, McEwan K, Crowe S et al (2012)
Circulating angiogenic cells can be derived
from cryopreserved peripheral blood mono-
nuclear cells. PLoS One 7:e48067
26. Werner N, Kosiol S, Schiegl T et al (2005)
Circulating endothelial progenitor cells and
cardiovascular outcomes. N Engl J Med 353:
999–1007
27. Khan SS, Solomon MA, McCoy JP Jr (2005)
Detection of circulating endothelial cells and
endothelial progenitor cells by flow cytometry.
Cytometry B Clin Cytom 64:1–8
28. Shepherd RM, Capoccia BJ, Devine SM et al
(2006) Angiogenic cells can be rapidly mobi-
lized and efficiently harvested from the blood
following treatment with AMD3100. Blood
108:3662–3667
29. Steinmetz M, Nickenig G, Werner N (2010)
Endothelial-regenerating cells: an expanding
universe. Hypertension 55:593–599
30. Yip HK, Chang LT, Chang WN et al (2008)
Level and value of circulating endothelial pro-
genitor cells in patients after acute ischemic
stroke. Stroke 39:69–74
31. Rafii S, Lyden D (2003) Therapeutic stem and
progenitor cell transplantation for organ vascular-
ization and regeneration. Nat Med 9:702–712
32. Werner N, Wassmann S, Ahlers P et al (2007)
Endothelial progenitor cells correlate with
endothelial function in patients with coronary
artery disease. Basic Res Cardiol 102:565–571
33. Delorme B, Basire A, Gentile C et al (2005)
Presence of endothelial progenitor cells, dis-
tinct from mature endothelial cells, within
human CD146+ blood cells. Thromb Haemost
94:1270–1279
Aleksandra Ostojic et al.
39
Milica Radisic and Lauren D. Black III (eds.), Cardiac Tissue Engineering: Methods and Protocols, Methods in Molecular Biology,
vol. 1181, DOI 10.1007/978-1-4939-1047-2_4, © Springer Science+Business Media New York 2014
Chapter 4
Isolation and Expansion of C-Kit-Positive Cardiac
Progenitor Cells by Magnetic Cell Sorting
Kristin M. French and Michael E. Davis
Abstract
Cell therapy techniques are a promising option for tissue regeneration; especially in cases such as heart
failure where transplantation is limited by donor availability. Multiple cell types have been examined for
myocardial regeneration, including mesenchymal stem cells (and other bone marrow-derived cells),
induced pluripotent stem cells, embryonic stem cells, cardiosphere-derived cells, and cardiac progeni-
tor cells (CPCs). CPCs are multipotent and clonogenic, can be harvested from mature tissue, and have
the distinct advantages of autologous transplant and lack of tumor formation in a clinical setting. Here
we focus on the isolation, expansion, and myocardial differentiation of rat CPCs. Brief adaptations of
the protocol for isolation from mouse and human tissue are also provided.
Key words Cardiac progenitor cells, C-kit, Cell therapy, MACS, Myocardial infarction
1 Introduction
Cardiac progenitor cells (CPCs), expressing the stem cell marker
c-kit, were first identified in the adult heart a decade ago and shown
to be self-renewing, clonogenic, and multipotent [1]. In addition
to c-kit, CPCs express the stem cell markers multiple drug resis-
tance (MDR) and Sca-1 as well as cardiac markers Nkx2.5 and
Gata-4. They are negative for markers of the hematopoietic lin-
eage. CPCs are reported to exist at a density of 1 per 10,000 myo-
cytes in the adult rat heart [1] and as high as 1.4 % of total cardiac
cells in humans [2]. The safety and efficacy of CPCs for myocardial
repair were demonstrated in the SCIPIO Phase I clinical trial [3].
This trial, in addition to various animal studies, suggests that CPCs
can improve cardiac function post-injury through reduction in scar
size and myocardial preservation/regeneration [4–6]. CPCs may
also be a stem cell candidate for generating cardiac patches for tis-
sue repair or drug screening [7].
In order to isolate CPCs, a tissue section must be excised from the
myocardium (atrial tissue is preferred). The cells are then enzymatically
Random documents with unrelated
content Scribd suggests to you:
Tapioca must be soaked in twice the quantity of water over night,
then add milk and water, and boil till it is soft. Flavor as above.
Dropped Egg.
Salt some boiling water, and drop in it a raw egg out of the shell,
taking care not to break the yolk; take it up as soon as the white is
hardened. Dip some toast in hot water, and put salt or butter on to
it, and lay the egg on the top.
Wheat Gruel for Young Children with weak stomachs, or for Invalids.
Tie half a pint of wheat flour in thick cotton, and boil it three or four
hours; then dry the lump and grate it when you use it. Prepare a
gruel of it by making a thin paste, and pouring it into boiling milk
and water, and flavor with salt. This is good for teething children.
Another Panada.
Boil a mixture of one-fourth wine, and three-fourths water, and
flavor it with nutmeg or lemon. Stir in grated bread or crackers, and
let it boil up once.
Herb Drinks.
Balm tea is often much relished by the sick. Sage tea also is good.
Balm, sage, and sorrel, mixed with sliced lemon and boiling water
poured on, and then sweetened, is a fine drink. Pennyroyal makes a
good drink to promote perspiration.
Herb drinks must often be renewed, as they grow insipid by
standing.
Other Simple Drinks.
Pour boiling water on to tamarinds, or mashed cranberries, or
mashed whortleberries, then pour off the water and sweeten it. Add
a little wine if allowed.
Toast bread very brown, and put it in cold water, and it is often
relished. Pour boiling water on to bread toasted very brown, and boil
it a minute, then strain it, and add a little cream and sugar. Make a
tea of parched corn pounded, and add sugar and cream.
Cream Tartar Whey.
Warm a pint of fresh milk, when scalding hot, stir in a teaspoonful of
cream tartar, and if this does not turn it, add more, till it does. Strain
it, and sweeten with loaf sugar. Those who cannot eat wine whey
can eat this without trouble, and it is good in fevers.
Simple Wine Whey.
Mix equal quantities of water, milk, and white wine. Warm the milk
and water, and then add the wine. Sweeten it to the taste.
A great Favorite with Invalids.
Take one third brisk cider and two thirds water, sweeten it, and
crumb in toasted bread, or toasted crackers, and grate on nutmeg.
Acid jellies will answer for this, when cider cannot be obtained.
A New Way of making Barley Water.
Put two tablespoonfuls of pearl barley into a quart jug, two great
spoonfuls of white sugar, a small pinch of salt, a small bit of orange,
or lemon peel, and a glass of calve’s foot jelly, and then fill the jug
with boiling water. Shake it, and then let it stand till quite cold. It is
best made over night, to use next day. When the liquor is all poured
off, it may be filled again with boiling water, and it is again very
good.
Panada.
Take two crackers, pour on boiling water, and let it simmer five
minutes; beat up an egg, sweeten and flavor it to your taste, and
then put the cracker to it.
Arrowroot Blanc Mange.
Take two tablespoonfuls of arrowroot to one quart of milk, and a
pinch of salt. Scald the milk, sweeten it, and then stir in the
arrowroot, which must first be wet up with some of the milk. Let it
boil up once. Orange water, rose water, or lemon peel, can be used
to flavor it. Pour it into moulds to cool.
Rice Flour Blanc Mange.
Four tablespoonfuls of ground rice and a pinch of salt wet up with a
little milk and stirred into a quart of boiling milk. Rub the rind of a
lemon with hard, refined sugar, till all the oil is absorbed, and use
the sugar to sweeten to your taste. Boil, stirring well, for eight
minutes; then cool it, and add the whites of three eggs cut to a
froth. Put it on to the fire, and stir constantly till boiling hot, then
turn it into moulds, or cups, and let it stand till cold.
Another Receipt for American Isinglass Jelly.
One ounce of gelatine, or American isinglass.
Three pints of boiling water.
A pound and a half of loaf sugar.
Three lemons, cut in slices, leaving out the peel of one.
The whites of four eggs, cut to a stiff froth.
Soak the isinglass half an hour in cold water, then take it out and
pour on the boiling water. When cool, add the sugar, lemon, and
whites of eggs; boil all three or four minutes, then strain through a
jelly-bag, and add wine to your taste.
Tapioca Jelly.
One cup full of tapioca.
Wash it two or three times, soak it in water, for five or six hours.
Then simmer it in the same water in which it has been soaked, with
a pinch of salt and bits of fresh lemon peel, until it becomes
transparent. Then add lemon juice, wine, and loaf sugar to flavor it.
Let all simmer well together, then pour into glasses to cool.
Caudle.
To rice, or water gruel, add a wine-glass of wine, or ale, and season
with nutmeg and sugar.
Sago Jelly.
Soak a tea-cup full of sago in cold water, half an hour, then pour off
the water, and add fresh, and soak it another half hour; and then
boil it slowly with a pinch of salt, a stick of cinnamon, or a bit of
orange, or lemon peel, stirring constantly. When thickened, add wine
and white sugar to suit the taste, and let it boil a minute; then turn
it into cups.
Spiced Chocolate.
One quart of milk.
Two squares of chocolate.
One stick of cinnamon.
A little nutmeg.
Grate the chocolate. Boil the milk, reserving a little cold to moisten
the chocolate, which must be mixed perfectly smooth to a thin
paste. When the milk boils (in which the cinnamon must be put
when cold, and boil in it), stir in the chocolate, and let it boil up
quickly, then pour into a pitcher, and grate on the nutmeg. Rich
cream added to the milk, will improve it.
Barley Water.
Put two ounces of pearl barley to half a pint of boiling water, and let
it simmer five minutes; pour off the water, and add two quarts of
boiling water, add two ounces sliced figs, two of stoned raisins, and
boil till it is reduced to a quart. Strain it for drink.
Water Gruel.
To two quarts of boiling water, add one gill of Indian meal and a
heaped tablespoonful of flour, made into a paste and stirred in the
water. Let it boil slowly twenty minutes. Salt, sugar, and nutmeg to
the taste.
Oatmeal makes a fine gruel in the same way.
Beef Tea.
Broil a pound of tender, juicy beef ten minutes, salt and pepper it,
cut it in small pieces, pour on a pint of boiling water, steep it half an
hour, and then pour it off to drink. Another way is slower, but better.
Cut the beef in small pieces, fill a junk bottle with them, and keep it
five hours in boiling water. Then pour out, and season the juice thus
obtained.
Tomato Syrup.
Express the juice of ripe tomatoes, and put a pound of sugar to each
quart of the juice, put it in bottles, and set it aside. In a few weeks it
will have the appearance and flavor of pure wine of the best kind,
and mixed with water is a delightful beverage for the sick. No
alcohol is needed to preserve it.
The medical properties of the tomato are in high repute, and it is
supposed that this syrup retains all that is contained in the fruit.
Arrowroot Custard for Invalids.
One tablespoonful of arrowroot.
One pint of milk. One egg.
One tablespoonful of sugar.
Mix the arrowroot with a little of the cold milk, put the milk into a
sauce-pan over the fire, and when it boils, stir in the arrowroot and
the egg and sugar, well beaten together. Let it scald, and pour into
cups to cool. A little cinnamon boiled in the milk flavors it pleasantly.
Sago for Invalids.
Wash one large spoonful of sago, boil it in a little water, with a pinch
of salt and one or two sticks of cinnamon, until it looks clear; then
add a pint of milk, boil all well together, and sweeten with loaf sugar.
Rice Jelly.
Make a thin paste of two ounces of rice flour, and three ounces of
loaf sugar, and boil them in a quart of water till transparent. Flavor
with rose, orange, or cinnamon water. It can be made also by boiling
whole rice long and slowly. A pinch of salt improves it.
Sassafras Jelly.
Take the pith of sassafras boughs, break it in small pieces, and let it
soak in cold water till the water becomes glutinous. It has the flavor
of sassafras, and is much relished by the sick, and is also good
nourishment.
Buttermilk Whey.
One quart of good buttermilk. When boiling, beat up the yolk of an
egg, and stir in, and, if it can be allowed, some thick cream, or a
little butter. Then beat the white to a stiff froth and stir in. Sugar and
spice if liked.
Alum Whey.
Mix half an ounce of pounded alum with one pint of milk. Strain it,
and add sugar and nutmeg to the whey. It is good in cases of
hemorrhages, and sometimes for colic.
Another Wine Whey.
One pint of boiling milk.
Two wine-glasses of wine.
Boil them one moment, stirring.
Take out the curd, and sweeten and flavor the whey.
Mulled Wine.
One pint of wine and one pint of water.
Beat eight eggs and add to the above, while boiling, stirring rapidly.
As soon as it begins to boil it is done.
Tamarind Whey.
Mix an ounce of tamarind pulp with a pint of milk, strain it, and add
a little white sugar to the whey.
Egg Tea and Egg Coffee (very fine).
Beat the yolk of an egg with a great spoonful of sugar, and put it to
a tea-cup of cold tea or cold coffee. Add a half a tea-cup of water,
cold in summer and boiling in winter, and as much cream. Then whip
the white of the egg to a stiff froth and stir it in. It is very much
relished by invalids.
Cranberry Tea.
Wash ripe cranberries, mash them, pour boiling water on them, and
then strain off the water and sweeten it, and grate on nutmeg.
Apple Tea.
Take good pippins, slice them thin, pour on boiling water, and let it
stand some time. Pour off the water, and sweeten and flavor it.
Egg and Milk.
Beat the yolk of an egg into a great spoonful of white sugar, or
more. Add a coffee cup of good milk, then beat the white of the egg
to a stiff froth, and stir it in. A little wine, or nutmeg to flavor it.
Sago Milk.
Soak one ounce of sago in a pint of cold water an hour. Pour off the
water, and add a pint and a half of new milk. Simmer it slowly till the
sago and milk are well mixed. Flavor with sugar, nutmeg, and wine.
Tapioca Milk.
Made like sago milk, only not boiled so long.
Bread and Milk.
Take a slice of good bread and soak it in milk, and then put on a
little butter, and it is often very acceptable to the sick. In some cases
sprinkle a little salt on instead of butter.
Egg Gruel.
Beat the yolk of an egg with a spoonful of white sugar, and then
beat the white separately, to a stiff froth. Pour water when boiling to
the yolk, then stir in the white and add spice, or any seasoning, to
suit the taste. When a person has taken a violent cold, after being
warm in bed give this as hot as it can be taken, and it is often a
perfect cure.
Ground Rice Gruel.
Take two tablespoonfuls of ground rice, and a pinch of salt, and mix
it with milk enough for a thin batter. Stir it with a pint of boiling
water, or boiling milk, and flavor with sugar and spice.
Oatmeal Gruel.
Four tablespoonfuls of grits (coarse oatmeal) and a pinch of salt,
into a pint of boiling water. Strain and flavor it while warm.
Or, take fine oatmeal and make a thin batter with a little cold water,
and pour it into a sauce-pan of boiling water.
Simple Barley Water.
Take two ounces and a half of pearl barley, cleanse it, and boil it ten
minutes in half a pint of water. Strain out this water and add two
quarts of boiling water, and boil it down to one quart. Then strain it,
and flavor it with slices of lemon and sugar, or sugar and nutmeg.
This is very acceptable to the sick in fevers.
Compound Barley Water.
Take two pints of simple barley water, a pint of hot water, two and a
half ounces of sliced figs, half an ounce of liquorice root sliced and
bruised, and two ounces and a half of raisins. Boil all down to two
pints, and strain it. This is slightly aperient.
Cream Tartar Beverage.
Take two even teaspoonfuls of cream tartar, and pour on a pint of
boiling water, and flavor it with white sugar and lemon peel to suit
the taste. If this is too acid, add more boiling water, as cold, or
lukewarm water, is not so good.
Seidlitz Powders.
Two drachms of Rochelle salts, and two scruples of bicarbonate of
soda, in a white paper; thirty-five grains of tartaric acid in a blue
one.
Dissolve that in the white paper in nearly half a tumbler of water,
then add the other powder, dissolved in another half tumbler of
water.
Syrup mixed with the water makes it more agreeable. It is a gentle
laxative.
Blackberry Syrup, for Cholera and Summer Complaint.
Two quarts of blackberry juice.
One pound of loaf sugar.
Half an ounce of nutmegs.
A quarter of an ounce of cloves.
Half an ounce of cinnamon.
Half an ounce of allspice.
Pulverize the spice, and boil all for fifteen or twenty minutes. When
cold, add a pint of brandy.
Remarks on the Combinations of Cooking.
The preceding receipts have been tested by the best housekeepers.
In reviewing them, it will be seen that there are several ways of
combining the various articles, all of which have, in the hands of
good housekeepers, proved successful. Still it will be found that
some methods are more successful than others.
In most cases, the receipts have been written as given by the ladies,
who endorse them as the best. But it is believed that the following
general rules will enable a housekeeper to modify some of them to
advantage.
In using the whites of eggs, it is found, as shown by several
receipts, that various combinations are much lighter when they are
cut to a froth, and put in the last thing. This is so in batter puddings,
and several other receipts. It seems, therefore, probable that in all
cases, cake and pies, and puddings that will allow it, will be lighter
by adding the cut whites of the eggs the last minute before cooking.
Sponge cake especially would most probably be most easily made
light by this method.
In using alkalies with acids to raise mixtures, the poorest is pearlash,
the next best is saleratus; bicarbonate of soda is still better, and sal
volatile is best of all.
But one thing must be remembered in reference to sal volatile, and
that is, that the lightness made by it is owing to the disengagement
of the gas by heat. It is mixed with the flour, and when set in the
oven, the heat volatilizes and expels the gas, and thus the lightness
is induced. Of course hot water must not be used to dissolve it, as it
would expel much of the gas. Sal volatile must be kept powdered,
and closely confined in glass bottles with ground glass stoppers. It is
certain to make any mixture light that can be raised by anything.
Cream tartar is best bought in lumps, and then pulverized and kept
corked.
When saleratus is used with sour milk or buttermilk, the flour should
be wet up with the sour milk, and then the alkali dissolved, and
worked in. This makes the effervescence take place in the mixture;
whereas, if the alkali is put into the sour liquid, much of the carbonic
acid generated is lost before it reaches the flour.
In all cases, then, where saleratus is used with acid wetting, it would
seem best to wet up the flour with at least a part of the sour liquid,
before putting in the alkali.
When the alkali is a light powder, it may sometimes be mixed
thoroughly with the flour, and then the sour liquid be mixed in. The
experiment can be made by any who like to learn the result. A lady
who understands chemistry may often improve her receipts by
applying chemical principles. All the lightness made by an acid and
an alkali is owing to the disengagement of carbonic acid, which is
retained by the gluten of the flour. Of course, then, that mode is
best which secures most effectually all the carbonic acid generated
by the combination.
Cooking is often much improved by a judicious use of sugar or
molasses. Thus, in soups, a very little sugar, say half a teaspoonful
to the quart, gives body to the soup, and just about as much
sweetness as is found in the juices of the best and sweetest kinds of
meat. It is very good when the meats used are of inferior kind, and
destitute of sweetness. So in preparing vegetables that are destitute
of sweetness, a little sugar is a great improvement. Mashed turnips,
squash, and pumpkin, are all of them much improved by extracting
all the water, and adding a little sugar, especially so when they are
poor.
A little molasses always improves all bread or cakes made of
unbolted wheat or rye.
A little lard or butter always improves cakes made of Indian meal, as
it makes them light and tender.
The careful use of salt is very important in cooking. Everything is
better to have the salt cooked in it, but there should always be a
little less salt than most would like, as it is easy for those who wish
more to add it, but none can subtract it.
When the shortening is butter, no salt is needed in cakes and
puddings, but in all combinations that have no salt in shortening, it
must be added. A little salt in sponge cake, custards, and the articles
used for desserts, made of gelatine, rice, sago, and tapioca, is a
great improvement, giving both body and flavor.
CHAPTER XXI.
ON MAKING BUTTER AND CHEESE.
The directions in this article were given by a practical amateur
cheese-maker of Goshen, Conn., a place distinguished all over the
nation for the finest butter and cheese.
Articles used in Making Cheese.
The articles used in making cheese are, a large tub, painted inside
and outside, to hold the milk, a large brass kettle to heat it, a cheese
basket, cheese hooks, cheese ladders, strainers of loose linen cloth,
and a cheese press.
It is indispensable that all the articles used be first washed
thoroughly, then scalded, and then dried thoroughly, before putting
away.
Mode of Preparing the Rennet.
Do not remove any part of the curd that may be found in the rennet
(which is the stomach of a calf), as it is the best part. Take out
everything mixed with the curd in the stomach. Soak the rennet in a
quart of water, then hang it to dry, where flies will not reach it, and
keep the water bottled for use. Rennet differs in strength, so that no
precise rule can be given for quantity, but say about half a tea-cup
full to two pails of milk.
To Make Cheese.
Strain the milk into the tub, keeping in all the cream. Heat a portion,
and then add it to the cold, till the whole is raised to 98° or 100°
Fahrenheit; no more and no less. Then put in the rennet, stirring
well, and take enough to have the curd form well in an hour. If it
does not form well, more must be stirred in.
When the curd is formed, cut it in small checks to the bottom, and
then break it gently with a skimmer, to make the whey separate. If
this is not done gently, the milk runs off, the whey turns white, and
the cheese is injured. The greener the whey, the better the cheese.
When the whey is separated, set the basket over the tub, spread the
strainer over, and dip the curd into it, occasionally lifting the corners
to hasten the draining.
Then draw up the corners, twist them, and set a stone on, to press
out the whey for ten minutes. Then again cut up the curd, and press
it again as before. Continue thus till it is thoroughly drained. Then
press it all into some regular form, and in cool weather it may stand
till next morning for more cheese to be added. But if the weather is
hot, it must be scalded the first day.
To Scald the Curd.
Cut, or chop the curd into cakes the fourth of an inch in size, put it
in the strainer, and immerse it in the brass kettle of warm water,
enough to cover it. Then raise the temperature to 105°. Stir it well
till warmed through, say half an hour. Then gradually add cold water,
till reduced to 88° or 90°. Then drain the curd thoroughly as before,
and salt it, allowing four ounces of salt to every ten pounds of curd,
and mixing very thoroughly. Then put it into the small strainers, and
then into the cheese hoops, laying the strainer over smoothly, and
placing the follower on it. Put it to press, and let it remain two days.
When taken from the press, grease it with common butter or butter
made of whey cream, and set it on a shelf in a dark, cool room.
Grease and turn it every day till firm, and for six months grease and
turn it often.
If the cheese is to be colored, boil anatto with ley, and put to the
milk with the rennet. To make sage cheese, put in sage juice (some
add to the sage spinach juice) when you put in the rennet. Sew
strips of strong cotton around large cheeses, when taken from the
press, to preserve their form. In fly time, put cayenne pepper in the
butter you rub on.
Stilton cheese is made of milk enriched by cream, and of a small
size.
Cottage cheese is made by pressing the curds of milk, and when free
from whey, adding cream or butter and salt.
Welsh rabbit is made by melting cheese and adding wine and other
seasoning.
Old and dry cheese is very good grated and mixed with a little
brandy, just enough to wet it up.
Directions for making Butter.
Two particulars are indispensable to success in making good butter;
the first is, that the churning be frequent, so that the cream will not
grow bitter, or sour; and the second is cleanliness in all the
implements and processes connected with it.
In hot weather it is important to keep the milk, cream, and butter as
cool as possible. For this purpose, those who have no ice-house, or
very cool milk-room, hang their cream down a well. In winter it is
needful to raise the temperature of the cream a little, while
churning, but care must be taken to do it very slightly, or the butter
is injured. The best way is to warm the churn, and churn in a warm
room.
After the weather gets cold, the cream rises more perfectly after
allowing the milk to stand say ten or eleven hours, to set it over a
furnace a while till it is warmed through, but not heated hot enough
to boil. Then take it back, let it stand eleven hours longer, and skim
it. This secures more, and better cream.
In hot weather, set the churn into a tub of cold water, and churn
steadily, as stopping puts back the process, and injures the butter. In
hot weather, do not churn very fast, as it makes soft butter. When
the butter has come, collect it in a wooden bowl, which is the best
article to work it in, having first scalded it, and then put it in cold
water till cold. Do not use the hand in working over butter, as it
injures it so much that a higher price is often paid for butter made
without using the hand.
A wooden spad made for the purpose is the proper article for
working over butter.
As soon as the butter is put in the bowl, pour in as much of the
coldest water as will allow you to work the butter, and keep adding
and pouring off cold water, as you work it, until the water will run off
clear. It is continuing this process until all the buttermilk is extracted,
which alone will secure butter that will keep good and sweet. Water
hastens the process, but butter keeps longer not to have it used.
No precise direction can be given for salting, as tastes vary so much
in regard to this. It is a good way to notice the proportions which
are most agreeable, and note the measure, and then measure ever
after.
In salting down in firkins, use the nicest rock salt, as much depends
on the nature of the salt. The firkins must be very thoroughly
seasoned, and the bottom covered with salt, and the sides rubbed
with it. Pack the butter in layers, with salt between. After a few
days, the butter will shrink from the sides, and then the space must
be filled with new and nice brine. Muslin spread under and over the
layer of salt, between the layers of butter is a good plan, as it saves
the butter.
It is said that butter will be preserved sweet a long time for
journeys, or voyages, by working into it very thoroughly a mixture
composed of one-fourth salt, one-fourth saltpetre, and two-fourths
white sugar.
In large dairies, the milk is churned soon after it is taken from the
cow.
The quality of butter depends very much upon the kind of cows.
Those who give a great deal of milk, are usually small and thin.
Every cow should have a tea-cup full of salt each week, and must be
well fed. Green cornstalks and carrots, are excellent for cows.
Turnips, cabbage, and parsnips spoil the milk. The waste of the
kitchen, with a quart of corn meal, and as much hay as she will eat
daily, is good fare. Skimmed milk for drink is good, and if it is
refused, withhold water, and the cow will learn to love it. Milk three
times a day, and you get much more cream; stripping must be done
thoroughly, or you lose cream, and dry up the cow. Never make a
cow run, as it injures the milk. Use tin vessels for milk.
A stoneware churn is best, and a tin one is better than wood. Keep
milk in a cool place, where air circulates freely; close air spoils milk
and cream. Never stir milk after it is set, as it stops the rising of the
cream. Skim milk as soon as it becomes loppard. Put a little ice in
each pan in hot weather, and you get more cream. In skimming
milk, do not scrape off the hardest portion that adheres to the pan,
as it injures the butter. Put a spoonful of salt to each pailful of milk
(except what is for family use), and it makes the butter sweeter, and
come easier. Salt your cream as you gather it, and it keeps better,
and makes sweeter butter. In hot weather churn in the coolest part
of the day, and in a cool place, and do not shut the air out of the
churn, as it is necessary to make the butter come. Butter is best, to
work it enough the first time. Never work it three times. It will keep
better to work out the buttermilk without putting in water. The more
entirely it is freed from buttermilk, the longer it will keep sweet.
A good brine is made for butter by dissolving a quart of fine salt, a
pound of loaf sugar, and a teaspoonful of saltpetre in two quarts of
water, and then strain it on to the butter. Packed butter is most
perfectly preserved sweet by setting the firkin into a larger firkin,
and filling in with good brine, and covering it. Butter will keep sweet
a year thus.
Buttermilk kept in potter’s ware dissolves the glazing, and becomes
poisonous.
Never scald strainers or milky vessels till thoroughly washed, as the
milk or cream put in them will be injured by it. The best way to scald
such vessels is to plunge them all over into scalding water, and then
every spot is scalded.
Butter will sometimes not come because the air is too much
excluded from the churn.
CHAPTER XXII.
ARTICLES AND CONVENIENCES FOR THE SICK.
“In some maladies,” says Dr. Pereira, “as fevers and acute
inflammatory diseases, an almost unlimited use of fluids is admitted,
under the names of slops, thin diet, fever diet, broth diet, &c. They
quench thirst, lessen the stimulating quality of the blood, increase its
fluidity, and promote the actions of the secreting organs. They are
sometimes useful, also, in lessening the irritating contents of the
alimentary canal.”
“But in some maladies it is necessary to restrict the quantity of fluids
taken, or, in other words, to employ a dry diet.”
As it is so customary for invalids to throng to watering places, the
following remarks contain very important cautions.
“The Congress Water at Saratoga,” says Dr. Lee, “though it
possesses active medical qualities, yet, except in diseases attended
with inflammatory action, seldom occasions unpleasant
consequences, unless drank in very large quantities, when it often
causes serious, if not dangerous effects.” Dr. Steel, a physician who
has devoted much attention to this subject, remarks, “About three
pints should be taken, an hour or two before breakfast, and be
followed by exercise, to produce a cathartic effect. Where more is
needed for this effect, add a teaspoonful or two of Epsom salts to
the first tumbler. It should not be drank at all during the remainder
of the day by those who wish to experience the full benefit of its
use. It would be better for those whose complaints render them fit
subjects for its administration, if the fountain should be locked up,
and no one suffered to approach it after the hour of nine or ten in
the morning.”
It is probable that multitudes who frequent mineral springs, not only
lose all benefit, but suffer injury by the excessive use of the water.
Such waters should, by invalids, be taken under the direction of a
physician well acquainted with their nature and uses.
Alcoholic drinks should never be given to the sick, except by
direction of a physician, as they are powerful medical agents, and in
some cases would increase disease.
The acid drinks are ordinarily those most relished by the sick, and
they are, usually, very serviceable, especially in febrile and other
inflammatory attacks. Ice cream and drinks are good for the sick,
especially in fevers.
When a person is debilitated by sickness, the stomach should never
be loaded with rich food. Nor should the palate be tempted by
favorite articles, when no appetite for food exists, as this is the
indication of nature that the stomach is in no order to digest food.
Dr. Lee remarks, “We regard rice as the most valuable of all the
articles of food, in cases of the derangement of the digestive organs.
It nourishes, while it soothes the irritable mucous membrane, and
while it supports strength, never seems to aggravate the existing
disease. For acute, or chronic affections of the alimentary canal, rice
water for drink, and rice jelly for food, seem peculiarly well adapted,
and appear to exert a specific influence in bringing about a recovery.
These preparations are invaluable also in convalescence from acute
fevers and other maladies, and in the summer complaints of young
children.”
“Isinglass is a very pure form of gelatine, and dissolved in milk,
sweetened and flavored, is taken with advantage by convalescents
when recovering from debility.”
Cod sounds, and the American gelatine, are equally good. Calve’s
foot jelly, blanc mange, and other gelatinous food, are among the
best kinds of nourishment, especially in cases of cholera infantum.
The slight quantity of spice or wine used to flavor such articles,
except in peculiar cases, cannot do any injury.
Buttered toast, either dry or dipped, rarely is a suitable article for the
sick, as melted oils are very difficult of digestion. Where there are
strong powers of digestion, it may be proper.
Many cases of illness, among both adults and children, are readily
cured by abstinence from all food. Headaches, disordered stomachs,
and many other attacks, are caused, often, by violating the rules of
health laid down in the preceding chapter, and in consequence,
some part of the system is overloaded, or some of the organs are
clogged. Omitting one, two, or three meals, as the case may be,
gives the system a chance to rest, and thus to gain strength, and
allows the clogged organs to dispose of their burdens. The practice
of giving drugs to “clear out the stomach,” though it may afford the
needed relief, always weakens the system, while abstinence secures
the good result, and yet does no injury.
Said a young gentleman to a distinguished medical practitioner of
Philadelphia,—“Doctor, what do you do for yourself, when you have
a turn of headache, or other slight attack?” “Go without my dinner,”
was the reply. “Well, if that will not do, what do you do then?” “Go
without my supper,” was the answer. “But if that does not cure you,
what then?” “Go without my breakfast. We physicians seldom take
medicines ourselves, or use them in our families, for we know that
starving is better, but we cannot make our patients believe it.”
Many cases of slight indisposition are cured by a change of diet.
Thus, if a person suffers from constipation, and, as the
consequence, has headaches, slight attacks of fever, or dyspepsia,
the cause often may be removed by eating rye mush and molasses
for breakfast, brown bread, baked apples, and other fruits.
In cases of diarrhœa, rice water for drink, and rice pudding or jelly,
will often remove the evil.
In cases of long-continued confinement from sickness, it is very
desirable to have a good variety of articles for the sick, as the invalid
is wearied with the same round, and perhaps may be suffering for
some ingredient of food, which is not found in the articles provided.
For this reason, a large number of receipts of articles for the sick
have been provided in this work. In preparing them, great care
should be used to have every article employed clean and pure, and
to prevent any burning in cooking, as the sensibilities of the sick to
bad tastes and smells are very acute.
It is often the case in dyspepsia and cholera infantum, that jellies of
American gelatine are very much better than any preparations of
farinaceous food, being much more easily digested.
It would be a happy thing for the sick, and a most benevolent
custom, if the young ladies of a place should practise cooking the
various articles for the sick, and carrying them to invalids as an
offering of kindness and sympathy. It would be twice a blessing, first
to the invalid, and quite as much to the young benefactress.
There are many little comforts and alleviations for the sick, which
should be carefully attended to, which are particularly pointed out in
the chapter on the Care of the Sick, in the Domestic Economy. Such,
for example, as keeping a room neat, clean, and in perfect order,
having every article in use sweet and clean, keeping a good supply
of cool water, providing pleasant perfumes, lemons, flowers, and
other objects agreeable to the senses, speaking softly, kindly, and
cheerfully, and reading the Bible and other cheering books of the
kind, whenever it will be acceptable. Be careful to change the linen
next the skin, and the bed linen, often. Be sure to ventilate the room
thoroughly, two or three times a day, as pure air is a great
restorative of health and strength. Wash the skin often, as it has a
great tendency to restore health, and never, except in very peculiar
cases, can do any harm.
Always request a physician to write all his directions, that no mistake
may be made, and nothing be forgotten. Always inquire of him as to
the exact mode of preparing every article ordered, and never
venture to alter, or omit, what he directs, unless you are sure that
you are better qualified to practise than he, in which case he should
be dismissed, and you should assume his duties.
Always keep all medicines in papers and vials, labelled, that poisons
be not given by mistake, or other injurious articles used.
The drawing at Fig. 5 represents a contrivance for the sick, which
ought to be prepared in every village, to rent out to those who need
it.
Fig. 5.
It is called the Water Bed, or Hydrostatic Couch, and is made at an
expense of from twelve to fifteen dollars. The object of it is to
relieve the sufferings of those who, from extreme emaciation, or
from ulcers, or bed sores, are great sufferers from the pressure of
the bed on these sore places. This kind of bed proves a great relief
from this kind of suffering.
It consists of a wooden box, six feet long, and two feet and a half
wide at the top, and the sides gradually sloping inward, making it
fourteen inches deep. This is lined with sheet zinc, to make it water
tight. Over this is thrown, and fastened to the edge of the box, a
sheet of thick India rubber, water-proof cloth, large enough for an
entire lining to the inside of the box. The edges of it are first made
to adhere to the upper edge of the box with spirit varnish, and then
a thin strip of board is nailed on, to fasten it firmly, and make it
water tight. Near the bottom, at A, is a hole and plug, to let off
water; and at B, a tin tube, soldered in the upper part of the outside,
to pour water in. When used, the box is to be filled half full of water,
about blood warmth. Then a woollen blanket and pillow are laid
upon the India rubber cloth, and the patient laid on them, and he
will float as he would in water, and there will be no pressure on any
part of the body greater than is felt when the body is in water.
This is important for all who suffer from bed sores, or sloughing in
protracted fevers, from diseases in the hip-joint, from diseases of
the spine, lingering consumption, and all diseases that compel to a
protracted recumbent position. None but those who have seen, or
experienced the relief and comfort secured to sufferers by this bed,
can conceive of its value. The writer saw the case of a young man,
who was enduring indescribable tortures with the most dreadful
ulcers all over his body, and who had for several days and nights
been unable to sleep, from extreme suffering. This bed was made
for him, as an experiment, after trying every other mode of relief in
vain. It was placed by his bedside, and the water poured in, and
then his friends raised him with the greatest care in a blanket, and
laid him on it. Instantly his groans ceased, an expression of relief
and delight stole over his countenance, and exhausted nature sunk
instantly into the most peaceful and protracted slumbers. And ever
after, he was relieved from his former sufferings. Every hospital,
every alms-house, and every village should have the means of
obtaining such a bed for the many classes of sufferers who would
thus find relief, and it is woman who should interest herself to
secure such a comfort for the sick, who especially are commended
to her benevolent ministries.
The drawing, AA, is the outline of a sick couch, such as would prove
a great comfort in every family, and at a small expense. The
following are the dimensions:—Length of seat inside, six feet three
inches; breadth, twenty-eight inches; height of the seat from the
floor, thirteen inches. The swing frame, dd, is three feet long, and is
fastened three feet from the head-board by a very large pin or
screw. The seat should be of sacking, and a thick hair mattrass, or
cushion for a bed or cushion and be divided into two parts, where
the swing frame is fastened. The frame is fastened by large screws
at ff. The supporter, ee, is fastened by large and strong hinges to
the upper part of the frame, and is moved into the notches made in
the frame of the seat. When in the notch a, the frame is very low;
when in notch b, it is higher; and when at c, it is as high as is
needful to raise the sick. A piece of sacking is to be fastened over
the frame, leaving it loose, especially at the top, and leaving a space
at o, so as to give room for a pillow, and so that the head can be
thrown a little back. The frame and supporter must be thick and
strong. When not in use for the sick, the frame can be laid down,
and the cushion laid over it, and then with a frill fastened in front, it
makes a good-looking and most comfortable chamber couch, or a
lolling sofa for a sitting-room.
Such a couch saves much labor to friends and nurses, because it is
so low, so easily moved, and the nurse can go around it and work on
both sides so easily, while the frame raises the patient with great
ease and comfort. It would be a good plan to engage some
carpenter or cabinet-maker, in every village, to provide such an
article to rent out, and probably it would be in constant demand.
The frame, supporter, hinges, and screws, must be very strong, or
they will break.
Fig. 6 represents a contrivance for securing exercise in the open air
for invalids, which would often prove convenient and agreeable.
Such an article can be easily made of the broken toy of a child,
called a velocipede, or the back wheels of a child’s wagon. Nothing
but shafts are needed, and a common rocking-chair, with a foot-
board nailed across the front rocker, on which to rest the feet. The
chair is then slipped along back to the axle of the wheels, so that the
shafts, when raised, are under the seat, and lift it up. In this way an
invalid can be rolled through yards and gardens with very little
effort, and with great comfort and relief to the imprisoned sufferer,
who perhaps can ride in no other way.
Fig. 6.
There is no way of relieving the weariness and nervousness of an
invalid more effectually than by rubbing the limbs and arms with the
bare hand of a healthful person.
Those who believe in animal magnetism would say, that by this
method, the well person imparts a portion of the magnetism of a
healthful body to aid in restoring the sick. Those who do not believe
in it, will say that it soothes and strengthens the nerves. Either way,
it is a great comfort to a suffering invalid.
It is unhealthful to sleep with a sick person, especially one who has
lung complaints, as the breath and effluvia from the skin sometimes
communicate disease, even in complaints not contagious. Young
children should not sleep with the aged, because their healthful
fluids will be absorbed.
CHAPTER XXIII.
THE PROVIDING AND CARE OF FAMILY
STORES.
The art of keeping a good table, consists, not in loading on a variety
at each meal, but rather in securing a successive variety, a table
neatly and tastefully set, and everything that is on it, cooked in the
best manner.
There are some families who provide an abundance of the most
expensive and choice articles, and spare no expenses in any respect,
who yet have everything cooked in such a miserable way, and a
table set in so slovenly a manner, that a person accustomed to a
really good table, can scarcely taste a morsel with any enjoyment.
On the contrary, there are many tables where the closest economy is
practised, and yet the table-cloth is so white and smooth, the dishes,
silver, glass, and other table articles so bright, and arranged with
such propriety, the bread so white, light, and sweet, the butter so
beautiful, and every other article of food so well cooked, and so
neatly and tastefully served, that everything seems good, and
pleases both the eye and the palate.
A habit of doing everything in the best manner, is of unspeakable
importance to a housekeeper, and every woman ought to aim at it,
however great the difficulties she may have to meet. If a young
housekeeper commences with a determination to try to do
everything in the best manner, and perseveres in the effort, meeting
all obstacles with patient cheerfulness, not only the moral, but the
intellectual tone of her mind is elevated by the attempt. Although
she may meet many insuperable difficulties, and may never reach
the standard at which she aims, the simple effort, persevered in, will
have an elevating influence on her character, while at the same time
she actually will reach a point of excellence far ahead of those who,
discouraged by many obstacles, give up in despair, and resolve to
make no more efforts, and let things go as they will. The grand
distinction between a noble and an ignoble mind is, that one will
control circumstances; the other yields, and allows circumstances to
control her.
It should be borne in mind, that the constitution of man demands a
variety of food, and that it is just as cheap to keep on hand a good
variety of materials in the store-closet, so as to make a frequent
change, as it is to buy one or two articles at once, and live on them
exclusively, till every person is tired of them, and then buy two or
three more of another kind.
It is too frequently the case, that families fall into a very limited
round of articles, and continue the same course from one year to
another, when there is a much greater variety within reach, of
articles which are just as cheap and easily obtained, and yet remain
unthought of and untouched.
A thrifty and generous provider, will see that her store-closet is
furnished with such a variety of articles, that successive changes can
be made, and for a good length of time. To aid in this, a slight
sketch of a well-provided store-closet will be given, with a
description of the manner in which each article should be stored and
kept, in order to avoid waste and injury. To this will be added, modes
of securing a successive variety, within the reach of all in moderate
circumstances.
It is best to have a store-closet open from a kitchen, because the
kitchen fire keeps the atmosphere dry, and this prevents the articles
stored from moulding, and other injury from dampness. Yet it must
not be kept warm, as there are many articles which are injured by
warmth.
A cool and dry place is indispensable for a store-room, and a small
window over the door, and another opening out-doors, is a great
advantage, by securing coolness, and a circulation of fresh air.
Flour should be kept in a barrel, with a flour scoop to dip it, a sieve
to sift it, and a pan to hold the sifted flour, either in the barrel, or
close at hand. The barrel should have a tight cover to keep out mice
and vermin. It is best, when it can be conveniently done, to find, by
trial, a lot of first-rate flour, and then buy a year’s supply. But this
should not be done, unless there are accommodations for keeping it
dry and cool, and protecting it from vermin.
Unbolted flour should be stored in barrels, and always be kept on
hand, as regularly as fine flour.
Indian meal should be purchased in small quantities, say fifteen or
twenty pounds at a time, and be kept in a covered tub or keg. When
new and sweet, it should not be scalded, but when not perfectly
fresh and good when used, it is improved by scalding. It must be
kept very cool and dry, and if occasionally stirred, is preserved more
surely from growing sour or musty.
Rye should be bought in small quantities, say forty or fifty pounds at
a time, and be kept in a keg, or half barrel with a cover.
Buckwheat, Rice, Hominy, and Ground Rice, must be purchased in
small quantities, and kept in covered kegs, or tubs. Several of these
articles are infested with small black insects, and examination must
occasionally be made for them.
Arrowroot, Tapioca, Sago, Pearl Barley, American Isinglass,
Macaroni, Vermacelli, and Oatmeal, are all articles which help to
make an agreeable variety, and it is just as cheap to buy a small
quantity of each, as it is to buy a larger quantity of two or three
articles. Eight or ten pounds of each of these articles of food can be
stored in covered jars, or covered wood boxes, and then they are
always at hand to help make a variety. All of them are very healthful
food, and help to form many delightful dishes for desserts. Some of
the most healthful puddings are those made of rice, tapioca, sago,
and macaroni, while isinglass, or American gelatine, form elegant
articles for desserts, and is also excellent for the sick.
Sugars should not be bought by the barrel, as the brown is apt to
turn to molasses, and run out on to the floor. It is best to keep four
qualities of sugar on hand. Refined loaf for tea, crushed sugar for
the nicest preserves and to use with fruit, nice brown sugar for
coffee, and common brown for cooking and more common use. The
loaf can be stored in the papers, on a shelf. The others should be
kept in close covered kegs, or covered wooden articles made for the
purpose.
Butter must be kept in the dryest and coldest place you can find, in
vessels of either stone, earthen, or wood, and never in tin.
Lard and Drippings must be kept in a dry, cold place, and should not
be salted. Usually the cellar is the best place for them. Earthen, or
stone jars are the best to store them in.
Salt must be kept in the dryest place that can be found. Rock salt is
the best for table salt. It should be washed, dried, pounded, sifted,
and stored in a glass jar, and covered close. It is common to find it
growing damp in the salt stands for the table. It should then be set
by the fire to dry, and afterwards be reduced to fine powder again.
Nothing is more disagreeable than coarse or damp salt on a table.
Vinegar is best made of wine, or cider. Buy a keg, or half barrel of it,
and set it in the cellar, and then keep a supply for the castors in a
junk bottle in the kitchen. If too strong, it eats the pickles.
Pickles never must be kept in glazed ware, as the vinegar forms a
poisonous compound with the glazing.
Oil must be kept in the cellar. Winter strained must be got in cold
weather, as the summer strained will not burn except in warm
weather. The best of lard oil is preferred to every other by those who
use it. Some lard oil is very poor.
Molasses, if bought by the barrel, or half barrel, should be kept in
the cellar. Sugar bakers’ is best for the table, and Porto Rico for
cooking. If bought in small quantities, it should be kept in a
demijohn. No vessel should be corked or bunged, if filled with
molasses, as it will swell, and burst the vessel, or run over.
Hard Soap should be bought by large quantity, and laid to harden on
a shelf, in a very dry place. It is much more economical to buy hard,
than soft soap, as those who use soft soap are very apt to waste it
in using it, as they cannot do with hard soap.
Starch it is best to buy by a large quantity. It comes very nicely put
up in papers, a pound or two in each paper, and packed in a box.
Starch, which by the single pound is five cents a pound, if bought by
the box, is only three cents a pound, and this makes a good deal of
difference, in a large family, by the year. The high-priced starch is
cheapest in the end.
Indigo is not always good. When a good lot is found by trial, it is
best to get enough for a year or two, and store it in a tight tin box.
Coffee it is best to buy by the bag, as it improves by keeping. Let it
hang in the bag, in a dry place, and it loses its rank smell and taste.
Tea, if bought by the box, is about five cents a pound cheaper than
by small quantities. If well put up in boxes lined with lead, it keeps
perfectly. But put up in paper, it soon loses its flavor. It therefore
should, if in small quantities, be put in glass, or tin, and shut tight.
Saleratus should be bought in small quantities, then powdered,
sifted, and kept tight corked in a large mouth glass bottle.
It grows damp if exposed to the air, and then cannot be used
properly.
Raisins should not be bought in large quantities, as they are injured
by time. It is best to buy the small boxes.
Currants for cake should be prepared as directed for cake, and set
by for use in a jar.
Lemon and Orange Peel should be dried, pounded, and set up in
corked glass jars.
Nutmeg, Cinnamon, Cloves, Mace, and Allspice, should be pounded
fine, and corked tight in small glass bottles with mouths large
enough for a junk bottle cork, and then put in a tight tin box, made
for the purpose. Or they can be put in small tin boxes with tight
covers. Essences are as good as spices.
Sweet Herbs should be dried, and the stalks thrown away, and the
rest be kept in corked large mouth bottles, or small tin boxes.
Cream Tartar, Citric and Tartaric Acids, Bicarbonate of Soda, and
Essences, should be kept in corked glass jars. Sal volatile must be
kept in a large-mouth bottle, with a ground glass stopper to make it
air-tight. Use cold water in dissolving it. It must be powdered.
Preserves and Jellies should be kept in glass or stone, in a cool, dry
place, well sealed, or tied with bladder covers. If properly made, and
thus put up, they never will ferment. If it is difficult to find a cool,
dry place, pack the jars in a box, and fill the interstices with sand,
very thoroughly dried. It is best to put jellies in tumblers, or small
glass jars, so as to open only a small quantity at a time.
The most easy way of keeping Hams perfectly is to wrap and tie
them in paper, and pack them in boxes or barrels with ashes. The
ashes must fill all interstices, but must not touch the hams, as it
absorbs the fat. It is much less labor, and quite as certain a mode as
the one previously mentioned. It keeps them sweet, and protects
from all kinds of insects.
After smoked beef, or ham, are cut, hang them in a coarse linen bag
in the cellar, and tie it up to keep out flies.
Keep Cheese in a cool, dry place, and after it is cut, wrap it in a linen
cloth, and keep it in a tight tin box.
Keep Bread in a tin covered box, and it will keep fresh and good
longer than if left exposed to the air.
Welcome to our website – the perfect destination for book lovers and
knowledge seekers. We believe that every book holds a new world,
offering opportunities for learning, discovery, and personal growth.
That’s why we are dedicated to bringing you a diverse collection of
books, ranging from classic literature and specialized publications to
self-development guides and children's books.
More than just a book-buying platform, we strive to be a bridge
connecting you with timeless cultural and intellectual values. With an
elegant, user-friendly interface and a smart search system, you can
quickly find the books that best suit your interests. Additionally,
our special promotions and home delivery services help you save time
and fully enjoy the joy of reading.
Join us on a journey of knowledge exploration, passion nurturing, and
personal growth every day!
ebookbell.com

Cardiac Tissue Engineering 2014th Milica Radisic Lauren D Black Iii

  • 1.
    Cardiac Tissue Engineering2014th Milica Radisic Lauren D Black Iii download https://ebookbell.com/product/cardiac-tissue-engineering-2014th- milica-radisic-lauren-d-black-iii-47742462 Explore and download more ebooks at ebookbell.com
  • 2.
    Here are somerecommended products that we believe you will be interested in. You can click the link to download. Cardiac Tissue Engineering 2nd Kareen Lk Coulombe Lauren D Black Iii https://ebookbell.com/product/cardiac-tissue-engineering-2nd-kareen- lk-coulombe-lauren-d-black-iii-47937244 Cardiac Regeneration And Repair Biomaterials And Tissue Engineering Renke Li https://ebookbell.com/product/cardiac-regeneration-and-repair- biomaterials-and-tissue-engineering-renke-li-4653098 Cardiac Reconstructions With Allograft Tissues 1st Edition Richard A Hopkins Md Auth https://ebookbell.com/product/cardiac-reconstructions-with-allograft- tissues-1st-edition-richard-a-hopkins-md-auth-4413200 Cardiac Reconstructions With Allograft Tissues Hopkins R A https://ebookbell.com/product/cardiac-reconstructions-with-allograft- tissues-hopkins-r-a-1098410
  • 3.
    Cardiac Gene TherapyMethods And Protocols 2nd Edition Kiyotake Ishikawa https://ebookbell.com/product/cardiac-gene-therapy-methods-and- protocols-2nd-edition-kiyotake-ishikawa-46073868 Cardiac Arrhythmias Pacing And Sudden Death Peter Kowey Jonathan P Piccini https://ebookbell.com/product/cardiac-arrhythmias-pacing-and-sudden- death-peter-kowey-jonathan-p-piccini-48356682 Cardiac Mri Certification Exam Sadeer G Alkindi Scott E Janus https://ebookbell.com/product/cardiac-mri-certification-exam-sadeer-g- alkindi-scott-e-janus-48401156 Cardiac Surgical Complications Strategic Analysis And Clinical Review Sandhya K Balaram https://ebookbell.com/product/cardiac-surgical-complications- strategic-analysis-and-clinical-review-sandhya-k-balaram-48949000 Cardiac Ultrasound 1st Edition Antoinette Kenny Kenny Antoinette https://ebookbell.com/product/cardiac-ultrasound-1st-edition- antoinette-kenny-kenny-antoinette-49200226
  • 5.
    CardiacTissue Engineering Milica Radisic Lauren D.Black III Editors Methods and Protocols Methods in Molecular Biology 1181
  • 6.
    ME T HO D S I N MO L E C U L A R BI O LO G Y Series Editor John M. Walker School of Life Sciences University of Hertfordshire Hatfield, Hertfordshire, AL10 9AB, UK For further volumes: http://www.springer.com/series/7651
  • 8.
    Cardiac Tissue Engineering Methodsand Protocols Edited by Milica Radisic InstituteofBiomaterialsandBiomedicalEngineering,DepartmentofChemical EngineeringandAppliedChemistry,UniversityofToronto,Toronto,ON,Canada; TorontoGeneralResearchInstitute,UniversityHealthNetwork,Toronto,ON,Canada Lauren D. Black III DepartmentofBiomedicalEngineering,TuftsUniversity,Medford,MA,USA; Cellular,MolecularandDevelopmentalBiologyProgram, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
  • 9.
    Videos to thisbook can be accessed at http:/ /www.springerimages.com/videos/978-1-4939-1046-5 ISSN 1064-3745 ISSN 1940-6029 (electronic) ISBN 978-1-4939-1046-5 ISBN 978-1-4939-1047-2 (eBook) DOI 10.1007 /978-1-4939-1047-2 Springer New York Heidelberg Dordrecht London Library of Congress Control Number: 2014940960 © Springer Science+Business Media New York 2014 This work is subject to copyright. All rights are reserved by the Publisher, whether the whole or part of the material is concerned, specifically the rights of translation, reprinting, reuse of illustrations, recitation, broadcasting, reproduction on microfilms or in any other physical way, and transmission or information storage and retrieval, electronic adaptation, computer software, or by similar or dissimilar methodology now known or hereafter developed. Exempted from this legal reservation are brief excerpts in connection with reviews or scholarly analysis or material supplied specifically for the purpose of being entered and executed on a computer system, for exclusive use by the purchaser of the work. Duplication of this publication or parts thereof is permitted only under the provisions of the Copyright Law of the Publisher’s location, in its current version, and permission for use must always be obtained from Springer. Permissions for use may be obtained through RightsLink at the Copyright Clearance Center. Violations are liable to prosecution under the respective Copyright Law. The use of general descriptive names, registered names, trademarks, service marks, etc. in this publication does not imply, even in the absence of a specific statement, that such names are exempt from the relevant protective laws and regulations and therefore free for general use. While the advice and information in this book are believed to be true and accurate at the date of publication, neither the authors nor the editors nor the publisher can accept any legal responsibility for any errors or omissions that may be made. The publisher makes no warranty, express or implied, with respect to the material contained herein. Printed on acid-free paper Humana Press is a brand of Springer Springer is part of Springer Science+Business Media (www.springer.com) Editors Milica Radisic Institute of Biomaterials and Biomedical Engineering Department of Chemical Engineering and Applied Chemistry University of Toronto Toronto, ON, Canada Toronto General Research Institute University Health Network Toronto, ON, Canada Lauren D. Black III Department of Biomedical Engineering Tufts University Medford, MA, USA Cellular, Molecular and Developmental Biology Program, Sackler School of Graduate Biomedical Sciences Tufts University School of Medicine Boston, MA, USA
  • 10.
    v Human hearts havea limited regenerative potential, motivating the development of the alternative treatment options for the conditions that result in the loss of beating cardio- myocytes. An example is myocardial infarction that results in a death of tens of millions of ventricular cardiomyocytes that cannot be replaced by the body. It is estimated that five to seven million patients live with myocardial infarction in North America alone. A majority of these patients do not need a surgical intervention, and medical management provides satisfac- tory results. However, over a period of 5 years, one-half of the patients who experience a myocardial infarction will develop heart failure, ultimately requiring heart transplantation. The long-term goal of cardiac tissue engineering is to provide a living, beating, ideally autologous, and non-immunogenic myocardial patch that can restore the contractile function of the failing heart. The engineered tissues could also be used for preclinical drug testing to discover new targets for cardiac therapy and eliminate drugs, cardiac and noncardiac, with serious side effects. It generally involves a combination of suitable cell types, human or non- human cardiomyocytes and supporting cells, with an appropriate biomaterial made out of either synthetic or natural components and cultivation in an environment that reproduces some of the complexity of the native cardiac environment (e.g., electrical, mechanical stimula- tion, passive tension, or topographical cues). This field is still young. The term cardiac tissue engineering usually refers to engineering of myocardial wall in vitro using living and beating cardiomyocytes. The pioneering papers appeared in the late 1990s, and they all utilized either neonatal rat cardiomyocytes or embryonic chick cardiomyocytes as a cell source. Since then, the field has matured signifi- cantly to include a range of approaches that all give cardiac tissues in vitro that are capable of developing contractile force and propagating electrical impulses. Advances in human embryonic stem cell research and induced pluripotent stem cell technology now provide the possibility of generating millions of bona fide human cardiomyocytes. When research in cardiac tissue engineering started some 25 years ago, the issue of a human cell source appeared insurmountable; however the researchers continued to make way, and there are many reports now on the use of human pluripotent stem cells as a source of cardiomyocytes for cardiac tissue engineering. Although early researchers thought that having purified car- diomyocytes in three-dimensional structures would be beneficial, based on analogies with monolayer studies where fibroblasts overgrow cardiomyocytes, there is a consensus in the field now that a mixed cell population is optimal for maintenance of cardiac phenotype and survival of cardiomyocytes in engineered tissues both in vitro and in vivo. The mixed popu- lation usually contains cardiomyocytes, endothelial cells, and a stromal cell type such as fibroblasts or mesenchymal stem cells. Also, there is a consensus that a form of physical stimulation, either mechanical or electrical, or passive tension is required for cardiomyo- cytes to achieve and maintain a differentiated phenotype and in vivo-like functional proper- ties during in vitro cultivation. This book gathers for the first time a collection of protocols on cardiac tissue engineering from pioneering and leading researchers around the globe. Protocols related to cell prepa- ration, biomaterial preparation, cell seeding, and cultivation in various systems are provided. Preface
  • 11.
    vi Our goal isto enable adoption of these protocols in laboratories that are interested in enter- ing the field as well as enable transfer of knowledge between laboratories that are already in this field. We hope that these efforts will lead to standardization, definition of best practices in cardiac tissue cultivation, and direct comparison of various production protocols using controlled in vivo studies that would ultimately lead to translational efforts. Although bio- material patches alone and hydrogels have been investigated in clinical studies focused on myocardial regeneration, a cardiac patch based on living, beating human cardiomyocytes has not yet been tested in humans. Only patches based on non-cardiomyocytes have been tested in humans with mixed results. Bringing a new therapy to the clinic is an overwhelm- ing task, one that we must approach in a collaborative rather than competitive spirit. We hope that sharing of the best protocols in cardiac tissue engineering will enable this goal. Toronto, ON, Canada Milica Radisic Medford, MA, USA Lauren D. Black III, Ph.D. Preface
  • 12.
    vii Contents Preface. . .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v Contributors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix 1 Second Generation Codon Optimized Minicircle (CoMiC) for Nonviral Reprogramming of Human Adult Fibroblasts . . . . . . . . . . . . . . . 1 Sebastian Diecke, Leszek Lisowski, Nigel G. Kooreman, and Joseph C. Wu 2 Scalable Cardiac Differentiation of Human Pluripotent Stem Cells as Microwell-Generated, Size Controlled Three-Dimensional Aggregates . . . . 15 Celine L. Bauwens and Mark D. Ungrin 3 Preparation and Characterization of Circulating Angiogenic Cells for Tissue Engineering Applications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27 Aleksandra Ostojic, Suzanne Crowe, Brian McNeill, Marc Ruel, and Erik J. Suuronen 4 Isolation and Expansion of C-Kit-Positive Cardiac Progenitor Cells by Magnetic Cell Sorting . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 39 Kristin M. French and Michael E. Davis 5 Synthesis of Aliphatic Polyester Hydrogel for Cardiac Tissue Engineering . . . . 51 Sanjiv Dhingra, Richard D. Weisel, and Ren-Ke Li 6 Fabrication of PEGylated Fibrinogen: A Versatile Injectable Hydrogel Biomaterial . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 61 Iris Mironi-Harpaz, Alexandra Berdichevski, and Dror Seliktar 7 Natural Cardiac Extracellular Matrix Hydrogels for Cultivation of Human Stem Cell-Derived Cardiomyocytes . . . . . . . . . . . . . . . . . . . . . . . . 69 Donald O. Freytes, John D. O’Neill, Yi Duan-Arnold, Emily A. Wrona, and Gordana Vunjak-Novakovic 8 Magnetically Actuated Alginate Scaffold: A Novel Platform for Promoting Tissue Organization and Vascularization. . . . . . . . . . . . . . . . . . 83 Yulia Sapir, Emil Ruvinov, Boris Polyak, and Smadar Cohen 9 Shrink-Induced Biomimetic Wrinkled Substrates for Functional Cardiac Cell Alignment and Culture . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 97 Nicole Mendoza, Roger Tu, Aaron Chen, Eugene Lee, and Michelle Khine 10 Injectable ECM Scaffolds for Cardiac Repair . . . . . . . . . . . . . . . . . . . . . . . . . . 109 Todd D. Johnson, Rebecca L. Braden, and Karen L. Christman 11 Generation of Strip-Format Fibrin-Based Engineered Heart Tissue (EHT) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 121 Sebastian Schaaf, Alexandra Eder, Ingra Vollert, Andrea Stöhr, Arne Hansen, and Thomas Eschenhagen
  • 13.
    viii 12 Cell Tri-Culturefor Cardiac Vascularization . . . . . . . . . . . . . . . . . . . . . . . . . . 131 Ayelet Lesman, Lior Gepstein, and Shulamit Levenberg 13 Cell Sheet Technology for Cardiac Tissue Engineering . . . . . . . . . . . . . . . . . . 139 Yuji Haraguchi, Tatsuya Shimizu, Katsuhisa Matsuura, Hidekazu Sekine, Nobuyuki Tanaka, Kenjiro Tadakuma, Masayuki Yamato, Makoto Kaneko, and Teruo Okano 14 Design and Fabrication of Biological Wires . . . . . . . . . . . . . . . . . . . . . . . . . . . 157 Jason W. Miklas, Sara S. Nunes, Boyang Zhang, and Milica Radisic 15 Collagen-Based Engineered Heart Muscle. . . . . . . . . . . . . . . . . . . . . . . . . . . . 167 Malte Tiburcy, Tim Meyer, Poh Loong Soong, and Wolfram-Hubertus Zimmermann 16 Creation of a Bioreactor for the Application of Variable Amplitude Mechanical Stimulation of Fibrin Gel-Based Engineered Cardiac Tissue . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 177 Kathy Y. Morgan and Lauren D. Black III 17 Preparation of Acellular Myocardial Scaffolds with Well-Preserved Cardiomyocyte Lacunae, and Method for Applying Mechanical and Electrical Simulation to Tissue Construct . . . . . . . . . . . . . . . . . . . . . . . . . 189 Bo Wang, Lakiesha N. Williams, Amy L. de Jongh Curry, and Jun Liao 18 Patch-Clamp Technique in ESC-Derived Cardiomyocytes . . . . . . . . . . . . . . . . 203 Jie Liu and Peter H. Backx 19 Optogenetic Control of Cardiomyocytes via Viral Delivery . . . . . . . . . . . . . . . 215 Christina M. Ambrosi and Emilia Entcheva 20 Methods for Assessing the Electromechanical Integration of Human Pluripotent Stem Cell-Derived Cardiomyocyte Grafts. . . . . . . . . . . 229 Wei-Zhong Zhu, Dominic Filice, Nathan J. Palpant, and Michael A. Laflamme 21 Quantifying Electrical Interactions Between Cardiomyocytes and Other Cells in Micropatterned Cell Pairs. . . . . . . . . . . . . . . . . . . . . . . . . . 249 Hung Nguyen, Nima Badie, Luke McSpadden, Dawn Pedrotty, and Nenad Bursac Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 263 Contents
  • 14.
    ix CHRISTINA M. AMBROSI,PH.D. • Department of Biomedical Engineering, Institute for Molecular Cardiology, Stony Brook University, Stony Brook, NY, USA PETER H. BACKX • Department of Physiology and Medicine, University of Toronto, Toronto, ON, Canada; The Heart and Stroke/Richard Lewar Centre of Excellence, Toronto, ON, Canada; Division of Cardiology, University Health Network, Toronto, ON, Canada NIMA BADIE • Department of Biomedical Engineering, Duke University, Durham, NC, USA CELINE L. BAUWENS • Centre for Commercialization of Regenerative Medicine, Toronto, ON, Canada ALEXANDRA BERDICHEVSKI • Faculty of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel LAUREN D. BLACK III, PH.D. • Department of Biomedical Engineering, Tufts University, Medford, MA, USA; Cellular, Molecular and Developmental Biology Program, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA REBECCA L. BRADEN, M.S. • Department of Bioengineering, University of California San Diego, La Jolla, CA, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA NENAD BURSAC, PH.D. • Department of Biomedical Engineering, Duke University, Durham, NC, USA AARON CHEN • Department of Chemical Engineering and Materials Science, University of California, Irvine, CA, USA KAREN L. CHRISTMAN, PH.D. • Department of Bioengineering, University of California San Diego, La Jolla, CA, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA SMADAR COHEN, PH.D. • Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, The Center for Regenerative Medicine and Stem Cell (RMSC) Research, Ben-Gurion University of the Negev, Beer-Sheva, Israel; The Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer-Sheva, Israel SUZANNE CROWE • Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, Canada MICHAEL E. DAVIS, PH.D. • Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA AMY L. DE JONGH CURRY • Department of Biomedical Engineering, University of Memphis, Memphis, TN, USA SANJIV DHINGRA • Regenerative Medicine Program, Institute of Cardiovascular Sciences, St. Boniface Research Centre, University of Manitoba, Winnipeg, MB, Canada SEBASTIAN DIECKE • Lorry I. Lokey Stem Cell Research Building, Stanford University School of Medicine, Stanford, CA, USA YI DUAN-ARNOLD • Department of Biomedical Engineering, Columbia University, New York, NY, USA ALEXANDRA EDER • Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; DZHK (German Centre for Cardiovascular Research), Hamburg, Germany Contributors
  • 15.
    x EMILIA ENTCHEVA, PH.D.• Department of Biomedical Engineering, Institute for Molecular Cardiology, Stony Brook University, Stony Brook, NY, USA THOMAS ESCHENHAGEN • Department of Experimental Pharmacology and Toxicology, University Medical CenterHamburg-Eppendorf (UKE), Hamburg, Germany; DZHK (German Centre for Cardiovascular Research), Hamburg, Germany DOMINIC FILICE • Department of Bioengineering, Institute for Stem Cells and Regenerative Medicine, University of Washington, Seattle, WA, USA KRISTIN M. FRENCH • Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA DONALD O. FREYTES • New York Stem Cell Foundation, New York, NY, USA LIOR GEPSTEIN • The Sohnis Family Research Laboratory for Cardiac Electrophysiology and Regenerative medicine, the Bruce Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa, Israel ARNE HANSEN • Department of Experimental Pharmacology and Toxicology, University Medical CenterHamburg-Eppendorf (UKE), Hamburg, Germany; DZHK (German Centre for Cardiovascular Research), Hamburg, Germany YUJI HARAGUCHI • Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women’s Medical University, Shinjuku-ku, Tokyo, Japan TODD D. JOHNSON, M.S. • Department of Bioengineering, University of California San Diego, La Jolla, CA, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA MAKOTO KANEKO • Department of Mechanical Engineering, Graduate School of Engineering, Osaka University, Yamadaoka, Suita, Japan MICHELLE KHINE, PH.D. • Department of Biomedical Engineering, University of California, Irvine, CA, USA; Department of Chemical Engineering, University of California, Irvine, CA, USA NIGEL G. KOOREMAN • Lorry I. Lokey Stem Cell Research Building, Stanford University School of Medicine, Stanford, CA, USA MICHAEL A. LAFLAMME, M.D., PH.D. • Department of Pathology, Institute for Stem Cells and Regenerative Medicine, University of Washington, Seattle, WA, USA EUGENE LEE • Department of Biomedical Engineering, University of California, Irvine, CA, USA AYELET LESMAN • Department of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA; Department of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel SHULAMIT LEVENBERG • Department of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel REN-KE LI, M.D., PH.D. • Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, ON, Canada; Division of Cardiac Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada JUN LIAO, PH.D. • Tissue Bioengineering Laboratory, Department of Biological Engineering, Mississippi State University, Mississippi State, MS, USA LESZEK LISOWSKI • Gene Transfer, Targeting and Therapeutics Facility, Salk Institute for Biological Studies, San Diego, CA, USA JIE LIU • Department of Physiology and Medicine, University of Toronto, Toronto, ON, Canada; Materials Science, University of California, Irvine, CA, USA KATSUHISA MATSUURA • Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women’s Medical University, Shinjuku-ku, Tokyo, Japan BRIAN MCNEILL • Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, Canada Contributors
  • 16.
    xi LUKE MCSPADDEN •Department of Biomedical Engineering, Duke University, Durham, NC, USA NICOLE MENDOZA • Department of Biomedical Engineering, University of California, Irvine, CA, USA TIM MEYER • Institute of Pharmacology, Heart Research Center Göttingen (HRCG), University Medical Center Göttingen, Göttingen, Germany; DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany JASON W. MIKLAS • Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada IRIS MIRONI-HARPAZ • Faculty of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel KATHY Y. MORGAN • Department of Biomedical Engineering, Tufts University, Medford, MA, USA HUNG NGUYEN • Department of Biomedical Engineering, Duke University, Durham, NC, USA SARA S. NUNES • Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Toronto General Research Institute, University Health Network, Toronto, ON, Canada TERUO OKANO • Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women’s Medical University, Shinjuku-ku, Tokyo, Japan JOHN D. O’NEILL • Department of Biomedical Engineering, Columbia University, New York, NY, USA ALEKSANDRA OSTOJIC • Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, Canada NATHAN J. PALPANT • Department of Pathology, Institute for Stem Cells and Regenerative Medicine, University of Washington, Seattle, WA, USA DAWN PEDROTTY • Department of Biomedical Engineering, Duke University, Durham, NC, USA BORIS POLYAK, PH.D. • Department of Surgery, Drexel University College of Medicine, Philadelphia, PA, USA; Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, USA MILICA RADISIC • Institute of Biomaterials and Biomedical Engineering, Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada; Toronto General Research Institute, University Health Network, Toronto, ON, Canada MARC RUEL • Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, Canada EMIL RUVINOV, PH.D. • Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, Beer-Sheva, Israel YULIA SAPIR, M.SC. • Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, Beer-Sheva, Israel SEBASTIAN SCHAAF • Department of Experimental Pharmacology and Toxicology, University Medical CenterHamburg-Eppendorf (UKE), Hamburg, Germany; DZHK (German Centre for Cardiovascular Research), Hamburg, Germany HIDEKAZU SEKINE • Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women’s Medical University, Shinjuku-ku, Tokyo, Japan DROR SELIKTAR • Faculty of Biomedical Engineering, Technion—Israel Institute of Technology, Haifa, Israel; Nanoscience and Nanotechnology Initiative, National University of Singapore, Singapore, Singapore Contributors
  • 17.
    xii TATSUYA SHIMIZU •Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women’s Medical University, Shinjuku-ku, Tokyo, Japan POH LOONG SOONG • Institute of Pharmacology, Heart Research Center Göttingen (HRCG), University Medical Center Göttingen, Göttingen, Germany; DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany ANDREA STÖHR • Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; DZHK (German Centre for Cardiovascular Research), Hamburg, Germany ERIK J. SUURONEN • Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, Canada KENJIRO TADAKUMA • Department of Mechanical Engineering, Graduate School of Engineering, Osaka University, Yamadaoka, Suita, Japan NOBUYUKI TANAKA • Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women’s Medical University, Shinjuku-ku, Tokyo, Japan MALTE TIBURCY • Institute of Pharmacology, Heart Research Center Göttingen (HRCG), University Medical Center Göttingen, Göttingen, Germany; DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany ROGER TU • Department of Biological Sciences, University of California, Irvine, CA, USA MARK D. UNGRIN • Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada INGRA VOLLERT • Department of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany; DZHK (German Centre for Cardiovascular Research), Hamburg, Germany GORDANA VUNJAK-NOVAKOVIC • Department of Biomedical Engineering, Columbia University, New York, NY, USA BO WANG • Tissue Bioengineering Laboratory, Department of Biological Engineering, Mississippi State University, Mississippi State, MS, USA; Department of Surgery, Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Northwestern University, Chicago, IL, USA RICHARD D. WEISEL • Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network, Toronto, ON, Canada; Department of Surgery, Division of Cardiac Surgery, University of Toronto, Toronto, ON, Canada LAKIESHA N. WILLIAMS • Tissue Bioengineering Laboratory, Department of Biological Engineering, Mississippi State University, Mississippi State, MS, USA EMILY A. WRONA • New York Stem Cell Foundation, New York, NY, USA JOSEPH C. WU, M.D., PH.D. • Lorry I. Lokey Stem Cell Research Building, Stanford University School of Medicine, Stanford, CA, USA MASAYUKI YAMATO • Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women’s Medical University, Shinjuku-ku, Tokyo, Japan BOYANG ZHANG • Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada WEI-ZHONG ZHU • Department of Pathology, Institute for Stem Cells and Regenerative Medicine, University of Washington, Seattle, WA, USA WOLFRAM-HUBERTUS ZIMMERMANN, M.D. • Institute of Pharmacology, Heart Research Center Göttingen (HRCG), University Medical Center Göttingen, Göttingen, Germany; DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany Contributors
  • 18.
    1 Milica Radisic andLauren D. Black III (eds.), Cardiac Tissue Engineering: Methods and Protocols, Methods in Molecular Biology, vol. 1181, DOI 10.1007/978-1-4939-1047-2_1, © Springer Science+Business Media New York 2014 Chapter 1 Second Generation Codon Optimized Minicircle (CoMiC) for Nonviral Reprogramming of Human Adult Fibroblasts Sebastian Diecke, Leszek Lisowski, Nigel G. Kooreman, and Joseph C. Wu Abstract The ability to induce pluripotency in somatic cells is one of the most important scientific achievements in the fields of stem cell research and regenerative medicine. This technique allows researchers to obtain pluripotent stem cells without the controversial use of embryos, providing a novel and powerful tool for disease modeling and drug screening approaches. However, using viruses for the delivery of reprogram- ming genes and transcription factors may result in integration into the host genome and cause random mutations within the target cell, thus limiting the use of these cells for downstream applications. To overcome this limitation, various non-integrating techniques, including Sendai virus, mRNA, minicircle, and plasmid- based methods, have recently been developed. Utilizing a newly developed codon optimized 4-in-1 minicircle (CoMiC), we were able to reprogram human adult fibroblasts using chemically defined media and without the need for feeder cells. Key words Human induced pluripotent stem cells (hiPSC), Minicircle, Reprogramming, Disease modeling, Chemically defined, Differentiation 1 Introduction Somatic cells, such as human adult fibroblasts, can be reverted to an embryonic stem cell (ESC)-like state by inducing the expression of specific genes and transcription factors that regulate the mainte- nance of pluripotency in embryonic stem cells [1]. These so-called core factors of pluripotency, namely OCT4, SOX2, NANOG, and two oncogenes (c-MYC and LIN28), initiate the resetting of the epigenetic profile of a terminal differentiated somatic cell and acti- vate the molecular circuitry of pluripotency [2–4]. The resultant induced pluripotent stem cells (iPSCs) have similar morphology, growth characteristics, and gene expression profiles as hESCs [5]. Furthermore, the capacity of iPSCs to differentiate into all other cell types of the human body has opened up new clinical opportunities for developing successful personalized stem cell-based therapies
  • 19.
    2 and drug discoveryapproaches. Initially iPSCs were produced using retroviral and lentiviral vectors that are known to integrate into the chromosomes of the target cells, potentially causing dis- ruption of gene transcription and subsequent tumor formation [6]. Therefore, various laboratories have developed additional non-integrating reprogramming techniques such as Sendai virus, mRNA, and protein- or DNA-based methods [7–11]. In order to induce the transformation of a nullipotent somatic cell into a pluripotent state, it is necessary to express the right stoi- chiometry of the reprogramming factors within the same trans- fected cell [12]. One elegant approach to achieve this requirement uses polycistronic constructs in which the four reprogramming factors are controlled by one promoter separated by a different self-cleaving 2a peptide sequence [13]. The disadvantage of such polycistronic plasmids is their large size, which might decrease the transfection efficiency in the adult human fibroblast. To over- come this problem, we cloned the polycistronic expression cassette into a minicircle plasmid backbone. Minicircles are special episomal DNA vectors devoid of any bacterial plasmid backbone (Fig. 1a). Therefore minicircles are significantly smaller than standard plasmids, minicircle after induction unpurified minicircle after induction purified 1 kb plus ladder 2 kb 7 kb chromosomal bacterial DNA 1 2 3 1 2 3 1 2 3 5 4 1 2 3 minicircle (Nde1 digest) Parental minicircle backbone (HindIII digest) Parental minicircle backbone (Nde1 digest) 4 5 minicircle (HindIII digest) Parental minicircle backbone (Nde1/HindIII digest) 2 kb 7 kb Before Arabinose Induction a b c After Arabinose Induction Parental 4-in-1 CoMiC 11.6 kb 4-in-1 CoMiC 7.6 kb Fig. 1 (a) Map of parental 4-in-1 reprogramming minicircle before and after arabinose induction. (b) Control digest to verify quality of the minicircle production. (c) Gel showing bands representing the minicircle after induction, both before and after purification steps Sebastian Diecke et al.
  • 20.
    3 which in turnenhances their transfection efficiency and the survival rate of the target cells [14]. Furthermore, it has been shown that minicircle constructs sustain a higher expression rate of their harboring transgenes over a longer period of time. Although the minicircle theoretically should not integrate into the genome of the target cell, there is still a relatively small chance of integration. Therefore, it is important to screen the derived iPSCs for random integration using specific PCR primers or Southern blot experi- ments [15]. The following protocol details the entire process of repro- gramming human adult fibroblasts into human iPSCs: from disso- ciating fibroblasts from patient skin biopsy samples, to the reprogramming and characterization of the newly derived iPSC line. The time from induction of pluripotency to expansion of the subsequent iPSC line to passage 3 is approximately 1 month (Fig. 2b). The workflow of the reprogramming process is summa- rized in Fig. 3. The final part of the protocol explains the pluripo- tency of the iPSCs is confirmed using immunostaining (Fig. 4a). However, it will also be necessary to perform further experiments, including directed differentiation into specific germ layers and teratoma formation assay, to verify pluripotency (Fig. 4b, c) [16]. It should also be mentioned that the minicircle reprogramming Transfection using unpurified minicircle a b Low transfection efficiency, high cell death Higher transfection efficiency, less cell death Transfection using purified minicircle 0 3 13 16 20 day Reprogramming timeline Phase Tomato TRA-1-60 iPSC colony on Matrigel 30 Fig.2 (a) Representative bright field and fluorescent (Tomato) images demonstrating the transfection efficiency and cell survival of unpurified and purified minicircle. (b) Timeline showing the expected phase contrast and fluorescent images at various stages of the reprogramming process.At day 20, pluripotency was confirmed by TRA-1-60 immunofluorescent staining iPSC Generation Using a Codon Optimized Minicircle Plasmid
  • 21.
    4 technique still hasa very low efficiency and therefore should be only used by researchers with iPSC experience as an alternative method for obtaining integration-free induced pluripotent stem cells, or if it is necessary to compare different reprogramming techniques. 1 3 10 Fibroblast-Media E8-Media 20 12 day + Nab + AA Transfection and seeding cells onto 2 Matrigel plates Picking of individual iPSC clones (use 10 µM Rock inhibitor Y27632) 7 9 11 13 5 0 -1 E7-Media split 1.1 x106 fibroblast onto a new dish 50% 50% Fig. 3 Timeline for reprogramming adult human fibroblasts into induced pluripotent stem cells using CoMiC Fig. 4 Immunofluorescence staining demonstrating (a) pluripotency markers NANOG, OCT4 and TRA-1-81, DAPI, and bright field images. (b) Immunofluorescent staining of iPSC-derived cardiomyocytes demonstrating positive staining of cardiac markers connexin 43 and cardiac troponin as well as DAPI. (c) Representative H&E staining from teratomas to confirm the pluripotency status of the generated iPSCs Sebastian Diecke et al.
  • 22.
    5 2 Materials 1. Fibroblastmedia: Low glucose Dulbecco’s Modified Eagle Media (DMEM low glucose) with 20 % fetal bovine serum (FBS). 2. iPSC media: Essential 8 (combine supplement with base media) and Essential 7 (Essential 6 combined with base media and supplemented with 100 ng/mL FGF2). 1. Phosphate buffered saline (PBS). 2. Collagenase IV. 3. Penicillin/streptomycin. 4. Lennox L Broth (LB) base. 5. Sodium hydroxide (NaOH) solution, 1 M. 6. L-(+)-Arabinose ≥99 %, distilled water (dH2O). 7. Kanamycin sulfate, 100×. 8. Matrigel—Growth factor reduced, hESC-qualified. 9. DMEM/F12. 10. TrypLE Express, store at room temperature (RT), there is no need to warm to 37 °C before use. 11. 0.25 % Trypsin/Ethylenediaminetetraacetic acid (EDTA). 12. Accutase. 13. Y27632 (ROCK inhibitor): 10 mM stock in Ultrapure water, store at −20 °C. 14. Ultrapure water. 15. Sodium butyrate (NaB). 16. Antibodies (see Table 1). 17. 4 % paraformaldehyde (PFA) solution. 18. Triton-X 100. 19. Bovine serum albumin (BSA). 20. Goat Serum. 21. Tween-20. 22. Vectashield mounting medium with DAPI. 23. Qiagen Plasmid Plus Maxi Kit. 24. DNAase (2,000 U/ml). 25. Gelatin. 26. Neon Buffer R. 27. Qiagen Plasmid Plus Maxi kit. 28. 4 % Paraformaldehyde (PFA). 29. New England biolabs enzymes HindIII and Nde1. 2.1 Media 2.2 Reagents iPSC Generation Using a Codon Optimized Minicircle Plasmid
  • 23.
    6 Table 1 List of antibodies and dilutions used for characterization of human induced pluripotent stem cells and differentiated cardiomyocytes. Staining protocols were optimized with the antibodies from the listed suppliers. Optimization may be required if different antibodies are used Antigen Supplier Product no. Dilution Raised in Secondary (1:400) Supplier Product no. Connexin-43 SIGMA C6219 1:400 Rabbit Alexa Fluor ® 488 Goat Anti-Rabbit IgG (H + L) Life Technologies A-11037 c Troponin-T Thermo Scientifi c MS-295-P 1:400 Mouse Alexa Fluor ® 488 Goat Anti-Mouse IgG (H + L) Life Technologies A-11001 NANOG Santa Cruz SC-33759 1:100 Rabbit Alexa Fluor ® 488 Goat Anti-Rabbit IgG (H + L) Life Technologies A-11037 OCT4 Santa Cruz SC-9081 1:100 Rabbit Alexa Fluor ® 488 Goat Anti-Rabbit IgG (H + L) Life Technologies A-11037 Tra-1-81 Millipore mab4381 1:250 Mouse Alexa Fluor ® 488 Goat Anti-Mouse IgG (H + L) Life Technologies A-11001 Sebastian Diecke etal.
  • 24.
    7 1. 6-well cellculture plates (surface area=9.8 cm2 ) coated with 2 mL Matrigel. 2. 12-well cell culture plates (surface area=3.8 cm2 ) coated with 1 mL Matrigel. 3. 10 cm tissue culture plates (surface area=58.95 cm2 ) coated with 12 mL Matrigel. 4. 90 mm petri dishes. 5. 15 and 50 mL polypropylene conical tubes. 6. 2 mL plastic aspiration pipettes. 7. 5, 10, 25, and 50 mL plastic pipettes. 8. 250 and 500 mL polyethersulfone (PES) media filters. 9. 8-chamber slides. 10. T225 flasks. 11. Coverslips. 12. Parafilm. 13. Countess Cell Counter, slides, and trypan blue. 14. Tissue culture incubator capable of 37 °C, 5 % CO2, and 85 % relative humidity (such as New Brunswick Galaxy 170R). 15. Dual gas tissue culture incubator capable of 37 °C, 5 % CO2, 5 % O2, and 85 % relative humidity with split inner door. 16. Centrifuge. 17. Inverted tissue culture microscope (such as Nikon Ti) with heated stage. 18. Autoclave. 19. Orbital shaker. 20. Spectrophotometer. 21. Waterbath. 22. Neon Transfection device Life Technologies. 23. Nalgene centrifuge bottle. 3 Methods 1. Prepare 1,200 mL of LB Broth media by adding 24 g of LB Broth Base to 1,200 mL of distilled water (hH2O) and divide this over the three culture flasks. 2. Autoclave the flasks using the “liquid cycle.” 3. After cooling down of the flasks to room temperature, add 4 mL of Kanamycin 100× to each one of the culture flasks. 4. In the morning, pick one clone from a parental plasmid trans- formed bacterial plate in 5 mL LB media containing kanamycin 2.3 Equipment 3.1 Minicircle Production iPSC Generation Using a Codon Optimized Minicircle Plasmid
  • 25.
    8 (50 μg/mL) andgrow this pre-culture until the evening at 37 °C with shaking at 250 rpm. 5. In the evening, grow an overnight culture by combining 100 μL of the pre-culture with each of the flasks containing media with kanamycin and culture overnight (16–18 h) at 37 °C with shaking at 250 rpm (see Note 1). 6. The optical density (OD, absorbance) of the overnight culture should be between 3.75 and 4.25. 7. Prepare a minicircle induction mix comprising 1 volume of fresh LB Broth (1,200 mL), 4 % 1 N NaOH (48 mL), and 1 % L-Arabinose (12 g) and mix well. 8. Add 400 mL of the induction mix to each of the three flasks with the overnight culture and mix well. 9. Incubate the combined mix at 32 °C with shaking at 250 rpm for 6–8 h. 10. Harvest all the culture flasks, transfer them to centrifuge bottle, and pellet the bacteria (6,000×g). 11. Isolate minicircles according to the Qiagen Plasmid Plus Maxi kit instructions with certain modifications (see Notes 2 and 3). 12. Confirm the successful minicircle induction by digestion using HindIII and Nde1. Nde1 linearizes only the parental back- bone but not the minicircle. Therefore, you should expect one band around 7.7 kb when digesting the minicircle with both enzymes (Fig. 1b). 1. All of these steps should be performed in a sterile tissue culture hood. Prepare a fibroblast digestion solution by dissolving 1 mg/mL Collagenase IV in DMEM/F12 and sterile filter it through a 0.2 μm pore size filter. 2. Transfer the skin punch biopsy into a 15 mL conical tube con- taining 3 mL PBS+penicillin/streptomycin (1:100), repeat this step three times. 3. Transfer the tissue into a sterile 10 cm plate and add 1 mL Collagenase IV. Use two scalpels to cross cut the tissue into small pieces while keeping the tissue immersed in Collagenase IV. 4. Additionally, add 2 mL Collagenase IV solution to the minced tissue and pipette it up and down. Transfer the Collagenase IV/tissue suspension into a 15 mL conical tube and add 25 μL of 0.5 M EDTA and 20 μL of DNase. Afterwards, incubate the mixture for 3 h in a tissue culture incubator at 37 °C. 5. Following the 3-h incubation time, pipette the tissue pieces up and down several times to obtain single-cell fibroblasts. 6. Add 12 mL fibroblast media containing 1× penicillin/ streptomycin to stop the Collagenase IV digestion. 3.2 Fibroblast Isolation from a Patient Skin Punch Biopsy Sebastian Diecke et al.
  • 26.
    9 7. Centrifuge for4 min at 300×g. 8. Resuspend the pellet in 2 mL fibroblast media containing 1× penicillin/streptomycin solution, and plate it in one well of a 0.1 % gelatin-coated 6-well plate. Depending on the size of your tissue sample, you may need to plate the cells in more than one well of a 6-well plate. To prepare gelatin-coated plates, gelatin stock (10 %) is dissolved in PBS, 1 ml of this solution is pipetted into a well of a 6-well plate and incubated for 1 h at 37 °C. 9. Incubate the cells for at least 4 days without changing the media, until some of the fibroblasts start to attach and propa- gate. After this, change the media every other day. 10. Once the cells become confluent, split using 0.25 % Trypsin– EDTA for 10 min. Plate the cells onto a 0.1 % gelatin-coated T225 flask to further expand and freeze multiple vials of fibro- blast after the flask becomes confluent. 1. Thaw stock bottle of Matrigel at 4 °C overnight. 2. Keep Matrigel on ice, and add 300 μL Matrigel to 50 mL of cold DMEM/F12 media. Add 2 mL to each well of a 6-well plate. (Alternatively use one Matrigel aliquot per 20 mL cold DMEM/F12 media and cover four 10 cm plates using 5 mL.) 3. Place the plates in the fridge and allow the Matrigel to polym- erize overnight. 1. The target fibroblast cell line should be thawed and cultured for at least 3 days before starting the reprogramming experi- ment. Additionally, it is beneficial to further purify the minicir- cle DNA using the Zymoclean Gel DNA Recovery Kit to eliminate residual chromosomal bacterial DNA of the minicir- cle preparation (Fig. 1c). This step will increase the cell survival after the electroporation (Fig. 2a). 2. Plate 1.1×106 human fibroblast in a 10 cm tissue culture plate 1 day before starting the reprogramming experiment. 3. On the following day (day 0), trypsinize the cells using Trypsin 0.25 % EDTA for 10 min to obtain a single cell suspension. 4. Wash off the trypsin using 10 mL of DMEM plus 20 % FBS and centrifuge for 3 min at 300×g. 5. Aspirate the media, and dissolve the cell pellet in 100–105 μL Neon Buffer R (the volume depends on the amount of plasmid you have to use to get 10–12 μg DNA). The final volume of buffer, DNA, and cells should be around 110 μL. 6. Perform the transfection using a 100 μL Neon tip and the following settings: 1,600 V, 10 ms, and 3 pulses (transfection efficiency should be more than 50 %). 3.3 Prepare Matrigel Plates 3.4 Reprogramming of Human Fibroblasts Using Neon Transfection and CoMiC iPSC Generation Using a Codon Optimized Minicircle Plasmid
  • 27.
    10 7. Plate thecells equally distributed onto two Matrigel-coated plates (5.5×105 cells) in fibroblast media. 8. On the following morning (day 1), change the media to fibroblast media and add sodium butyrate (0.2 mM) plus 50 μg/mL ascorbic acid. 9. On day 3, change the media to fibroblast media and add sodium butyrate (0.2 mM) plus 50 μg/mL ascorbic acid. 10. On day 5, change the media to 50:50 fibroblast media:Essential 7 media and add sodium butyrate (0.2 mM) plus 50 μg/mL ascorbic acid. 11. On days 7–13, change the media every other day until the first pre-iPSC colonies appear. Change the media to Essential 7 and add sodium butyrate (0.2 mM) plus 50 μg/mL ascorbic acid. 12. Between days 14 and 20, after you recognize the first colonies, switch to Essential 8 media and hypoxic culture conditions. Around day 20, the iPSC colonies should be big enough for manual picking under the microscope. Pick six individual iPSC clones using the Vitrolife stem cell cutting tool with a 10 μL tip and transfer them into six different wells of a Matrigel-coated plate with Essential 8 media in presence of 10 μM ROCK Inhibitor (Y-27632). If it is difficult to detach the iPSC colonies from the surrounding fibroblasts, you also can scrape those fibroblasts away to make room for the outgrowing iPSC colony. After 2 days, the iPSC clone should be large enough to pick. 1. After 5–7 days, the individual colonies become big enough to dissociate into single cells using 0.5 mL TrypLE for 8 min. Detach the remaining colonies by pipetting them up and down with a 1 mL pipette. 2. Wash the cells once with 5 mL Essential 8 media. 3. Centrifuge for 3 min at 300×g. 4. Passage the cells into one well of a Matrigel-coated 6-well plate containing 2 mL Essential 8 media. 5. After 5–7 days, colonies will have grown out and become dense enough to split onto a Matrigel-coated 10 cm plate. Therefore incubate the cells with 1 mL Accutase for 8 min, wash using 9 mL Essential 8 media, and centrifuge for 3 min at 300×g. 6. Seed the cells onto a Matrigel-coated 10 cm plate in Essential 8 media in the presence of 10 μM Y27632. 7. Once the 10 cm plate become confluent, freeze down 5 vials (passage 3 iPS cells) and continue to grow at a 1:6 split ratio, freezing vials every 10 passages. 8. Grow cells to at least passage 5 to test for the expression of the pluripotency marker genes and to passage 15 before testing for efficient differentiation. 3.5 Colony Purification and Expansion Sebastian Diecke et al.
  • 28.
    11 1. Coat 8-chamberslides with Matrigel (200 μL per chamber) as described above in Subheading 3.3. 2. Seed iPSCs (1×104 cells/chamber) or differentiated cardio- myocytes (1.75×104 cells/chamber) as a negative control, on 8-chamber slides in a final volume of 200 μL of the appropriate medium. 3. Place the chamber slides in 90 mm Petri dishes and store in a 37 °C incubator. 4. Allow cells to attach and grow for ~24–72 h and fix as follows (see Note 4). 5. Aspirate medium and wash cells 3× with PBS. 6. Add to each chamber 100 μL 4 % Paraformaldehyde (PFA) in PBS for 20 min at RT. 7. Aspirate PFA and wash 3× with 200 μL PBS. 8. Aspirate PBS and replace with 500 μL of fresh PBS. 9. Seal slides with parafilm and place at 4ºC until needed for staining (keep for up to 7 days). 10. Incubate with 100 μL of permeabilization solution (0.2 % Triton X-100 in PBS), for 15 min at room temperature (see Note 5). 11. Wash twice with PBS, for 5 min at room temperature. 12. Incubate with blocking solution (2 % BSA or 4 % Goat Serum or 2 % FBS in PBS), for 1 h at room temperature or overnight at 4ºC. 13. Wash once in PBS, for 5 min at room temperature. 14. Incubate overnight at 4 °C with primary antibody (Table 1) diluted appropriately in blocking solution (100 μL per chamber). 15. Wash in 1 % goat serum, 0.1 % Tween-20 in PBS four times, for 10 min. 16. Incubate with the appropriate secondary antibody diluted in blocking solution, for 1 h at RT (100 μL per chamber). Wrap in aluminum foil as soon as secondary antibody is added. 17. Wash in 1 % Goat Serum, 0.1 % Tween-20 in PBS four times, for 10 min. 18. Remove the chambers from the slide. 19. Place a small amount of Vectashield mounting medium with DAPI over the area where each chamber used to be. 20. Place a clean coverslip over the slide and gently blot off any liquid remaining after the staining protocol. 21. Seal with nail varnish. 22. Store slides at 4 °C in the dark. 23. Image at least 3 h later. 3.6 Immunofluores- cence Staining Protocol iPSC Generation Using a Codon Optimized Minicircle Plasmid
  • 29.
    12 1. Takahashi Ket al (2007) Induction of pluripo- tent stem cells from adult human fibroblasts by defined factors. Cell 131(5):861–872 2. Mitalipov S, Wolf D (2009) Totipotency, pluri- potency and nuclear reprogramming. Adv Biochem Eng Biotechnol 114:185–199 3. Feng B, Ng JH, Heng JC, Ng HH (2009) Molecules that promote or enhance reprogram- ming of somatic cells to induced pluripotent stem cells. Cell Stem Cell 4(4):301–312 4. Maherali N et al (2007) Directly repro- grammed fibroblasts show global epigenetic remodeling and widespread tissue contribu- tion. Cell Stem Cell 1(1):55–70 5. Wernig M et al (2007) In vitro reprogramming of fibroblasts into a pluripotent ES-cell-like state. Nature 448(7151):318–324 6. Sommer CA et al (2010) Excision of reprogram- ming transgenes improves the differentiation potential of iPS cells generated with a single excisable vector. Stem Cells 28(1):64–74 7. Stadtfeld M, Hochedlinger K (2010) Induced pluripotency: history, mechanisms, and appli- cations. Genes Dev 24(20):2239–2263 8. Stadtfeld M, Nagaya M, Utikal J, Weir G, Hochedlinger K (2008) Induced pluripotent stem cells generated without viral integration. Science 322(5903):945–949 4 Notes 1. Do not start off with too much parental plasmid, as an overgrown culture will limit the induction efficiency. 2. To ensure a high yield, make sure that the pellet is completely resuspended in Qiagen Maxi Kit P1 buffer and use the same volume of P2 and S3. For example, when using 400 mL spin- ning tubes, resuspend the pellet in 10 mL P1, P2, and S3 buf- fer instead of 8 mL. 3. To avoid bacterial genomic DNA contamination, avoid exces- sive shaking of the resuspended pellet when adding buffer P2. 4. Solutions should be made fresh for each experiment. Keep at 4ºC until day 2. 5. The permeabilization step is only required for the detection of nuclear protein (e.g., OCT4, SOX2, or NANOG); when sur- face markers are detected, skip the permeabilization step and go straight to the primary antibody incubation. Acknowledgements We thank Nicholas Mordwinkin for proof-reading the manuscript. We thank funding support from the National Institutes of Health (NIH) R01 HL113006, NIH U01 HL099776, NIH R24 HL117756, and the American Heart Association Established Investigator Award (JCW). This work was also supported by DFG (German Research Foundation) (to S.D.). References Sebastian Diecke et al.
  • 30.
    13 9. Warren Let al (2010) Highly efficient repro- gramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA. Cell Stem Cell 7(5): 618–630 10. Zhou H et al (2009) Generation of induced pluripotent stem cells using recombinant pro- teins. Cell Stem Cell 4(5):381–384 11. Okita K et al (2011) A more efficient method to generate integration-free human iPS cells. Nat Methods 8(5):409–412 12. Tiemann U et al (2011) Optimal reprogram- ming factor stoichiometry increases colony numbers and affects molecular characteristics of murine induced pluripotent stem cells. Cytometry A 79(6):426–435 13. Warlich E et al (2011) Lentiviral vector design and imaging approaches to visualize the early stages of cellular reprogramming. Mol Ther 19(4):782–789 14. Jia F et al (2010) A nonviral minicircle vector for deriving human iPS cells. Nat Methods 7(3):197–199 15. Gonzalez F, Boue S, Izpisua Belmonte JC (2011) Methods for making induced pluripo- tent stem cells: reprogramming a la carte. Nat Rev Genet 12(4):231–242 16. Lan F et al (2013) Abnormal calcium handling properties underlie familial hypertrophic car- diomyopathy pathology in patient-specific induced pluripotent stem cells. Cell Stem Cell 12(1):101–113 iPSC Generation Using a Codon Optimized Minicircle Plasmid
  • 31.
    15 Milica Radisic andLauren D. Black III (eds.), Cardiac Tissue Engineering: Methods and Protocols, Methods in Molecular Biology, vol. 1181, DOI 10.1007/978-1-4939-1047-2_2, © Springer Science+Business Media New York 2014 Chapter 2 Scalable Cardiac Differentiation of Human Pluripotent Stem Cells as Microwell-Generated, Size Controlled Three-Dimensional Aggregates Celine L. Bauwens and Mark D. Ungrin Abstract The formation of cells into more physiologically relevant three-dimensional multicellular aggregates is an important technique for the differentiation and manipulation of stem cells and their progeny. As industrial and clinical applications for these cells increase, it will be necessary to execute this procedure in a readily scalable format. We present here a method employing microwells to generate large numbers of human pluripotent stem cell aggregates and control their subsequent differentiation towards a cardiac fate. Key words Cardiac differentiation, Microwells, Human pluripotent stem cells, Size controlled cell aggregates, Forced cell aggregation 1 Introduction It has been known for a long time that cellular behavior in two- dimensional adherent culture does not fully recapitulate three- dimensional in vivo systems, and consequently there is a growing trend in many fields of research to employ three-dimensional tissue constructs. These may be divided into scaffolded constructs, where cells are cultured on or in an exogenous supporting matrix [1], and unscaffolded systems that consist primarily or exclusively of cells (which may then proceed to generate their own endogenous matrices) [2, 3]. Here, we will discuss a process for generating large numbers of uniform, unscaffolded aggregates of human pluripotent stem cells for differentiation towards a cardiac fate. Conventionally, size controlled cellular aggregates have been generated in microcentrifuge tubes [4], as hanging drops [5], by micropatterning human embryonic stem cells (hESCs) colonies [6] or by centrifugation into U- or V-bottom plates [7, 8]; how- ever throughput is limited using these approaches. Microwell- based systems are similar in concept to V-bottom plates; however
  • 32.
    16 the smaller sizeof the microwells permits very large numbers of uniform aggregates to be generated from a single culture-plate well in a mechanically simple system [9]. This approach has been used for a variety of applications including differentiation of plu- ripotent stem cells to ectodermal [10], endodermal [11], mesoder- mal [12], and extraembryonic [13] fates; chondrogenesis from mesenchymal stem cells [14]; and generation of uniform substrates for toxicological screening [15] and investigations of mechanobi- ology [16]. Using this approach to control aggregate size, we consequently determine surface area to volume ratio, which in turn modulates extraembryonic endoderm (ExE) commitment. Cardiogenic sig- nals from this cell lineage then specify subsequent cardiomyogen- esis [12]. 2 Materials All reagent preparation and cell culture handling should be carried out in a biological safety cabinet under sterile conditions. 1. Biological safety cabinet. 2. Pipette aid. 3. Serological pipettes (5–25 mL). 4. Aspirator. 5. Aspirator or Pasteur pipettes. 6. 15 and 50 mL conical tubes. 7. Fume hood. 8. 0.22 μm syringe filter. 9. 5 % CO2, 5 % O2, and humidity controlled cell culture incubator (Subheadings 3.1 and 3.4). 10. 5 % CO2, 20 % O2, and humidity controlled cell culture incubator (Subheading 3.5). 11. Low speed centrifuge with a swinging bucket rotor fitted with a plate holder. 12. P2, P20, P200, and P1000 micropipettors with appropriate tips. 13. Inverted microscope with 4×, 10×, and 20× phase objectives. 14. Ultra-Low Attachment (ULA) 24 well plates. 15. 1.5 mL microcentrifuge tubes. 16. Bench-top microcentrifuge. 1. L-Ascorbic Acid: Prepare a stock solution of 5 mg/mL in 4ºC, sterile ultrapure distilled water. Leave on ice and vortex 2.1 General Equipment and Supplies 2.2 Basal Medium Celine L. Bauwens and Mark D. Ungrin
  • 33.
    17 periodically until completelydissolved. Filter-sterilize using a 0.22 μm syringe filter, aliquot at 1 mL volume, and store the Ascorbic Acid aliquots at −20ºC. Use a freshly thawed aliquot each time medium is prepared. 2. Monothioglycerol: MTG should be aliquoted (1 mL) and stored frozen (−20ºC). When aliquots are thawed, they can be used for 3 months and then discarded. Aliquoting of MTG is strongly recommended as it minimizes the amount of oxida- tion due to repeated opening. On the day of media prepara- tion, dilute 13 μL MTG in 1 mL StemPro-34. Discard unused diluted MTG. 3. Transferrin: Transferrin (stock concentration: 30 mg/mL) should be aliquoted (1 mL aliquots) and stored at −20ºC. 4. All cytokines are stored lyophilized at −20ºC until ready to be aliquoted in the following buffers and at the following concen- trations, Table 1. Continue to store unused aliquots at −20 °C. 5. 4 mM Hydrochloric acid (HCl), 0.1 % BSA Buffer: In a fume hood, transfer 30 μL of 6.0 N HCl solution to 50 mL of ultra- pure distilled water. Filter-sterilize the solution using a 0.22 μm syringe filter. Add 2 mL of 25 % BSA solution to the 50 mL HCl solution. 6. PBS, 0.1 % BSA Buffer: Add 20 μL of 25 % BSA solution for every mL PBS. 7. Supplemented DMEM/F-12: 1 % Penicillin/Streptomycin (Pen/Strep) and 1 % L-glutamine in DMEM/F-12. 8. hESC Wash Medium: 5 % Knockout Serum Replacement in Supplemented DMEM/F12. Table 1 Cytokines and buffers used for media preparation Cytokine Buffer Stock Conc. (μg/mL) Human Bone morphogenetic protein 4 (hBMP-4) 4 mM Hydrochloric acid, 0.1 % BSA 10 Human Fibroblast Growth Factor 2 (basic) (hbFGF) Phosphate buffered saline (PBS), 0.1 % BSA 10 Human Vascular Endothelial Growth Factor (hVEGF) Phosphate buffered saline (PBS), 0.1 % BSA 5 Activin A Phosphate buffered saline (PBS), 0.1 % BSA 10 Human Dickkopf 1 homolog (hDkk-1) Phosphate buffered saline (PBS), 0.1 % BSA 50 Scalable Cardiac Differentiation of Human Pluripotent Stem Cells…
  • 34.
    18 9. StemPro-34: StemPro-34is sold as a kit with two components. The supplement is kept at −20ºC and the liquid media at 4ºC. When combined, the media is stable for 30 days (see Note 1). StemPro-34 is always used with the supplement added in this protocol. 10. Modified StemPro-34: StemPro-34 modified with the addi- tion of 1 % Pen/Strep, 1 % L-glutamine, 0.5 % Transferrin, 1 % Ascorbic Acid, 0.3 % MTG. This medium should be prepared on the day of use with freshly thawed Ascorbic Acid. 1. One 6 well plate culture of feeder-free or feeder-depleted (see Note 2) hESCs adapted to single cell passaging (e.g., enzy- matic dissociation with TrypLE). 2. TrypLE Select. 3. hESC Wash Medium (preparation previously described in Subheading 2.2, item 8). 4. Hemocytometer. 5. Trypan Blue. 6. Aggrewell™ 400 plates (StemCell Technologies 27845). 7. Aggrewell™ Rinsing Solution (StemCell Technologies 07010). 8. Aggregation Medium: Modified StemPro-34, 0.5 ng/mL BMP4, 10 μM Y-27632 ROCK Inhibitor. 1 mL of medium is required per well of a 24 well Aggrewell™ 400 plate. 9. OPTIONAL: 37 μm strainer. 1. Stage 1 Induction Medium: Modified StemPro-34, 10 ng/mL BMP4, 5 ng/mL bFGF, 6 ng/mL Activin A. 1 mL of medium is required per well of a 24 well Aggrewell™ 400 plate (see Note 3). 2. Stage 2 Induction Medium: Modified StemPro-34, 10 ng/mL VEGF, 150 ng/mL DKK1. 1 mL of medium is required per well of a 24 well ULA plate (see Note 3). 3. Stage 3 Induction Medium: Modified StemPro-34, 10 ng/mL VEGF, 150 ng/mL DKK1, 5 ng/mL bFGF. 1 mL of medium is required per well of a 24 well ULA plate (see Note 3). 4. hESC Wash Medium (prepared as previously described in Subheading 2.2, item 8). 1. 1 mg/mL Collagenase Type II in Hank’s Balanced Salt Solution. 2. TrypLE Select. 3. hESC Wash Medium (preparation previously described in Subheading 2.2, item 8). 2.3 hESC Aggregate Formation Components 2.4 Cardiac Induction Components 2.5 Flow Cytometry Celine L. Bauwens and Mark D. Ungrin
  • 35.
    19 4. DNase: DNaseshould be diluted in ultrapure distilled water to a concentration of 1 mg/mL, aliquoted, and stored at −20 °C. 5. 96 well assay plate (or 1.5 mL microcentrifuge tubes). 6. IntraPrep fixation and permeabilization kit. 7. Cardiac Troponin T antibody. 8. Allophycocyanin (APC)-conjugated goat-anti-mouse IgG antibody. 9. HF buffer solution: 2 % Fetal Bovine Serum (FBS) in Hank’s Balanced Salt Solution. 10. 5 mL round-bottom flow cytometry analysis tubes. 11. Flow cytometer with the appropriate laser and filter to excite (i.e., 633–640 nm) and detect (i.e., 660/20 nm) APC. 3 Methods A schematic of the process is shown in Fig. 1. 1. Add 0.5 mL Aggrewell™ Rinsing Solution to each well of the Aggrewell™ plate used. Ensure that the solution contacts the entire surface inside each microwell by centrifuging the plate at 840×g for 2 min. Incubate the Aggrewell™ plate containing the Rinsing Solution for 30–60 min at room temperature. Aspirate the solution and wash twice with 1 mL PBS per well, centrifuging at 840×g for 2 min after each PBS addition. 2. Completely dissociate the hESCs to single cells: Aspirate the cul- ture medium from each culture well, rinse each well with 1 mL TrypLE Select, and aspirate the TrypLE Select. Residual TrypLE should be sufficient to dissociate the cells. Incubate the plate at 37 °C for 3 min, then add 1 mL of hESC Wash Medium to each well and mechanically dissociate the cells from the tissue cul- ture surface by pipetting with a P1000 micropipettor. Transfer the cells (in hESC Wash Medium) to a 15 mL conical tube. 3.1 hESC Aggregate Formation Fig. 1 Schematic of cardiac differentiation protocol. The sequence of steps in Subheadings 3.1–3.6 is diagrammed, along with an indication of the timescale over which they occur Scalable Cardiac Differentiation of Human Pluripotent Stem Cells…
  • 36.
    20 Optionally, if thedissociation appears to be incomplete and cell clumps remain, the suspension may be passed through a strainer at this point. 3. Perform a cell count: Take a 10 μL sample of the cell suspen- sion collected in the previous step. Add 30 μL of Trypan Blue and mix the suspension well by pipetting up and down. Transfer 10 μL of Trypan Blue-stained cells to each chamber of a hemo- cytometer and visualize under the inverted microscope with the 10× objective. Count the cells. 4. From the cell count results, calculate how many wells can be seeded at 1.2×106 cells per well. Resuspend the dissociated hESCs in Aggregation Medium at a density of 1.2×106 cells/mL (see Note 4). 5. Aspirate the PBS wash solution from each well on the Aggrewell™ Plate. Using a P1000 micropipettor, evenly dis- tribute 1 mL of the cell suspension to each well on the Aggrewell™ Plate. If seeding a large numbers of wells, periodi- cally mix the container of cell suspension to prevent settling. Centrifuge the plate at 200×g for 5 min. Visualize the plate under the microscope to confirm cells have spun to the bottom of each microwell (see Note 5). 6. Incubate the plate for 24 h at 37 °C in a 5 % CO2, 5 % O2 (hypoxic) incubator. 1. Prepare the required volume of Stage 1 Induction Medium (for a 24 well plate, volume=1 mL×number of wells). Place the medium in a 37 °C water bath for at least 15 min. 2. Remove the Aggrewell™ plate from the incubator. Inspect the aggregates under the microscope. Compared to immedi- ately post-centrifuge aggregation, they should appear intact with smooth edges (more round and less square than the pre- vious day). 3. To remove the supernatant but retain the aggregates in their individual microwells: Use a P1000 micropipettor. For each well on the Aggrewell™ plate, place the tip of the micropipettor at the surface of the culture medium in the well and against the edge of the well (keep the plate level horizontally). Slowly remove the medium, being careful not to disturb the aggre- gates at the bottom of the microwells. Once the medium level has been reduced to about 1–2 mm from the surface of the textured microwell surface, the plate can be slowly tilted to collect medium at one side of the well (once the volume is this low, fluid motion is greatly reduced and it is easier to avoid aggregates getting lifted out of their individual microwells). Slowly pipette out the remaining medium in the well. 4. To add fresh Stage 1 Induction Medium while ensuring the aggregates remain in their individual microwells: Take up 1 mL 3.2 Cardiac Induction Stage 1 (24±2 h Following Aggregation) Celine L. Bauwens and Mark D. Ungrin
  • 37.
    21 of Stage 1Induction Medium with a P1000 micropipettor. Hold the pipette tip against the inside edge of the well and very slowly dispense the medium against the inside wall of the well. Repeat for the remaining wells. 5. Return the plate to the incubator under hypoxic conditions for 4 days. Stage 2 Induction directs mesoderm differentiation. Therefore at this time-point it is very important to remove all residual Activin A from the Stage 1 Induction Medium. Activin A is a very potent signaling molecule and at this stage of differentiation, even trace levels would promote differentiation to the endoderm lineage at the expense of cardiac induction. Therefore, at this stage the aggre- gates are removed from the Aggrewell™ plates, washed well, and transferred to bulk aggregate cultures in 24 well ULA plates. 1. Place hESC Wash Medium (volume=number of wells×2 mL) in a 37 °C water bath for 10–15 min. 2. Prepare the necessary volume of Stage 2 Induction medium (volume=number of wells×1 mL) and place in a 37 °C water bath for 10–15 min. 3. Using a 5 mL serological pipette, harvest the aggregates from each well of the Aggrewell™ plate, and collect the aggregate suspension in a 15 mL conical tube (up to 10 wells per tube). 4. Allow the aggregates to settle for 15 min in hypoxic (5 % O2) conditions. This step is performed to separate single cells and cellular debris from the intact aggregates. Aspirate the super- natant carefully and resuspend the aggregates in 10 mL hESC Wash Medium to wash out residual inductive cytokines (especially Activin A which is a potent signaling molecule even at very low concentrations). 5. Centrifuge the aggregates at 50×g for 2 min and aspirate the supernatant. 6. Resuspend the pelleted aggregates in the Induction 2 Medium prepared in step 2 above. 7. Dispense the aggregate suspension into a 24 well ULA plate at 1 mL per well. Visualize the aggregates under the microscope. They should appear as uniform, tight cell clusters. 8. Incubate under hypoxic conditions until day 8. Typically, in a successful cardiac induction, spontaneously con- tracting aggregates will be observed under the microscope between day 8 and day 12. 1. Prepare the necessary volume of Stage 3 Induction Medium (volume=number of wells×2 mL) and place in a 37ºC water bath for 10–15 min. 3.3 Cardiac Induction Stage 2 (Day 4: 96±2 h After Aggregate Formation) 3.4 Cardiac Induction Stage 3 (Day 8: 192±2 h After Aggregate Formation) Scalable Cardiac Differentiation of Human Pluripotent Stem Cells…
  • 38.
    22 2. Use a5 mL serological pipette to transfer the aggregates to 15 mL conical tubes, pooling up to 10 mL of aggregates per tube. Allow 10 min for the aggregates to settle. Aspirate the supernatant and resuspend the aggregates in Stage 3 Induction Medium. Using a 5 mL serological pipette, redistribute the aggregates into a 24 well ULA plate at 1 mL per well. 3. Incubate under hypoxic conditions until day 12. 1. Prepare the necessary volume of Stage 3 Induction Medium (volume=number of wells×1 mL) and place in a 37ºC water bath for 10–15 min. 2. Use a 5 mL serological pipette to transfer the aggregates to 15 mL conical tubes, pooling up to 10 mL of aggregates per tube. Allow 10 min for the aggregates to settle. Aspirate the supernatant and resuspend the aggregates in Stage 3 Induction Medium. Using a 5 mL serological pipette, redistribute the aggregates into a 24 well ULA plate at 1 mL per well. 3. Incubate the cells at normoxic oxygen levels for the remainder of the culture period (37ºC, 20 % O2, 5 % CO2). After this time point, cells are no longer cultured under hypoxic conditions. 4. Repeat this complete medium exchange (steps 1–3) at day 16. 5. Cardiac aggregates are ready for harvest on day 20. For flow cytometry analysis of cardiac Troponin T (cTnT) expres- sion frequency, it is recommended that 4 wells of day 20 cardiac aggregates be harvested. 1. Using a 5 mL serological pipette, transfer 1 well of aggregates to a 15 mL conical tube. Centrifuge the aggregates at 50×g for 2 min and carefully aspirate the supernatant. 2. Add 1 mL of freshly dissolved 1 mg/ml collagenase Type II solution to the aggregates, transfer the aggregate suspension to a 1.5 mL microcentrifuge tube, and incubate the aggre- gates in collagenase Type II overnight at room temperature. 3. The next day, using a P1000 micropipettor, gently pipette the aggregates to dissociate them into a homogenous single cell suspension. If the aggregates do not readily dissociate, settle the aggregates, aspirate the supernatant, and incubate the aggregates in 700 μL TrypLE for 1–2 min at room tempera- ture. Gently pipette the aggregates 1–2 times with a P1000 micropipette to dissociate. Add 700 μL hESC Wash Medium containing 14 μL of 1 mg/mL DNAse stock solution to dilute the TrypLE. Take a 10 μL sample for cell counting, and centrifuge the remaining suspension using a bench-top microcentri- fuge for 2 min at 300×g. 4. Stain the 10 μL counting sample with an equal volume of Trypan Blue and count using a hemocytometer. 3.5 Cardiac Induction Stage 3 (Day 12 to Harvest) 3.6 Flow Cytometry Analysis of Cardiac Troponin T Expression Frequency of Aggrewell™ Culture Output Celine L. Bauwens and Mark D. Ungrin
  • 39.
    23 5. Remove the1.5 mL microcentrifuge tube containing the remaining cells from the microcentrifuge. Aspirate the super- natant and resuspend the cells in HF at a concentration of 200,000–500,000 cells per 100 μL. 6. Transfer 100 μL per well to 2 wells of a 96 well plate (see Note 6) per experimental condition or replicate (one well will be for cTnT staining and the other will be the unstained control). 7. Pellet by centrifuging the plate at 300×g for 2 min in a swinging bucket centrifuge. 8. Fix the cells in 200 μL of Intraprep Reagent 1 per well for 15 min at room temperature. 9. Centrifuge the plate at 300×g for 2 min. Carefully aspirate the supernatant and dispose of in a paraformaldehyde waste container. 10. Wash the fixed cells twice: Add 200 μL HF to each well. Centrifuge the plate at 300×g for 2 min, aspirate the superna- tant, and repeat the wash step once more. Fixed cells can be stored for up to 1 week in HF at 4 °C. 11. Permeabilize the cells: Centrifuge the plate at 300×g for 2 min. Add 100 μL of Intraprep Reagent 2 per well and incu- bate the plate for 5 min at room temperature. Centrifuge the plate at 300×g for 2 min and aspirate the supernatant. 12. Prepare a staining solution of anti-cTnT (Neomarkers MS-295) in HF at the optimal concentration for the given lot number (determined by titration: typically ranges from 1:500 to 1:2,000 dilution). 13. Add 100 μL staining solution to one well of cells and 100 μL of plain HF to the other well (negative control). Incubate the cells for 30 min at 4 °C. 14. Centrifuge the cells at 300×g for 2 min. Aspirate the superna- tant and add 200 μL HF per well. Repeat the wash step one more time. 15. Prepare a master mix of the secondary antibody: Transfer a volume of HF that corresponds to 100 μL for every well (con- trol and cTnT-stained) being treated. Add 1 μL of goat anti- mouse-APC secondary antibody per 200 μL of HF (1:200 dilution). 16. Stain the samples: Add 100 μL of staining solution to each well (negative and anti-cTnT-stained wells). Incubate cells for 30 min at 4 °C (keep the plate covered or in the dark after adding fluorescent secondary antibody to avoid photobleaching). 17. Centrifuge the cells at 300×g for 2 min. Aspirate the superna- tant and add 200 μL HF per well. Repeat the wash step one more time. Scalable Cardiac Differentiation of Human Pluripotent Stem Cells…
  • 40.
    24 18. Transfer thesamples to 5 mL round-bottom flow cytometer analysis tubes and analyze on a flow cytometer using the red laser. 4 Notes 1. If 500 mL of complete StemPro-34 medium (with supple- ment) will not be used within 30 days, it is recommended that the basal medium and supplement be aliquoted as follows: prepare ten 50 mL aliquots of the basal medium and store at 4 °C, prepare ten 1.3 mL aliquots of the supplement and store at −20 °C. When preparing complete medium from the ali- quots, completely thaw 1 aliquot of supplement in a 37 °C water bath and then add the supplement to a 50 mL aliquot of basal medium. 2. To prepare feeder-depleted hESCs, passage the cells onto Growth Factor Reduced Matrigel (BD 354230) coated 6 well plates at a split ratio that will produce 1–2×106 cells per well after 1–2 days. Use the same medium that was being used to maintain the hESCs on feeders. 3. Depending on the volume of Induction Medium to be pre- pared, the volume of some of the growth factor stock solutions to be added on a given day may be less than 1 μL. To ensure accurate volumes of growth factor stock solutions are being added to the medium preparation, the stock may first be diluted 1:10 in StemPro-34 basal medium, and then the appro- priate volume of diluted stock may be added to the medium preparation. 4. At this seeding density, aggregates containing 1,000 cells will be formed. This is based on our observations with cardiac induction from the HES-2 cell line maintained on feeders [12]. The ideal seeding density may vary and should be optimized for the specific cell lines and culture conditions being used. 5. If the cells do not appear to form a tight pellet at the bottom of the microwells, try eliminating air bubbles in the microwells by spinning some medium into the wells prior to adding the cells: Add 0.4 mL of medium per well and centrifuge the plate at 200×g for 5 min. Then add the appropriate number of cells to each well in 0.6 mL medium per well. 6. If the total number of samples being prepared is small, staining can also be performed in 1.5 mL microcentrifuge tubes. If microcentrifuge tubes are used, supernatant is removed by aspiration and it is recommended that minimum wash volumes of 0.5 mL and centrifuge settings of 900×g (in a bench-top microcentrifuge) for 3 min be used for the wash steps. Celine L. Bauwens and Mark D. Ungrin
  • 41.
    25 Acknowledgements We thank Dr.Peter Zandstra, in whose laboratory this protocol was developed, and Drs. Mark Gagliardi and Gordon Keller who provided assistance in establishing the initial methods on which this process was based. Protocol development was supported by an Ontario Graduate Scholarship in Science and Technology to C.B. and a grant from the Heart and Stroke Foundation of Ontario to Peter Zandstra. References 1. Wintermantel E, Mayer J, Blum J et al (1996) Tissue engineering scaffolds using superstruc- tures. Biomaterials 17:83–91 2. Lazar A, Peshwa MV, Wu FJ et al (1995) Formation of porcine hepatocyte spheroids for use in a bioartificial liver. Cell Transplant 4: 259–268 3. Sachlos E, Auguste DT (2008) Embryoid body morphology influences diffusive trans- port of inductive biochemicals: a strategy for stem cell differentiation. Biomaterials 29: 4471–4480 4. Johnstone B, Hering TM, Caplan AI et al (1998) In vitro chondrogenesis of bone marrow-derived mesenchymal progenitor cells. Exp Cell Res 238:265–272 5. Steinberg MS (1970) Does differential adhe- sion govern self-assembly processes in histo- genesis? Equilibrium configurations and the emergence of a hierarchy among populations of embryonic cells. J Exp Zool 173:395–433 6. Bauwens CL, Peerani R, Niebruegge S et al (2008) Control of human embryonic stem cell colony and aggregate size heterogeneity influ- ences differentiation trajectories. Stem Cells 26:2300–2310 7. Koike M, Kurosawa H, Amano Y (2005) A round-bottom 96-well polystyrene plate coated with 2-methacryloyloxyethyl phos- phorylcholine as an effective tool for embryoid body formation. Cytotechnology 47:3–10 8. Ng ES, Davis RP, Azzola L et al (2005) Forced aggregation of defined numbers of human embryonic stem cells into embryoid bodies fosters robust, reproducible hematopoietic dif- ferentiation. Blood 106:1601–1603 9. Ungrin MD, Joshi C, Nica A et al (2008) Reproducible, ultra high-throughput forma- tion of multicellular organization from single cell suspension-derived human embryonic stem cell aggregates. PLoS One 3:e1565 10. Kozhich O, Hamilton R, Mallon B (2013) Standardized generation and differentiation of neural precursor cells from human pluripotent stem cells. Stem Cell Rev Rep 9:531–536 11. Ungrin MD, Clarke G, Yin T et al (2012) Rational bioprocess design for human pluripo- tent stem cell expansion and endoderm differ- entiation based on cellular dynamics. Biotechnol Bioeng 109:853–866 12. Bauwens CL, Song H, Thavandiran N et al (2011) Geometric control of cardiomyogenic induction in human pluripotent stem cells. Tissue Eng Part A 17:1901–1909 13. Golos TG, Giakoumopoulos M, Garthwaite MA (2010) Embryonic stem cells as models of trophoblast differentiation: progress, opportu- nities, and limitations. Reproduction 140:3–9 14. Markway B, Tan G-K, Brooke G et al (2010) Enhanced chondrogenic differentiation of human bone marrow-derived mesenchymal stem cells in low oxygen environment micro- pellet cultures. Cell Transplant 19(1):29–42 15. Fey SJ, Wrzesinski K (2012) Determination of drug toxicity using 3D spheroids constructed from an immortal human hepatocyte cell line. Toxicol Sci 127(2):403–411 16. Wallace L, Reichelt J (2013) Using 3D culture to investigate the role of mechanical signaling in keratinocyte stem cells. In: Turksen K (ed) Skin stem cells. Humana Press, Totowa, NJ, pp 153–164 Scalable Cardiac Differentiation of Human Pluripotent Stem Cells…
  • 42.
    27 Milica Radisic andLauren D. Black III (eds.), Cardiac Tissue Engineering: Methods and Protocols, Methods in Molecular Biology, vol. 1181, DOI 10.1007/978-1-4939-1047-2_3, © Springer Science+Business Media New York 2014 Chapter 3 Preparation and Characterization of Circulating Angiogenic Cells for Tissue Engineering Applications Aleksandra Ostojic, Suzanne Crowe, Brian McNeill, Marc Ruel, and Erik J. Suuronen Abstract Circulating angiogenic cells (CACs) are a heterogeneous cell population of bone marrow (BM) origin. These cells are most commonly derived from the peripheral blood, bone marrow, and cord blood, and are one of the leading candidates for promoting vascularization in tissue engineering therapies. CACs can be isolated by culturing peripheral blood mononuclear cells (PBMCs) on fibronectin or by flow cytometry to obtain more specific subpopulations. Here we will describe how to generate a population of CACs, and how to characterize the cells and confirm their phenotype. Also, we will provide select methods that can be used to assess the angiogenic and endothelial cell-like properties of the CACs. Key words Circulating angiogenic cells (CACs), Angiogenic cell therapy, Neovascularization, Peripheral blood mononuclear cells, Tissue engineering 1 Introduction Endothelial progenitor cells (EPCs) were initially isolated from peripheral blood in 1997, and identified by their co-expression of CD34 and vascular endothelial growth factor receptor-2 (VEGFR-2) [1]. EPCs have been further shown to originate from bone marrow (BM) and other tissues, including skeletal muscle and adipose, dermis, and vascular tissues [2–4]. A true definition has not yet been established for these cells, since their phenotype and function can vary depending on their source and/or method of selection. Regardless of origin, it is generally accepted that EPCs play a role in contributing to neovascularization in ischemic tis- sues, directly through participation in the generation of new blood vessels, or indirectly through the production of various pro- angiogenic cytokines [2, 5, 6]. Based on this knowledge, EPCs are actively being tested as cells for therapy (±biomaterials) in multiple diseases such as diabetes,
  • 43.
    28 wound repair, andcardiovascular disease [7]. The potential for revascularization therapy using EPCs and biomaterials has been demonstrated in animal models [8–11] and the feasibility and safety of cell therapy for cardiovascular regeneration has been dem- onstrated in clinical studies, albeit with relatively modest structural and functional benefits [12, 13]. Furthermore, EPCs have been used successfully as a strategy to vascularize engineered tissues [14–16]. Together, this evidence demonstrates the promise that EPCs have for promoting vascularization in tissue engineering and regenerative medicine applications. In 2008, Hirschi et al. assigned the name circulating angio- genic cells (CACs) to a specific type of EPC, due to the fact that they are isolated from the peripheral blood and possess angiogenic properties [17]. The most common method for obtaining CACs from peripheral blood is to isolate the mononuclear cells (MNCs) and culture them on fibronectin-coated plates. After 4–7 days, the non-adherent cells are removed and the remaining adherent cells are considered CACs [18–20]. Starting with ~100 ml of peripheral blood, this isolation procedure can consistently yield 25–60×106 cells, which is plenty for performing multiple experiments using the same donor. Within the population of CACs, CD34+ cells have been identi- fied as one of the most pro-angiogenic subpopulations [21, 22]. CD34+ cells, or other subpopulations of interest, can be isolated from the CACs using fluorescence-activated cell sorting (FACS). This method labels specific cell-surface antigens to identify and sort specific subpopulations of CACs. Common markers used to identify the endothelial precursor/progenitor population are CD34, CD133, and VEGFR-2 [23–26]. The outlined set of experiments describes the procedure for isolating CACs from human peripheral blood, and how to charac- terize the phenotype of the population with flow cytometry. Furthermore, methods are provided to evaluate the angiogenic properties of the cells and to confirm that they are CACs (using DiI-acLDL and FITC-lectin staining). 2 Materials 1. Wash buffer: 1 % FBS, 0.11 % EDTA in Phosphate Buffered Saline (PBS). 2. Histopaque 1077 (Sigma). 3. 12×9 ml Ethylenediaminetetraacetic acid (EDTA) vacutainer tubes. 4. 50 ml conical tubes. 5. Human donors to donate peripheral blood. 2.1 Isolation of Peripheral Blood Mononuclear Cells Aleksandra Ostojic et al.
  • 44.
    29 1. Fibronectin: 20μg/ml fibronectin in PBS. Store at 4 °C. 2. EBM medium (Lonza): 500 ml EBM supplemented with EGM-2 components (Lonza), containing fetal bovine serum (FBS), recombinant long R insulin-like growth factor-1 in aqueous solution (R3 -IGF-1), endothelial growth factor vascu- lar human recombinant (VEGF), gentamicin sulfate ampho- tericin-B, and epidermal growth factor human recombinant in a buffered BSA saline solution (see Note 1). Store at 4 °C. 3. 4×10 cm culture dishes. 1. PBS. Store at 4 °C. 2. EBM medium+supplements (see Subheading 2.2). 1. PBS. Store at 4 °C. 2. EBM medium+supplements (see Subheading 2.2). 3. Antibodies of interest for flow cytometry. 1. Human umbilical vein endothelial cells (HUVECs). 2. HUVEC Medium: M200 medium supplemented with LSGS kit. Store at 4 °C. 3. ECMatrix™ (EMD Millipore; ECM625): includes 10× diluent and matrix solution. 4. 10 cm plates. 5. PBS. Store at 4 °C. 6. CellTracker™ Orange (Invitrogen). 7. 4′,6-diamidino-2-phenylindol (DAPI): 50 μl stock (1 mg/ml) in 950 μl PBS. 8. 96-well plate. 9. EBM medium (see Subheading 2.2). 1. 1,1′-dioctadecyl-3,3,3′,3′-tetramethylindocarbocyanine (DiI)- labeled acetylated low-density lipoprotein (acLDL, Molecular Probes; L-3484): stock at 1 mg/ml. 2. FITC-labeled Ulex-lectin (Sigma; L-9006): stock at 1 mg/ml. 3. PBS. Store at 4 °C. 4. Cytofix buffer (BD Bioscience). 3 Methods Carry out all procedures at room temperature unless otherwise specified. All reagents that are stored at 4 °C should first be warmed to room temperature before use, unless otherwise specified. 2.2 Culture of Peripheral Blood Mononuclear Cells 2.3 Lifting Cells 2.4 CAC Characterization by Fluorescence- Activated Cell Sorting (FACS) 2.5 Angiogenesis Assay 2.6 DiI and Lectin Staining Circulating Angiogenic Cells for Tissue Engineering
  • 45.
    30 For ~100 mlblood (12 vials of EDTA vacutainer tubes): 1. Pour 20 ml Histopaque into 4×50 ml tubes (see Note 2). 2. Collect 12 × 9 ml blood into EDTA vacutainer tubes. Mix gently for a minimum of 5 min on rocker before processing (see Note 3). 3. Using a sterile pipette, slowly and carefully layer blood from three tubes on top of the 20 ml Histopaque (see Note 4). Repeat for each of the 4×50 ml tubes. 4. Centrifuge the tubes for 30 min at 840×g at room tempera- ture (do not use centrifuge brakes—see Note 5). 5. Remove the tubes which now contain the separated blood layers (Fig. 2). Aspirate off top plasma layer to within 1 cm of buffy coat (see Note 6). 3.1 Isolation of Mononuclear Cells from Peripheral Blood (Fig. 1) Blood collection PBMC isolation with Histopaque and differential centrifugation 4 days Fibronectin coated plates + EBM-2 Conduct assays Fig. 1 PBMC isolation and CAC culture protocol plasma Red blood cells Buffy coat Histopaque Fig. 2 Following density gradient centrifugation, the peripheral blood is separated into four layers: (1) red blood cell layer; (2) clear Histopaque layer; (3) “Buffy coat” layer consisting of the mononuclear cells; and (4) plasma layer Aleksandra Ostojic et al.
  • 46.
    31 6. Using asterile transfer pipette, remove the top 1 cm of plasma layer and all of buffy coat and place it in clean, sterile 50 ml tubes (should have four separate tubes). Bring final volume in each tube to 40 ml with wash buffer. 7. Centrifuge the tubes at 300×g for 10 min (do not use centri- fuge brakes). 8. Remove and discard the supernatant. Resuspend the cell pellets in a few ml of wash buffer and combine the cell pellet from each of the four tubes into a single tube. Bring the final volume up to 20 ml using wash buffer. 9. Centrifuge the tubes at 300×g for 10 min (do not use centri- fuge brakes). 10. Resuspend the pellet in 1 ml EBM culture medium. 1. Coat 4×10 cm plate with 3 ml of fibronectin solution (see Note 7). 2. Incubate at room temperature for 30–60 min. 3. Aspirate the excess liquid and air dry for 15 min. 4. Rinse the plates 2× with PBS: leave PBS on for 5 min, aspirate, and repeat. 5. Add 10 ml EBM medium with supplements, warmed to room temperature. 6. Add ¼ of the cells to each of the four plates and culture at 5 % CO2 at 37 °C for 4 days without changing the medium. 7. After 4 days, the adherent population is considered to be CACs (Fig. 3). 3.2 Plating Fibronectin for Selection of CACs Fig. 3 Adherent population of CACs generated from the culture of PBMCs on fibronectin-coated plates for 4 days Circulating Angiogenic Cells for Tissue Engineering
  • 47.
    32 1. Aspirate themedium. 2. Lift cells using gentle pipetting (see Note 8). 3. Centrifuge at 300×g for 5 min (do not use centrifuge brakes). 4. Remove the supernatant and resuspend the CACs in desired volume of EBM medium with supplements, warmed to room temperature. 1. Lift and resuspend the cells as previously described. 2. Count cells with an automated system or a hemocytometer. 3. Place 5 × 105 CACs in a 1.5 ml centrifuge tube in 200 μl of PBS. 4. Stain cells for 30 min at 4 °C with pre-conjugated antibodies. Common antibodies used for characterization are CD34, KDR (VEGFR-2), CD133, CD31, and L-selectin, among others (see Table 1). 5. Wash with PBS and centrifuge at 300×g for 5 min. 6. Aspirate medium and resuspend in 500 μl EBM medium with supplements, warmed to room temperature. 7. Place another 5×105 CACs in a 1.5 ml centrifuge tube in 200 μl of PBS and stain with appropriate IgG isotype-matched control antibodies (see Note 9). Repeat steps 5 and 6. This can be done concurrently with steps 3–6. 8. Analyze and quantify cells with flow cytometry (see Note 10) [25, 27]. 3.3 Lifting Cells 3.4 CAC Characterization by Fluorescence- Activated Cell Sorting (FACS) Table 1 Common markers used to characterize CAC populations. This Table lists some of the markers that have been previously used to characterize the phenotypes of cells in CAC populations. The listed functions are general and not exclusive Marker Function References CD14 Monocyte marker [28, 29] CD31/PECAM-1 Endothelial adhesion molecule [28–30] CD34 Progenitor cell marker [1, 25, 29, 31, 32] CD45 Leukocyte marker [28, 29, 33] CD133 Progenitor cell marker [25, 29, 31, 32] CD144/VE-cadherin Endothelial adhesion molecule [28, 29, 31] CD146 Endothelial adhesion molecule [29, 31, 33] c-Kit Progenitor cell marker [25, 31] DiI-acLDL uptake Endothelial/macrophage detection [28, 31] VEGFR-2 (KDR, Flk-1) Endothelial cell receptor [1, 25, 29, 31] L-selectin Leukocyte adhesion molecule [25, 30] Aleksandra Ostojic et al.
  • 48.
    33 1. HUVECs areprovided from the manufacturer in a frozen state, and stored this way until needed. Thaw the HUVECs at 37 °C in a water bath. 2. Transfer cells to a 15 ml tube with 4 ml pre-warmed HUVEC medium. 3. Centrifuge at 300×g for 5 min. 4. Remove supernatant, and resuspend cells in pre-warmed HUVEC medium. 5. Plate cells in 2× 10 cm dishes, split 50:50, which is equivalent to ~5×105 cells/dish. 6. Incubate at 37 °C and 5 % CO2 until 70–80 % confluent (see Note 11). 7. Change HUVEC medium the following day, and every second day thereafter. 1. This protocol uses the ECMatrix™ angiogenesis kit (EMD Millipore). 2. Thaw the 10× diluent and the gel solution on ice (see Note 12). 3. Mix 100 μl diluent with 900 μl of the gel solution. Aliquot 80 μl of gel mixture into individual wells of a 96-well plate (see Note 13). 4. Allow to set at 37 °C for approximately 1 h. 5. Gels are then ready to use. 1. Remove the supernatant from the HUVEC cultures. 2. Add 1 ml of 0.25 % trypsin and incubate for 5 min at RT with constant agitation (see Note 14). 3. Add 5 ml of EBM medium to the plate to inactivate the tryp- sin. Gently pipette the cells to loosen them from the dish. 4. Rinse the plate with PBS and transfer contents to a 15 ml tube. 5. Centrifuge at 300×g for 5 min and then remove the supernatant. 6. Stain the cells by resuspending them in CellTracker™ Orange (2 μl aliquot of CellTracker™ Orange+998 μl PBS) for 30 min at 37 °C (see Note 15). 7. Add 5 ml of PBS, and then spin the cells as in step 5. 8. Resuspend in desired volume of EBM medium with supple- ments, warmed to room temperature. 1. After completing step 3 of Subheading 3.3, remove the supernatant. 2. Resuspend the CACs in 4′,6-diamidino-2-phenylindole (DAPI) for 30 min at 37 °C. DAPI solution is prepared by mixing 50 μl of stock (1 mg/ml) with 950 μl PBS. 3.5 Angiogenesis Assay 3.5.1 Human Umbilical Vein Endothelial Cell (HUVEC) Culture 3.5.2 Making the In Vitro Angiogenesis Gels 3.5.3 Lifting HUVECs 3.5.4 Performing the Angiogenesis Assay Circulating Angiogenic Cells for Tissue Engineering
  • 49.
    34 3. Add 5ml PBS, and centrifuge at 300×g for 5 min. 4. Remove the supernatant and resuspend CACs in the desired volume of EBM medium with supplements, warmed to room temperature. 5. Count cells using a hemocytometer. 6. Add 10,000 HUVECs and 10,000 CACs together to each well containing ECMatrix™ gel (see Note 16). 7. Fill the wells with 150 μl EBM medium with supplements and incubate at 37 °C overnight (should be 18 h, and no more than a 24 h incubation) (see Note 17). 8. Take photographs of the capillary-like structure formations using fluorescence microscopy. Evaluate the number of CACs (blue DAPI+ cells) that have incorporated into the tubule structures (orange HUVECs), as well as the total length of tubule formation (see Note 18) (Fig. 4). 1. Prepare (DiI)-labeled acetylated low-density lipoprotein (acLDL) stain (stock at 1 mg/ml). Take 2 μl and add to 998 μl PBS. 2. Aspirate medium from CAC cultures and rinse the plates 2× with PBS. 3. Add 1 ml of DiI-acLDL per dish and incubate at 37 °C for 1 h. 4. Aspirate the DiI-acLDL dye and rinse the cells 2× with PBS. 5. Fix the cells with 1.0 ml of Cytofix buffer for 15 min at 37 °C. 3.6 DiI and Lectin Staining Fig. 4 Angiogenesis assay. HUVECs are stained with CellTracker™ Orange (red/ orange) and CACs are stained with DAPI (blue). Arrows indicate CACs that have incorporated into the capillary-like structures Aleksandra Ostojic et al.
  • 50.
    35 6. During thisincubation step, prepare the lectin stain. Dilute FITC-labeled Ulex-lectin to 10 μg/ml by adding 10 μl of stock solution (1 mg/ml) to 990 μl of PBS. 7. Rinse cells 2× with PBS and add 1 ml FITC-labeled Ulex-lectin per dish and incubate at 37 °C for 1 h. 8. Rinse cells 2× with PBS. 9. Add 1 ml of PBS to each dish and image the cells by fluores- cence microscopy (Fig. 5). Dual-staining cells are considered to be CACs (see Note 19). 4 Notes 1. Use a pipette when adding the supplements to minimize vol- ume loss during the transfer. 2. 20 ml of Histopaque is the recommended amount per 30 ml of blood. This amount can be reduced to 10 ml of Histopaque per 30 ml of blood to save on reagents. However, the more Histopaque there is, the easier it is to identify the buffy coat. 3. This step is necessary to mix the blood with the EDTA and prevent clotting. If using the blood right away, mixing can be achieved by inverting the collection tubes multiple times, oth- erwise leave on the rocker. 4. Cannot have layers mix because it will affect the separation efficacy, and the buffy coat layer will not be as visible. 5. It is very important that braking is not applied during centrifu- gation, as this may disrupt proper separation of the layers. Fig. 5 DiI-acLDL (red) and FITC-lectin (green) staining of a CAC culture Circulating Angiogenic Cells for Tissue Engineering
  • 51.
    36 6. Make surenot to aspirate any cells that may be stuck to the side of falcon tube. Gently scrape them off with a pipette before proceeding with the transfer. 7. Cover the plate and gently swirl to spread the fibronectin. Make sure all areas are covered; otherwise cells may not uni- formly adhere. Fibronectin coating step can be performed immediately prior to CAC isolation. This way the plates will be ready right about the same time that the cells are. 8. PBS can be added and cells can be washed multiple times. If absolutely necessary, a scraper can be used to help lift the cells. Do not allow areas of the plate to dry during this step. Add PBS and look under the microscope to ensure that most of the cells have been removed. 9. If flow cytometry analysis cannot be performed immediately, then the cells can be fixed with 4 % PFA for 20 min, washed with PBS, and spun for 5 min at 300×g. Cells can then be left in the fridge for an extended period of time (weeks) before staining (with primary or IgG control antibodies). 10. Use flow cytometry for analysis using a set number of events (cells). A recommended number of events is 100,000–200,000, but more or less can be used depending on the desired analysis. 11. If cells are over-confluent, they may suffer contact inhibition and lose their growth and proliferative potential, and may also become very difficult to trypsinize. Monitor cell density and make sure to split cells before confluence is reached (>80–90 %). 12. The gel components typically take about 2 h to thaw on ice. When ECMatrix™ is prepared, it is imperative to keep it on ice, otherwise solidification will occur. 13. When pipetting the gel into the wells, do not expel all the way in order to avoid bubbles. As an alternative to 96-well plates, Ibidi microslides can be used, which will reduce reagent consumption. 14. Carefully tap the side of the plate to help lift the cells. 15. Depending on the setup of various microscope systems, other cytoplasmic dyes/fluorochromes may be more suitable to visualize the cells, such as CellTracker™ Green (Invitrogen). 16. Must have 1:1 ratio. Adding too little cells will not allow the HUVECs to form tubule structures and adding too many cells may result in overcrowding or tubules forming too quickly and then regressing. 17. Allowing the angiogenesis assay to proceed for longer than 24 h will lead to regression of the tubule structures. As an alternative to EBM medium, the assay can be performed using M200 medium (this is the medium used for HUVEC culture). Aleksandra Ostojic et al.
  • 52.
    37 18. It isrecommended that a minimum of three, and ideally six images of random fields-of-view (FOV) be taken per well. There are three suggested ways to analyze the results using imaging analysis software (such as ImageJ; http://rsbweb.nih. gov/ij/). (A) Count the number of DAPI+ CACs (blue) that have incorporated into tubule structures per FOV (HUVECs, stained with CellTracker™ Orange). (B) Quantify the total length of tubule formation per field-of-view. This is done by measuring the length of each tubule branch and adding them all together. (C) Quantify the total area of tubule formation. This is done by measuring the area of all the tubule formations and adding them together. The results from all 3–6 FOV are then averaged to obtain the final value per well. 19. DiI-acLDL and FITC-lectin staining is a traditional method originally used to identify adherent endothelial progenitor cells (EPCs) cells from the peripheral blood. However, this means of identifying EPCs is not considered sufficient, and additional characterization is necessary [7]. We suggest this staining sim- ply as a means of confirming the successful generation of an adherent CAC population. Acknowledgements This work was supported by the Canadian Institutes of Health Research (to MR and EJS). BM was supported by a University of Ottawa Cardiology Research Endowment Fellowship. References 1. Asahara T, Murohara T, Sullivan A et al (1997) Isolation of putative progenitor endothelial cells for angiogenesis. Science 275:964–967 2. Asahara T, Takahashi T, Masuda H et al (1999) VEGF contributes to postnatal neovasculariza- tion by mobilizing bone marrow-derived endo- thelial progenitor cells. EMBO J 18:3964–3972 3. Grenier G, Scime A, Le Grand F et al (2007) Resident endothelial precursors in muscle, adipose, and dermis contribute to postnatal vasculogenesis. Stem Cells 25:3101–3110 4. Tamaki T, Akatsuka A, Ando K et al (2002) Identification of myogenic-endothelial progen- itor cells in the interstitial spaces of skeletal muscle. J Cell Biol 157:571–577 5. Asahara T, Kawamoto A, Masuda H (2011) Concise review: circulating endothelial pro- genitor cells for vascular medicine. Stem Cells 29:1650–1655 6. Moreno PR, Sanz J, Fuster V (2009) Promoting mechanisms of vascular health: circulating progenitor cells, angiogenesis, and reverse cholesterol transport. J Am Coll Cardiol 53:2315–2323 7. Fadini GP, Losordo D, Dimmeler S (2012) Critical reevaluation of endothelial progenitor cell phenotypes for therapeutic and diagnostic use. Circ Res 110:624–637 8. Gilbert PM, Blau HM (2011) Engineering a stem cell house into a home. Stem Cell Res Ther 2:3 9. Kuraitis D, Giordano C, Ruel M et al (2012) Exploiting extracellular matrix-stem cell interac- tions: a review of natural materials for therapeutic muscle regeneration. Biomaterials 33:428–443 10. Segers VF, Lee RT (2011) Biomaterials to enhance stem cell function in the heart. Circ Res 109:910–922 Circulating Angiogenic Cells for Tissue Engineering
  • 53.
    38 11. Nunes SS,Song H, Chiang CK et al (2011) Stem cell-based cardiac tissue engineering. J Cardiovasc Transl Res 4:592–602 12. Delewi R, Andriessen A, Tijssen JG et al (2013) Impact of intracoronary cell therapy on left ven- tricular function in the setting of acute myocar- dial infarction: a meta-analysis of randomised controlled clinical trials. Heart 99:225–232 13. Jeevanantham V, Butler M, Saad A et al (2012) Adult bone marrow cell therapy improves sur- vival and induces long-term improvement in cardiac parameters: a systematic review and meta-analysis. Circulation 126:551–568 14. Amini AR, Laurencin CT, Nukavarapu SP (2012) Differential analysis of peripheral blood- and bone marrow-derived endothelial progenitor cells for enhanced vascularization in bone tissue engineering. J Orthop Res 30: 1507–1515 15. Critser PJ, Voytik-Harbin SL, Yoder MC (2011) Isolating and defining cells to engineer human blood vessels. Cell Prolif 44(Suppl 1):15–21 16. Zhang Z, Ito WD, Hopfner U et al (2011) The role of single cell derived vascular resident endothelial progenitor cells in the enhance- ment of vascularization in scaffold-based skin regeneration. Biomaterials 32:4109–4117 17. Hirschi KK, Ingram DA, Yoder MC (2008) Assessing identity, phenotype, and fate of endothelial progenitor cells. Arterioscler Thromb Vasc Biol 28:1584–1595 18. Favre J, Terborg N, Horrevoets AJ (2013) The diverse identity of angiogenic monocytes. Eur J Clin Invest 43:100–107 19. Kalka C, Masuda H, Takahashi T et al (2000) Transplantation of ex vivo expanded endothe- lial progenitor cells for therapeutic neovascu- larization. Proc Natl Acad Sci U S A 97: 3422–3427 20. Kuraitis D, Hou C, Zhang Y et al (2011) Ex vivo generation of a highly potent population of circulating angiogenic cells using a collagen matrix. J Mol Cell Cardiol 51:187–197 21. Mackie AR, Losordo DW (2011) CD34- positive stem cells: in the treatment of heart and vascular disease in human beings. Tex Heart Inst J 38:474–485 22. Yang J, Ii M, Kamei N et al (2011) CD34+ cells represent highly functional endothelial progenitor cells in murine bone marrow. PLoS One 6:e20219 23. Garmy-Susini B, Varner JA (2005) Circulating endothelial progenitor cells. Br J Cancer 93: 855–858 24. Peichev M, Naiyer AJ, Pereira D et al (2000) Expression of VEGFR-2 and AC133 by circu- lating human CD34(+) cells identifies a popu- lation of functional endothelial precursors. Blood 95:952–958 25. Sofrenovic T, McEwan K, Crowe S et al (2012) Circulating angiogenic cells can be derived from cryopreserved peripheral blood mono- nuclear cells. PLoS One 7:e48067 26. Werner N, Kosiol S, Schiegl T et al (2005) Circulating endothelial progenitor cells and cardiovascular outcomes. N Engl J Med 353: 999–1007 27. Khan SS, Solomon MA, McCoy JP Jr (2005) Detection of circulating endothelial cells and endothelial progenitor cells by flow cytometry. Cytometry B Clin Cytom 64:1–8 28. Shepherd RM, Capoccia BJ, Devine SM et al (2006) Angiogenic cells can be rapidly mobi- lized and efficiently harvested from the blood following treatment with AMD3100. Blood 108:3662–3667 29. Steinmetz M, Nickenig G, Werner N (2010) Endothelial-regenerating cells: an expanding universe. Hypertension 55:593–599 30. Yip HK, Chang LT, Chang WN et al (2008) Level and value of circulating endothelial pro- genitor cells in patients after acute ischemic stroke. Stroke 39:69–74 31. Rafii S, Lyden D (2003) Therapeutic stem and progenitor cell transplantation for organ vascular- ization and regeneration. Nat Med 9:702–712 32. Werner N, Wassmann S, Ahlers P et al (2007) Endothelial progenitor cells correlate with endothelial function in patients with coronary artery disease. Basic Res Cardiol 102:565–571 33. Delorme B, Basire A, Gentile C et al (2005) Presence of endothelial progenitor cells, dis- tinct from mature endothelial cells, within human CD146+ blood cells. Thromb Haemost 94:1270–1279 Aleksandra Ostojic et al.
  • 54.
    39 Milica Radisic andLauren D. Black III (eds.), Cardiac Tissue Engineering: Methods and Protocols, Methods in Molecular Biology, vol. 1181, DOI 10.1007/978-1-4939-1047-2_4, © Springer Science+Business Media New York 2014 Chapter 4 Isolation and Expansion of C-Kit-Positive Cardiac Progenitor Cells by Magnetic Cell Sorting Kristin M. French and Michael E. Davis Abstract Cell therapy techniques are a promising option for tissue regeneration; especially in cases such as heart failure where transplantation is limited by donor availability. Multiple cell types have been examined for myocardial regeneration, including mesenchymal stem cells (and other bone marrow-derived cells), induced pluripotent stem cells, embryonic stem cells, cardiosphere-derived cells, and cardiac progeni- tor cells (CPCs). CPCs are multipotent and clonogenic, can be harvested from mature tissue, and have the distinct advantages of autologous transplant and lack of tumor formation in a clinical setting. Here we focus on the isolation, expansion, and myocardial differentiation of rat CPCs. Brief adaptations of the protocol for isolation from mouse and human tissue are also provided. Key words Cardiac progenitor cells, C-kit, Cell therapy, MACS, Myocardial infarction 1 Introduction Cardiac progenitor cells (CPCs), expressing the stem cell marker c-kit, were first identified in the adult heart a decade ago and shown to be self-renewing, clonogenic, and multipotent [1]. In addition to c-kit, CPCs express the stem cell markers multiple drug resis- tance (MDR) and Sca-1 as well as cardiac markers Nkx2.5 and Gata-4. They are negative for markers of the hematopoietic lin- eage. CPCs are reported to exist at a density of 1 per 10,000 myo- cytes in the adult rat heart [1] and as high as 1.4 % of total cardiac cells in humans [2]. The safety and efficacy of CPCs for myocardial repair were demonstrated in the SCIPIO Phase I clinical trial [3]. This trial, in addition to various animal studies, suggests that CPCs can improve cardiac function post-injury through reduction in scar size and myocardial preservation/regeneration [4–6]. CPCs may also be a stem cell candidate for generating cardiac patches for tis- sue repair or drug screening [7]. In order to isolate CPCs, a tissue section must be excised from the myocardium (atrial tissue is preferred). The cells are then enzymatically
  • 55.
    Random documents withunrelated content Scribd suggests to you:
  • 56.
    Tapioca must besoaked in twice the quantity of water over night, then add milk and water, and boil till it is soft. Flavor as above. Dropped Egg. Salt some boiling water, and drop in it a raw egg out of the shell, taking care not to break the yolk; take it up as soon as the white is hardened. Dip some toast in hot water, and put salt or butter on to it, and lay the egg on the top. Wheat Gruel for Young Children with weak stomachs, or for Invalids. Tie half a pint of wheat flour in thick cotton, and boil it three or four hours; then dry the lump and grate it when you use it. Prepare a gruel of it by making a thin paste, and pouring it into boiling milk and water, and flavor with salt. This is good for teething children. Another Panada. Boil a mixture of one-fourth wine, and three-fourths water, and flavor it with nutmeg or lemon. Stir in grated bread or crackers, and let it boil up once. Herb Drinks. Balm tea is often much relished by the sick. Sage tea also is good. Balm, sage, and sorrel, mixed with sliced lemon and boiling water poured on, and then sweetened, is a fine drink. Pennyroyal makes a good drink to promote perspiration. Herb drinks must often be renewed, as they grow insipid by standing. Other Simple Drinks. Pour boiling water on to tamarinds, or mashed cranberries, or mashed whortleberries, then pour off the water and sweeten it. Add a little wine if allowed.
  • 57.
    Toast bread verybrown, and put it in cold water, and it is often relished. Pour boiling water on to bread toasted very brown, and boil it a minute, then strain it, and add a little cream and sugar. Make a tea of parched corn pounded, and add sugar and cream. Cream Tartar Whey. Warm a pint of fresh milk, when scalding hot, stir in a teaspoonful of cream tartar, and if this does not turn it, add more, till it does. Strain it, and sweeten with loaf sugar. Those who cannot eat wine whey can eat this without trouble, and it is good in fevers. Simple Wine Whey. Mix equal quantities of water, milk, and white wine. Warm the milk and water, and then add the wine. Sweeten it to the taste. A great Favorite with Invalids. Take one third brisk cider and two thirds water, sweeten it, and crumb in toasted bread, or toasted crackers, and grate on nutmeg. Acid jellies will answer for this, when cider cannot be obtained. A New Way of making Barley Water. Put two tablespoonfuls of pearl barley into a quart jug, two great spoonfuls of white sugar, a small pinch of salt, a small bit of orange, or lemon peel, and a glass of calve’s foot jelly, and then fill the jug with boiling water. Shake it, and then let it stand till quite cold. It is best made over night, to use next day. When the liquor is all poured off, it may be filled again with boiling water, and it is again very good. Panada. Take two crackers, pour on boiling water, and let it simmer five minutes; beat up an egg, sweeten and flavor it to your taste, and then put the cracker to it.
  • 58.
    Arrowroot Blanc Mange. Taketwo tablespoonfuls of arrowroot to one quart of milk, and a pinch of salt. Scald the milk, sweeten it, and then stir in the arrowroot, which must first be wet up with some of the milk. Let it boil up once. Orange water, rose water, or lemon peel, can be used to flavor it. Pour it into moulds to cool. Rice Flour Blanc Mange. Four tablespoonfuls of ground rice and a pinch of salt wet up with a little milk and stirred into a quart of boiling milk. Rub the rind of a lemon with hard, refined sugar, till all the oil is absorbed, and use the sugar to sweeten to your taste. Boil, stirring well, for eight minutes; then cool it, and add the whites of three eggs cut to a froth. Put it on to the fire, and stir constantly till boiling hot, then turn it into moulds, or cups, and let it stand till cold. Another Receipt for American Isinglass Jelly. One ounce of gelatine, or American isinglass. Three pints of boiling water. A pound and a half of loaf sugar. Three lemons, cut in slices, leaving out the peel of one. The whites of four eggs, cut to a stiff froth. Soak the isinglass half an hour in cold water, then take it out and pour on the boiling water. When cool, add the sugar, lemon, and whites of eggs; boil all three or four minutes, then strain through a jelly-bag, and add wine to your taste. Tapioca Jelly. One cup full of tapioca.
  • 59.
    Wash it twoor three times, soak it in water, for five or six hours. Then simmer it in the same water in which it has been soaked, with a pinch of salt and bits of fresh lemon peel, until it becomes transparent. Then add lemon juice, wine, and loaf sugar to flavor it. Let all simmer well together, then pour into glasses to cool. Caudle. To rice, or water gruel, add a wine-glass of wine, or ale, and season with nutmeg and sugar. Sago Jelly. Soak a tea-cup full of sago in cold water, half an hour, then pour off the water, and add fresh, and soak it another half hour; and then boil it slowly with a pinch of salt, a stick of cinnamon, or a bit of orange, or lemon peel, stirring constantly. When thickened, add wine and white sugar to suit the taste, and let it boil a minute; then turn it into cups. Spiced Chocolate. One quart of milk. Two squares of chocolate. One stick of cinnamon. A little nutmeg. Grate the chocolate. Boil the milk, reserving a little cold to moisten the chocolate, which must be mixed perfectly smooth to a thin paste. When the milk boils (in which the cinnamon must be put when cold, and boil in it), stir in the chocolate, and let it boil up quickly, then pour into a pitcher, and grate on the nutmeg. Rich cream added to the milk, will improve it. Barley Water.
  • 60.
    Put two ouncesof pearl barley to half a pint of boiling water, and let it simmer five minutes; pour off the water, and add two quarts of boiling water, add two ounces sliced figs, two of stoned raisins, and boil till it is reduced to a quart. Strain it for drink. Water Gruel. To two quarts of boiling water, add one gill of Indian meal and a heaped tablespoonful of flour, made into a paste and stirred in the water. Let it boil slowly twenty minutes. Salt, sugar, and nutmeg to the taste. Oatmeal makes a fine gruel in the same way. Beef Tea. Broil a pound of tender, juicy beef ten minutes, salt and pepper it, cut it in small pieces, pour on a pint of boiling water, steep it half an hour, and then pour it off to drink. Another way is slower, but better. Cut the beef in small pieces, fill a junk bottle with them, and keep it five hours in boiling water. Then pour out, and season the juice thus obtained. Tomato Syrup. Express the juice of ripe tomatoes, and put a pound of sugar to each quart of the juice, put it in bottles, and set it aside. In a few weeks it will have the appearance and flavor of pure wine of the best kind, and mixed with water is a delightful beverage for the sick. No alcohol is needed to preserve it. The medical properties of the tomato are in high repute, and it is supposed that this syrup retains all that is contained in the fruit. Arrowroot Custard for Invalids. One tablespoonful of arrowroot. One pint of milk. One egg.
  • 61.
    One tablespoonful ofsugar. Mix the arrowroot with a little of the cold milk, put the milk into a sauce-pan over the fire, and when it boils, stir in the arrowroot and the egg and sugar, well beaten together. Let it scald, and pour into cups to cool. A little cinnamon boiled in the milk flavors it pleasantly. Sago for Invalids. Wash one large spoonful of sago, boil it in a little water, with a pinch of salt and one or two sticks of cinnamon, until it looks clear; then add a pint of milk, boil all well together, and sweeten with loaf sugar. Rice Jelly. Make a thin paste of two ounces of rice flour, and three ounces of loaf sugar, and boil them in a quart of water till transparent. Flavor with rose, orange, or cinnamon water. It can be made also by boiling whole rice long and slowly. A pinch of salt improves it. Sassafras Jelly. Take the pith of sassafras boughs, break it in small pieces, and let it soak in cold water till the water becomes glutinous. It has the flavor of sassafras, and is much relished by the sick, and is also good nourishment. Buttermilk Whey. One quart of good buttermilk. When boiling, beat up the yolk of an egg, and stir in, and, if it can be allowed, some thick cream, or a little butter. Then beat the white to a stiff froth and stir in. Sugar and spice if liked. Alum Whey. Mix half an ounce of pounded alum with one pint of milk. Strain it, and add sugar and nutmeg to the whey. It is good in cases of hemorrhages, and sometimes for colic.
  • 62.
    Another Wine Whey. Onepint of boiling milk. Two wine-glasses of wine. Boil them one moment, stirring. Take out the curd, and sweeten and flavor the whey. Mulled Wine. One pint of wine and one pint of water. Beat eight eggs and add to the above, while boiling, stirring rapidly. As soon as it begins to boil it is done. Tamarind Whey. Mix an ounce of tamarind pulp with a pint of milk, strain it, and add a little white sugar to the whey. Egg Tea and Egg Coffee (very fine). Beat the yolk of an egg with a great spoonful of sugar, and put it to a tea-cup of cold tea or cold coffee. Add a half a tea-cup of water, cold in summer and boiling in winter, and as much cream. Then whip the white of the egg to a stiff froth and stir it in. It is very much relished by invalids. Cranberry Tea. Wash ripe cranberries, mash them, pour boiling water on them, and then strain off the water and sweeten it, and grate on nutmeg. Apple Tea. Take good pippins, slice them thin, pour on boiling water, and let it stand some time. Pour off the water, and sweeten and flavor it.
  • 63.
    Egg and Milk. Beatthe yolk of an egg into a great spoonful of white sugar, or more. Add a coffee cup of good milk, then beat the white of the egg to a stiff froth, and stir it in. A little wine, or nutmeg to flavor it. Sago Milk. Soak one ounce of sago in a pint of cold water an hour. Pour off the water, and add a pint and a half of new milk. Simmer it slowly till the sago and milk are well mixed. Flavor with sugar, nutmeg, and wine. Tapioca Milk. Made like sago milk, only not boiled so long. Bread and Milk. Take a slice of good bread and soak it in milk, and then put on a little butter, and it is often very acceptable to the sick. In some cases sprinkle a little salt on instead of butter. Egg Gruel. Beat the yolk of an egg with a spoonful of white sugar, and then beat the white separately, to a stiff froth. Pour water when boiling to the yolk, then stir in the white and add spice, or any seasoning, to suit the taste. When a person has taken a violent cold, after being warm in bed give this as hot as it can be taken, and it is often a perfect cure. Ground Rice Gruel. Take two tablespoonfuls of ground rice, and a pinch of salt, and mix it with milk enough for a thin batter. Stir it with a pint of boiling water, or boiling milk, and flavor with sugar and spice. Oatmeal Gruel.
  • 64.
    Four tablespoonfuls ofgrits (coarse oatmeal) and a pinch of salt, into a pint of boiling water. Strain and flavor it while warm. Or, take fine oatmeal and make a thin batter with a little cold water, and pour it into a sauce-pan of boiling water. Simple Barley Water. Take two ounces and a half of pearl barley, cleanse it, and boil it ten minutes in half a pint of water. Strain out this water and add two quarts of boiling water, and boil it down to one quart. Then strain it, and flavor it with slices of lemon and sugar, or sugar and nutmeg. This is very acceptable to the sick in fevers. Compound Barley Water. Take two pints of simple barley water, a pint of hot water, two and a half ounces of sliced figs, half an ounce of liquorice root sliced and bruised, and two ounces and a half of raisins. Boil all down to two pints, and strain it. This is slightly aperient. Cream Tartar Beverage. Take two even teaspoonfuls of cream tartar, and pour on a pint of boiling water, and flavor it with white sugar and lemon peel to suit the taste. If this is too acid, add more boiling water, as cold, or lukewarm water, is not so good. Seidlitz Powders. Two drachms of Rochelle salts, and two scruples of bicarbonate of soda, in a white paper; thirty-five grains of tartaric acid in a blue one. Dissolve that in the white paper in nearly half a tumbler of water, then add the other powder, dissolved in another half tumbler of water.
  • 65.
    Syrup mixed withthe water makes it more agreeable. It is a gentle laxative. Blackberry Syrup, for Cholera and Summer Complaint. Two quarts of blackberry juice. One pound of loaf sugar. Half an ounce of nutmegs. A quarter of an ounce of cloves. Half an ounce of cinnamon. Half an ounce of allspice. Pulverize the spice, and boil all for fifteen or twenty minutes. When cold, add a pint of brandy. Remarks on the Combinations of Cooking. The preceding receipts have been tested by the best housekeepers. In reviewing them, it will be seen that there are several ways of combining the various articles, all of which have, in the hands of good housekeepers, proved successful. Still it will be found that some methods are more successful than others. In most cases, the receipts have been written as given by the ladies, who endorse them as the best. But it is believed that the following general rules will enable a housekeeper to modify some of them to advantage. In using the whites of eggs, it is found, as shown by several receipts, that various combinations are much lighter when they are cut to a froth, and put in the last thing. This is so in batter puddings, and several other receipts. It seems, therefore, probable that in all cases, cake and pies, and puddings that will allow it, will be lighter by adding the cut whites of the eggs the last minute before cooking.
  • 66.
    Sponge cake especiallywould most probably be most easily made light by this method. In using alkalies with acids to raise mixtures, the poorest is pearlash, the next best is saleratus; bicarbonate of soda is still better, and sal volatile is best of all. But one thing must be remembered in reference to sal volatile, and that is, that the lightness made by it is owing to the disengagement of the gas by heat. It is mixed with the flour, and when set in the oven, the heat volatilizes and expels the gas, and thus the lightness is induced. Of course hot water must not be used to dissolve it, as it would expel much of the gas. Sal volatile must be kept powdered, and closely confined in glass bottles with ground glass stoppers. It is certain to make any mixture light that can be raised by anything. Cream tartar is best bought in lumps, and then pulverized and kept corked. When saleratus is used with sour milk or buttermilk, the flour should be wet up with the sour milk, and then the alkali dissolved, and worked in. This makes the effervescence take place in the mixture; whereas, if the alkali is put into the sour liquid, much of the carbonic acid generated is lost before it reaches the flour. In all cases, then, where saleratus is used with acid wetting, it would seem best to wet up the flour with at least a part of the sour liquid, before putting in the alkali. When the alkali is a light powder, it may sometimes be mixed thoroughly with the flour, and then the sour liquid be mixed in. The experiment can be made by any who like to learn the result. A lady who understands chemistry may often improve her receipts by applying chemical principles. All the lightness made by an acid and an alkali is owing to the disengagement of carbonic acid, which is retained by the gluten of the flour. Of course, then, that mode is best which secures most effectually all the carbonic acid generated by the combination.
  • 67.
    Cooking is oftenmuch improved by a judicious use of sugar or molasses. Thus, in soups, a very little sugar, say half a teaspoonful to the quart, gives body to the soup, and just about as much sweetness as is found in the juices of the best and sweetest kinds of meat. It is very good when the meats used are of inferior kind, and destitute of sweetness. So in preparing vegetables that are destitute of sweetness, a little sugar is a great improvement. Mashed turnips, squash, and pumpkin, are all of them much improved by extracting all the water, and adding a little sugar, especially so when they are poor. A little molasses always improves all bread or cakes made of unbolted wheat or rye. A little lard or butter always improves cakes made of Indian meal, as it makes them light and tender. The careful use of salt is very important in cooking. Everything is better to have the salt cooked in it, but there should always be a little less salt than most would like, as it is easy for those who wish more to add it, but none can subtract it. When the shortening is butter, no salt is needed in cakes and puddings, but in all combinations that have no salt in shortening, it must be added. A little salt in sponge cake, custards, and the articles used for desserts, made of gelatine, rice, sago, and tapioca, is a great improvement, giving both body and flavor.
  • 68.
    CHAPTER XXI. ON MAKINGBUTTER AND CHEESE. The directions in this article were given by a practical amateur cheese-maker of Goshen, Conn., a place distinguished all over the nation for the finest butter and cheese. Articles used in Making Cheese. The articles used in making cheese are, a large tub, painted inside and outside, to hold the milk, a large brass kettle to heat it, a cheese basket, cheese hooks, cheese ladders, strainers of loose linen cloth, and a cheese press. It is indispensable that all the articles used be first washed thoroughly, then scalded, and then dried thoroughly, before putting away. Mode of Preparing the Rennet. Do not remove any part of the curd that may be found in the rennet (which is the stomach of a calf), as it is the best part. Take out everything mixed with the curd in the stomach. Soak the rennet in a quart of water, then hang it to dry, where flies will not reach it, and keep the water bottled for use. Rennet differs in strength, so that no precise rule can be given for quantity, but say about half a tea-cup full to two pails of milk. To Make Cheese. Strain the milk into the tub, keeping in all the cream. Heat a portion, and then add it to the cold, till the whole is raised to 98° or 100° Fahrenheit; no more and no less. Then put in the rennet, stirring well, and take enough to have the curd form well in an hour. If it does not form well, more must be stirred in.
  • 69.
    When the curdis formed, cut it in small checks to the bottom, and then break it gently with a skimmer, to make the whey separate. If this is not done gently, the milk runs off, the whey turns white, and the cheese is injured. The greener the whey, the better the cheese. When the whey is separated, set the basket over the tub, spread the strainer over, and dip the curd into it, occasionally lifting the corners to hasten the draining. Then draw up the corners, twist them, and set a stone on, to press out the whey for ten minutes. Then again cut up the curd, and press it again as before. Continue thus till it is thoroughly drained. Then press it all into some regular form, and in cool weather it may stand till next morning for more cheese to be added. But if the weather is hot, it must be scalded the first day. To Scald the Curd. Cut, or chop the curd into cakes the fourth of an inch in size, put it in the strainer, and immerse it in the brass kettle of warm water, enough to cover it. Then raise the temperature to 105°. Stir it well till warmed through, say half an hour. Then gradually add cold water, till reduced to 88° or 90°. Then drain the curd thoroughly as before, and salt it, allowing four ounces of salt to every ten pounds of curd, and mixing very thoroughly. Then put it into the small strainers, and then into the cheese hoops, laying the strainer over smoothly, and placing the follower on it. Put it to press, and let it remain two days. When taken from the press, grease it with common butter or butter made of whey cream, and set it on a shelf in a dark, cool room. Grease and turn it every day till firm, and for six months grease and turn it often. If the cheese is to be colored, boil anatto with ley, and put to the milk with the rennet. To make sage cheese, put in sage juice (some add to the sage spinach juice) when you put in the rennet. Sew strips of strong cotton around large cheeses, when taken from the
  • 70.
    press, to preservetheir form. In fly time, put cayenne pepper in the butter you rub on. Stilton cheese is made of milk enriched by cream, and of a small size. Cottage cheese is made by pressing the curds of milk, and when free from whey, adding cream or butter and salt. Welsh rabbit is made by melting cheese and adding wine and other seasoning. Old and dry cheese is very good grated and mixed with a little brandy, just enough to wet it up. Directions for making Butter. Two particulars are indispensable to success in making good butter; the first is, that the churning be frequent, so that the cream will not grow bitter, or sour; and the second is cleanliness in all the implements and processes connected with it. In hot weather it is important to keep the milk, cream, and butter as cool as possible. For this purpose, those who have no ice-house, or very cool milk-room, hang their cream down a well. In winter it is needful to raise the temperature of the cream a little, while churning, but care must be taken to do it very slightly, or the butter is injured. The best way is to warm the churn, and churn in a warm room. After the weather gets cold, the cream rises more perfectly after allowing the milk to stand say ten or eleven hours, to set it over a furnace a while till it is warmed through, but not heated hot enough to boil. Then take it back, let it stand eleven hours longer, and skim it. This secures more, and better cream. In hot weather, set the churn into a tub of cold water, and churn steadily, as stopping puts back the process, and injures the butter. In hot weather, do not churn very fast, as it makes soft butter. When
  • 71.
    the butter hascome, collect it in a wooden bowl, which is the best article to work it in, having first scalded it, and then put it in cold water till cold. Do not use the hand in working over butter, as it injures it so much that a higher price is often paid for butter made without using the hand. A wooden spad made for the purpose is the proper article for working over butter. As soon as the butter is put in the bowl, pour in as much of the coldest water as will allow you to work the butter, and keep adding and pouring off cold water, as you work it, until the water will run off clear. It is continuing this process until all the buttermilk is extracted, which alone will secure butter that will keep good and sweet. Water hastens the process, but butter keeps longer not to have it used. No precise direction can be given for salting, as tastes vary so much in regard to this. It is a good way to notice the proportions which are most agreeable, and note the measure, and then measure ever after. In salting down in firkins, use the nicest rock salt, as much depends on the nature of the salt. The firkins must be very thoroughly seasoned, and the bottom covered with salt, and the sides rubbed with it. Pack the butter in layers, with salt between. After a few days, the butter will shrink from the sides, and then the space must be filled with new and nice brine. Muslin spread under and over the layer of salt, between the layers of butter is a good plan, as it saves the butter. It is said that butter will be preserved sweet a long time for journeys, or voyages, by working into it very thoroughly a mixture composed of one-fourth salt, one-fourth saltpetre, and two-fourths white sugar. In large dairies, the milk is churned soon after it is taken from the cow.
  • 72.
    The quality ofbutter depends very much upon the kind of cows. Those who give a great deal of milk, are usually small and thin. Every cow should have a tea-cup full of salt each week, and must be well fed. Green cornstalks and carrots, are excellent for cows. Turnips, cabbage, and parsnips spoil the milk. The waste of the kitchen, with a quart of corn meal, and as much hay as she will eat daily, is good fare. Skimmed milk for drink is good, and if it is refused, withhold water, and the cow will learn to love it. Milk three times a day, and you get much more cream; stripping must be done thoroughly, or you lose cream, and dry up the cow. Never make a cow run, as it injures the milk. Use tin vessels for milk. A stoneware churn is best, and a tin one is better than wood. Keep milk in a cool place, where air circulates freely; close air spoils milk and cream. Never stir milk after it is set, as it stops the rising of the cream. Skim milk as soon as it becomes loppard. Put a little ice in each pan in hot weather, and you get more cream. In skimming milk, do not scrape off the hardest portion that adheres to the pan, as it injures the butter. Put a spoonful of salt to each pailful of milk (except what is for family use), and it makes the butter sweeter, and come easier. Salt your cream as you gather it, and it keeps better, and makes sweeter butter. In hot weather churn in the coolest part of the day, and in a cool place, and do not shut the air out of the churn, as it is necessary to make the butter come. Butter is best, to work it enough the first time. Never work it three times. It will keep better to work out the buttermilk without putting in water. The more entirely it is freed from buttermilk, the longer it will keep sweet. A good brine is made for butter by dissolving a quart of fine salt, a pound of loaf sugar, and a teaspoonful of saltpetre in two quarts of water, and then strain it on to the butter. Packed butter is most perfectly preserved sweet by setting the firkin into a larger firkin, and filling in with good brine, and covering it. Butter will keep sweet a year thus. Buttermilk kept in potter’s ware dissolves the glazing, and becomes poisonous.
  • 73.
    Never scald strainersor milky vessels till thoroughly washed, as the milk or cream put in them will be injured by it. The best way to scald such vessels is to plunge them all over into scalding water, and then every spot is scalded. Butter will sometimes not come because the air is too much excluded from the churn.
  • 74.
    CHAPTER XXII. ARTICLES ANDCONVENIENCES FOR THE SICK. “In some maladies,” says Dr. Pereira, “as fevers and acute inflammatory diseases, an almost unlimited use of fluids is admitted, under the names of slops, thin diet, fever diet, broth diet, &c. They quench thirst, lessen the stimulating quality of the blood, increase its fluidity, and promote the actions of the secreting organs. They are sometimes useful, also, in lessening the irritating contents of the alimentary canal.” “But in some maladies it is necessary to restrict the quantity of fluids taken, or, in other words, to employ a dry diet.” As it is so customary for invalids to throng to watering places, the following remarks contain very important cautions. “The Congress Water at Saratoga,” says Dr. Lee, “though it possesses active medical qualities, yet, except in diseases attended with inflammatory action, seldom occasions unpleasant consequences, unless drank in very large quantities, when it often causes serious, if not dangerous effects.” Dr. Steel, a physician who has devoted much attention to this subject, remarks, “About three pints should be taken, an hour or two before breakfast, and be followed by exercise, to produce a cathartic effect. Where more is needed for this effect, add a teaspoonful or two of Epsom salts to the first tumbler. It should not be drank at all during the remainder of the day by those who wish to experience the full benefit of its use. It would be better for those whose complaints render them fit subjects for its administration, if the fountain should be locked up, and no one suffered to approach it after the hour of nine or ten in the morning.” It is probable that multitudes who frequent mineral springs, not only lose all benefit, but suffer injury by the excessive use of the water.
  • 75.
    Such waters should,by invalids, be taken under the direction of a physician well acquainted with their nature and uses. Alcoholic drinks should never be given to the sick, except by direction of a physician, as they are powerful medical agents, and in some cases would increase disease. The acid drinks are ordinarily those most relished by the sick, and they are, usually, very serviceable, especially in febrile and other inflammatory attacks. Ice cream and drinks are good for the sick, especially in fevers. When a person is debilitated by sickness, the stomach should never be loaded with rich food. Nor should the palate be tempted by favorite articles, when no appetite for food exists, as this is the indication of nature that the stomach is in no order to digest food. Dr. Lee remarks, “We regard rice as the most valuable of all the articles of food, in cases of the derangement of the digestive organs. It nourishes, while it soothes the irritable mucous membrane, and while it supports strength, never seems to aggravate the existing disease. For acute, or chronic affections of the alimentary canal, rice water for drink, and rice jelly for food, seem peculiarly well adapted, and appear to exert a specific influence in bringing about a recovery. These preparations are invaluable also in convalescence from acute fevers and other maladies, and in the summer complaints of young children.” “Isinglass is a very pure form of gelatine, and dissolved in milk, sweetened and flavored, is taken with advantage by convalescents when recovering from debility.” Cod sounds, and the American gelatine, are equally good. Calve’s foot jelly, blanc mange, and other gelatinous food, are among the best kinds of nourishment, especially in cases of cholera infantum. The slight quantity of spice or wine used to flavor such articles, except in peculiar cases, cannot do any injury.
  • 76.
    Buttered toast, eitherdry or dipped, rarely is a suitable article for the sick, as melted oils are very difficult of digestion. Where there are strong powers of digestion, it may be proper. Many cases of illness, among both adults and children, are readily cured by abstinence from all food. Headaches, disordered stomachs, and many other attacks, are caused, often, by violating the rules of health laid down in the preceding chapter, and in consequence, some part of the system is overloaded, or some of the organs are clogged. Omitting one, two, or three meals, as the case may be, gives the system a chance to rest, and thus to gain strength, and allows the clogged organs to dispose of their burdens. The practice of giving drugs to “clear out the stomach,” though it may afford the needed relief, always weakens the system, while abstinence secures the good result, and yet does no injury. Said a young gentleman to a distinguished medical practitioner of Philadelphia,—“Doctor, what do you do for yourself, when you have a turn of headache, or other slight attack?” “Go without my dinner,” was the reply. “Well, if that will not do, what do you do then?” “Go without my supper,” was the answer. “But if that does not cure you, what then?” “Go without my breakfast. We physicians seldom take medicines ourselves, or use them in our families, for we know that starving is better, but we cannot make our patients believe it.” Many cases of slight indisposition are cured by a change of diet. Thus, if a person suffers from constipation, and, as the consequence, has headaches, slight attacks of fever, or dyspepsia, the cause often may be removed by eating rye mush and molasses for breakfast, brown bread, baked apples, and other fruits. In cases of diarrhœa, rice water for drink, and rice pudding or jelly, will often remove the evil. In cases of long-continued confinement from sickness, it is very desirable to have a good variety of articles for the sick, as the invalid
  • 77.
    is wearied withthe same round, and perhaps may be suffering for some ingredient of food, which is not found in the articles provided. For this reason, a large number of receipts of articles for the sick have been provided in this work. In preparing them, great care should be used to have every article employed clean and pure, and to prevent any burning in cooking, as the sensibilities of the sick to bad tastes and smells are very acute. It is often the case in dyspepsia and cholera infantum, that jellies of American gelatine are very much better than any preparations of farinaceous food, being much more easily digested. It would be a happy thing for the sick, and a most benevolent custom, if the young ladies of a place should practise cooking the various articles for the sick, and carrying them to invalids as an offering of kindness and sympathy. It would be twice a blessing, first to the invalid, and quite as much to the young benefactress. There are many little comforts and alleviations for the sick, which should be carefully attended to, which are particularly pointed out in the chapter on the Care of the Sick, in the Domestic Economy. Such, for example, as keeping a room neat, clean, and in perfect order, having every article in use sweet and clean, keeping a good supply of cool water, providing pleasant perfumes, lemons, flowers, and other objects agreeable to the senses, speaking softly, kindly, and cheerfully, and reading the Bible and other cheering books of the kind, whenever it will be acceptable. Be careful to change the linen next the skin, and the bed linen, often. Be sure to ventilate the room thoroughly, two or three times a day, as pure air is a great restorative of health and strength. Wash the skin often, as it has a great tendency to restore health, and never, except in very peculiar cases, can do any harm. Always request a physician to write all his directions, that no mistake may be made, and nothing be forgotten. Always inquire of him as to the exact mode of preparing every article ordered, and never
  • 78.
    venture to alter,or omit, what he directs, unless you are sure that you are better qualified to practise than he, in which case he should be dismissed, and you should assume his duties. Always keep all medicines in papers and vials, labelled, that poisons be not given by mistake, or other injurious articles used. The drawing at Fig. 5 represents a contrivance for the sick, which ought to be prepared in every village, to rent out to those who need it. Fig. 5. It is called the Water Bed, or Hydrostatic Couch, and is made at an expense of from twelve to fifteen dollars. The object of it is to relieve the sufferings of those who, from extreme emaciation, or from ulcers, or bed sores, are great sufferers from the pressure of the bed on these sore places. This kind of bed proves a great relief from this kind of suffering. It consists of a wooden box, six feet long, and two feet and a half wide at the top, and the sides gradually sloping inward, making it fourteen inches deep. This is lined with sheet zinc, to make it water tight. Over this is thrown, and fastened to the edge of the box, a sheet of thick India rubber, water-proof cloth, large enough for an entire lining to the inside of the box. The edges of it are first made to adhere to the upper edge of the box with spirit varnish, and then a thin strip of board is nailed on, to fasten it firmly, and make it water tight. Near the bottom, at A, is a hole and plug, to let off water; and at B, a tin tube, soldered in the upper part of the outside,
  • 79.
    to pour waterin. When used, the box is to be filled half full of water, about blood warmth. Then a woollen blanket and pillow are laid upon the India rubber cloth, and the patient laid on them, and he will float as he would in water, and there will be no pressure on any part of the body greater than is felt when the body is in water. This is important for all who suffer from bed sores, or sloughing in protracted fevers, from diseases in the hip-joint, from diseases of the spine, lingering consumption, and all diseases that compel to a protracted recumbent position. None but those who have seen, or experienced the relief and comfort secured to sufferers by this bed, can conceive of its value. The writer saw the case of a young man, who was enduring indescribable tortures with the most dreadful ulcers all over his body, and who had for several days and nights been unable to sleep, from extreme suffering. This bed was made for him, as an experiment, after trying every other mode of relief in vain. It was placed by his bedside, and the water poured in, and then his friends raised him with the greatest care in a blanket, and laid him on it. Instantly his groans ceased, an expression of relief and delight stole over his countenance, and exhausted nature sunk instantly into the most peaceful and protracted slumbers. And ever after, he was relieved from his former sufferings. Every hospital, every alms-house, and every village should have the means of obtaining such a bed for the many classes of sufferers who would thus find relief, and it is woman who should interest herself to secure such a comfort for the sick, who especially are commended to her benevolent ministries.
  • 80.
    The drawing, AA,is the outline of a sick couch, such as would prove a great comfort in every family, and at a small expense. The following are the dimensions:—Length of seat inside, six feet three inches; breadth, twenty-eight inches; height of the seat from the floor, thirteen inches. The swing frame, dd, is three feet long, and is fastened three feet from the head-board by a very large pin or screw. The seat should be of sacking, and a thick hair mattrass, or cushion for a bed or cushion and be divided into two parts, where the swing frame is fastened. The frame is fastened by large screws at ff. The supporter, ee, is fastened by large and strong hinges to the upper part of the frame, and is moved into the notches made in the frame of the seat. When in the notch a, the frame is very low; when in notch b, it is higher; and when at c, it is as high as is needful to raise the sick. A piece of sacking is to be fastened over the frame, leaving it loose, especially at the top, and leaving a space at o, so as to give room for a pillow, and so that the head can be thrown a little back. The frame and supporter must be thick and strong. When not in use for the sick, the frame can be laid down, and the cushion laid over it, and then with a frill fastened in front, it makes a good-looking and most comfortable chamber couch, or a lolling sofa for a sitting-room. Such a couch saves much labor to friends and nurses, because it is so low, so easily moved, and the nurse can go around it and work on both sides so easily, while the frame raises the patient with great ease and comfort. It would be a good plan to engage some carpenter or cabinet-maker, in every village, to provide such an article to rent out, and probably it would be in constant demand. The frame, supporter, hinges, and screws, must be very strong, or they will break. Fig. 6 represents a contrivance for securing exercise in the open air for invalids, which would often prove convenient and agreeable. Such an article can be easily made of the broken toy of a child, called a velocipede, or the back wheels of a child’s wagon. Nothing
  • 81.
    but shafts areneeded, and a common rocking-chair, with a foot- board nailed across the front rocker, on which to rest the feet. The chair is then slipped along back to the axle of the wheels, so that the shafts, when raised, are under the seat, and lift it up. In this way an invalid can be rolled through yards and gardens with very little effort, and with great comfort and relief to the imprisoned sufferer, who perhaps can ride in no other way. Fig. 6. There is no way of relieving the weariness and nervousness of an invalid more effectually than by rubbing the limbs and arms with the bare hand of a healthful person. Those who believe in animal magnetism would say, that by this method, the well person imparts a portion of the magnetism of a healthful body to aid in restoring the sick. Those who do not believe in it, will say that it soothes and strengthens the nerves. Either way, it is a great comfort to a suffering invalid. It is unhealthful to sleep with a sick person, especially one who has lung complaints, as the breath and effluvia from the skin sometimes communicate disease, even in complaints not contagious. Young
  • 82.
    children should notsleep with the aged, because their healthful fluids will be absorbed.
  • 83.
    CHAPTER XXIII. THE PROVIDINGAND CARE OF FAMILY STORES. The art of keeping a good table, consists, not in loading on a variety at each meal, but rather in securing a successive variety, a table neatly and tastefully set, and everything that is on it, cooked in the best manner. There are some families who provide an abundance of the most expensive and choice articles, and spare no expenses in any respect, who yet have everything cooked in such a miserable way, and a table set in so slovenly a manner, that a person accustomed to a really good table, can scarcely taste a morsel with any enjoyment. On the contrary, there are many tables where the closest economy is practised, and yet the table-cloth is so white and smooth, the dishes, silver, glass, and other table articles so bright, and arranged with such propriety, the bread so white, light, and sweet, the butter so beautiful, and every other article of food so well cooked, and so neatly and tastefully served, that everything seems good, and pleases both the eye and the palate. A habit of doing everything in the best manner, is of unspeakable importance to a housekeeper, and every woman ought to aim at it, however great the difficulties she may have to meet. If a young housekeeper commences with a determination to try to do everything in the best manner, and perseveres in the effort, meeting all obstacles with patient cheerfulness, not only the moral, but the intellectual tone of her mind is elevated by the attempt. Although she may meet many insuperable difficulties, and may never reach the standard at which she aims, the simple effort, persevered in, will have an elevating influence on her character, while at the same time she actually will reach a point of excellence far ahead of those who,
  • 84.
    discouraged by manyobstacles, give up in despair, and resolve to make no more efforts, and let things go as they will. The grand distinction between a noble and an ignoble mind is, that one will control circumstances; the other yields, and allows circumstances to control her. It should be borne in mind, that the constitution of man demands a variety of food, and that it is just as cheap to keep on hand a good variety of materials in the store-closet, so as to make a frequent change, as it is to buy one or two articles at once, and live on them exclusively, till every person is tired of them, and then buy two or three more of another kind. It is too frequently the case, that families fall into a very limited round of articles, and continue the same course from one year to another, when there is a much greater variety within reach, of articles which are just as cheap and easily obtained, and yet remain unthought of and untouched. A thrifty and generous provider, will see that her store-closet is furnished with such a variety of articles, that successive changes can be made, and for a good length of time. To aid in this, a slight sketch of a well-provided store-closet will be given, with a description of the manner in which each article should be stored and kept, in order to avoid waste and injury. To this will be added, modes of securing a successive variety, within the reach of all in moderate circumstances. It is best to have a store-closet open from a kitchen, because the kitchen fire keeps the atmosphere dry, and this prevents the articles stored from moulding, and other injury from dampness. Yet it must not be kept warm, as there are many articles which are injured by warmth. A cool and dry place is indispensable for a store-room, and a small window over the door, and another opening out-doors, is a great advantage, by securing coolness, and a circulation of fresh air.
  • 85.
    Flour should bekept in a barrel, with a flour scoop to dip it, a sieve to sift it, and a pan to hold the sifted flour, either in the barrel, or close at hand. The barrel should have a tight cover to keep out mice and vermin. It is best, when it can be conveniently done, to find, by trial, a lot of first-rate flour, and then buy a year’s supply. But this should not be done, unless there are accommodations for keeping it dry and cool, and protecting it from vermin. Unbolted flour should be stored in barrels, and always be kept on hand, as regularly as fine flour. Indian meal should be purchased in small quantities, say fifteen or twenty pounds at a time, and be kept in a covered tub or keg. When new and sweet, it should not be scalded, but when not perfectly fresh and good when used, it is improved by scalding. It must be kept very cool and dry, and if occasionally stirred, is preserved more surely from growing sour or musty. Rye should be bought in small quantities, say forty or fifty pounds at a time, and be kept in a keg, or half barrel with a cover. Buckwheat, Rice, Hominy, and Ground Rice, must be purchased in small quantities, and kept in covered kegs, or tubs. Several of these articles are infested with small black insects, and examination must occasionally be made for them. Arrowroot, Tapioca, Sago, Pearl Barley, American Isinglass, Macaroni, Vermacelli, and Oatmeal, are all articles which help to make an agreeable variety, and it is just as cheap to buy a small quantity of each, as it is to buy a larger quantity of two or three articles. Eight or ten pounds of each of these articles of food can be stored in covered jars, or covered wood boxes, and then they are always at hand to help make a variety. All of them are very healthful food, and help to form many delightful dishes for desserts. Some of the most healthful puddings are those made of rice, tapioca, sago, and macaroni, while isinglass, or American gelatine, form elegant articles for desserts, and is also excellent for the sick.
  • 86.
    Sugars should notbe bought by the barrel, as the brown is apt to turn to molasses, and run out on to the floor. It is best to keep four qualities of sugar on hand. Refined loaf for tea, crushed sugar for the nicest preserves and to use with fruit, nice brown sugar for coffee, and common brown for cooking and more common use. The loaf can be stored in the papers, on a shelf. The others should be kept in close covered kegs, or covered wooden articles made for the purpose. Butter must be kept in the dryest and coldest place you can find, in vessels of either stone, earthen, or wood, and never in tin. Lard and Drippings must be kept in a dry, cold place, and should not be salted. Usually the cellar is the best place for them. Earthen, or stone jars are the best to store them in. Salt must be kept in the dryest place that can be found. Rock salt is the best for table salt. It should be washed, dried, pounded, sifted, and stored in a glass jar, and covered close. It is common to find it growing damp in the salt stands for the table. It should then be set by the fire to dry, and afterwards be reduced to fine powder again. Nothing is more disagreeable than coarse or damp salt on a table. Vinegar is best made of wine, or cider. Buy a keg, or half barrel of it, and set it in the cellar, and then keep a supply for the castors in a junk bottle in the kitchen. If too strong, it eats the pickles. Pickles never must be kept in glazed ware, as the vinegar forms a poisonous compound with the glazing. Oil must be kept in the cellar. Winter strained must be got in cold weather, as the summer strained will not burn except in warm weather. The best of lard oil is preferred to every other by those who use it. Some lard oil is very poor. Molasses, if bought by the barrel, or half barrel, should be kept in the cellar. Sugar bakers’ is best for the table, and Porto Rico for cooking. If bought in small quantities, it should be kept in a
  • 87.
    demijohn. No vesselshould be corked or bunged, if filled with molasses, as it will swell, and burst the vessel, or run over. Hard Soap should be bought by large quantity, and laid to harden on a shelf, in a very dry place. It is much more economical to buy hard, than soft soap, as those who use soft soap are very apt to waste it in using it, as they cannot do with hard soap. Starch it is best to buy by a large quantity. It comes very nicely put up in papers, a pound or two in each paper, and packed in a box. Starch, which by the single pound is five cents a pound, if bought by the box, is only three cents a pound, and this makes a good deal of difference, in a large family, by the year. The high-priced starch is cheapest in the end. Indigo is not always good. When a good lot is found by trial, it is best to get enough for a year or two, and store it in a tight tin box. Coffee it is best to buy by the bag, as it improves by keeping. Let it hang in the bag, in a dry place, and it loses its rank smell and taste. Tea, if bought by the box, is about five cents a pound cheaper than by small quantities. If well put up in boxes lined with lead, it keeps perfectly. But put up in paper, it soon loses its flavor. It therefore should, if in small quantities, be put in glass, or tin, and shut tight. Saleratus should be bought in small quantities, then powdered, sifted, and kept tight corked in a large mouth glass bottle. It grows damp if exposed to the air, and then cannot be used properly. Raisins should not be bought in large quantities, as they are injured by time. It is best to buy the small boxes. Currants for cake should be prepared as directed for cake, and set by for use in a jar.
  • 88.
    Lemon and OrangePeel should be dried, pounded, and set up in corked glass jars. Nutmeg, Cinnamon, Cloves, Mace, and Allspice, should be pounded fine, and corked tight in small glass bottles with mouths large enough for a junk bottle cork, and then put in a tight tin box, made for the purpose. Or they can be put in small tin boxes with tight covers. Essences are as good as spices. Sweet Herbs should be dried, and the stalks thrown away, and the rest be kept in corked large mouth bottles, or small tin boxes. Cream Tartar, Citric and Tartaric Acids, Bicarbonate of Soda, and Essences, should be kept in corked glass jars. Sal volatile must be kept in a large-mouth bottle, with a ground glass stopper to make it air-tight. Use cold water in dissolving it. It must be powdered. Preserves and Jellies should be kept in glass or stone, in a cool, dry place, well sealed, or tied with bladder covers. If properly made, and thus put up, they never will ferment. If it is difficult to find a cool, dry place, pack the jars in a box, and fill the interstices with sand, very thoroughly dried. It is best to put jellies in tumblers, or small glass jars, so as to open only a small quantity at a time. The most easy way of keeping Hams perfectly is to wrap and tie them in paper, and pack them in boxes or barrels with ashes. The ashes must fill all interstices, but must not touch the hams, as it absorbs the fat. It is much less labor, and quite as certain a mode as the one previously mentioned. It keeps them sweet, and protects from all kinds of insects. After smoked beef, or ham, are cut, hang them in a coarse linen bag in the cellar, and tie it up to keep out flies. Keep Cheese in a cool, dry place, and after it is cut, wrap it in a linen cloth, and keep it in a tight tin box. Keep Bread in a tin covered box, and it will keep fresh and good longer than if left exposed to the air.
  • 89.
    Welcome to ourwebsite – the perfect destination for book lovers and knowledge seekers. We believe that every book holds a new world, offering opportunities for learning, discovery, and personal growth. That’s why we are dedicated to bringing you a diverse collection of books, ranging from classic literature and specialized publications to self-development guides and children's books. More than just a book-buying platform, we strive to be a bridge connecting you with timeless cultural and intellectual values. With an elegant, user-friendly interface and a smart search system, you can quickly find the books that best suit your interests. Additionally, our special promotions and home delivery services help you save time and fully enjoy the joy of reading. Join us on a journey of knowledge exploration, passion nurturing, and personal growth every day! ebookbell.com