SlideShare a Scribd company logo
1 of 14
Download to read offline
HAL Id: hal-01774301
https://hal.archives-ouvertes.fr/hal-01774301
Submitted on 13 Nov 2018
HAL is a multi-disciplinary open access
archive for the deposit and dissemination of sci-
entific research documents, whether they are pub-
lished or not. The documents may come from
teaching and research institutions in France or
abroad, or from public or private research centers.
Lā€™archive ouverte pluridisciplinaire HAL, est
destinƩe au dƩpƓt et Ơ la diffusion de documents
scientifiques de niveau recherche, publiƩs ou non,
Ć©manant des Ć©tablissements dā€™enseignement et de
recherche franƧais ou Ʃtrangers, des laboratoires
publics ou privƩs.
An overview of treatment strategies for
Hutchinson-Gilford Progeria Syndrome
Karim Harhouri, Diane Frankel, Catherine Bartoli, Patrice Roll, Annachiara
de Sandre-Giovannoli, Nicolas LĆ©vy
To cite this version:
Karim Harhouri, Diane Frankel, Catherine Bartoli, Patrice Roll, Annachiara de Sandre-Giovannoli,
et al.. An overview of treatment strategies for Hutchinson-Gilford Progeria Syndrome. Nucleus,
European Nuclear Society, 2018, 9 (1), pp.265-276. ąø€10.1080/19491034.2018.1460045ąø€. ąø€hal-01774301ąø€
Full Terms & Conditions of access and use can be found at
http://www.tandfonline.com/action/journalInformation?journalCode=kncl20
Nucleus
ISSN: 1949-1034 (Print) 1949-1042 (Online) Journal homepage: http://www.tandfonline.com/loi/kncl20
An overview of treatment strategies for
Hutchinson-Gilford Progeria syndrome
Karim Harhouri, Diane Frankel, Catherine Bartoli, Patrice Roll, Annachiara
De Sandre-Giovannoli & Nicolas LĆ©vy
To cite this article: Karim Harhouri, Diane Frankel, Catherine Bartoli, Patrice Roll, Annachiara
De Sandre-Giovannoli & Nicolas LĆ©vy (2018) An overview of treatment strategies for Hutchinson-
Gilford Progeria syndrome, Nucleus, 9:1, 265-276, DOI: 10.1080/19491034.2018.1460045
To link to this article: https://doi.org/10.1080/19491034.2018.1460045
Ā© 2018 The Author(s). Published by Informa
UK Limited, trading as Taylor & Francis
Group
Accepted author version posted online: 05
Apr 2018.
Published online: 05 Apr 2018.
Submit your article to this journal
Article views: 1423
View Crossmark data
Citing articles: 3 View citing articles
An overview of treatment strategies for Hutchinson-Gilford Progeria syndrome
Karim Harhouri a
, Diane Frankel a,b
, Catherine Bartolia
, Patrice Roll a,b
,
Annachiara De Sandre-Giovannoli a,c
and Nicolas L
evy a,c
a
Aix Marseille Univ, INSERM, MMG ā€“ U1251, Marseille, France; b
APHM, H^
opital la Timone, Service de Biologie Cellulaire, Marseille, France;
c
APHM, H^
opital la Timone, D
epartement de G
en
etique M
edicale, Marseille, France
ARTICLE HISTORY
Received 16 January 2018
Revised 21 March 2018
Accepted 27 March 2018
ABSTRACT
Hutchinson-Gilford progeria syndrome (HGPS) is a sporadic, autosomal dominant disorder
characterized by premature and accelerated aging symptoms leading to death at the mean age of
14.6 years usually due to cardiovascular complications. HGPS is caused by a de novo point mutation
in the LMNA gene encoding the intermediate ļ¬lament proteins lamins A and C which are structural
components of the nuclear lamina. This mutation leads to the production of a truncated toxic form
of lamin A, issued from aberrant splicing and called progerin. Progerin accumulates in HGPS cellsā€™
nuclei and is a hallmark of the disease. Small amounts of progerin are also produced during normal
aging. HGPS cells and animal preclinical models have provided insights into the molecular and
cellular pathways that underlie the disease and have also highlighted possible mechanisms
involved in normal aging. This review reports recent medical advances and treatment approaches
for patients affected with HGPS.
KEYWORDS
HGPS; Progerin; FTI; ZOPRA;
AON; Rapamycin; Metformin;
MG132
Introduction
Hutchinson-Gilford progeria syndrome (HGPS; OMIM
#176670) is a rare genetic disorder which affects 1 in
4ā€“8 million children with symptoms resembling physio-
logical aging that include growth impairment, very thin
skin, loss of subcutaneous fat, alopecia, osteoporosis
and heart disease leading to shortened life span and
death at about 14.6 years [1,2]. HGPS was ļ¬rst
described at the end of the XIX century by Jonathan
Hutchinson and Hastings Gilford [3,4]. It was only
more than 100 years later, in 2003, that the heterozy-
gous, de novo mutation c.1824CT, p.G608G
(NM_170707.3) responsible for this accelerated-aging
disease was found to be located within exon 11 of the
LMNA gene that encodes lamins A and C [5,6]. Lamin
A is a nuclear protein belonging to type V intermediate
ļ¬laments. It is synthetized as a precursor called prela-
min A. Prelamin A undergoes a multistep post-transla-
tional processing, including cysteine farnesylation by
farnesyl transferase (FTase) on its C-terminal CaaX
motif, then cleavage of the remaining 3 amino acids by
the metallopeptidase ZMPSTE24. The C-terminal cyste-
ine is then carboxymethylated by the isoprenylcysteine
carboxylmethyltransferase (ICMT). Finally, the last 15
amino acids are cleaved again by ZMPSTE24 to pro-
duce the unfarnesylated, mature lamin A. HGPS muta-
tion activates a cryptic donor splice site in exon 11 of
the LMNA gene that leads to deletion of 50 amino acids
near the C terminus, abrogating the second ZMPSTE24
cleavage site and resulting in accumulation of a trun-
cated and permanently farnesylated prelamin A called
progerin that incorporates abnormally into the nuclear
lamina and exerts multiple toxic effects [7]. At the cellu-
lar level, HGPS is characterized by dramatic defects in
nuclear envelope structure and function [8,9]. Primary
ļ¬broblasts from HGPS patients exhibit reduced prolifer-
ation as well as premature senescence [10], impaired
DNA repair mechanisms [2,11ā€“13], increased reactive
oxygen species production [14], mitochondrial dysfunc-
tion [15], loss of peripheral heterochromatin [16ā€“18]
and telomere attrition [18]. Through these alterations,
progerin accumulation results in loss of peripheral het-
erochromatin caused by the decrease in the repressive
histone marks H3K9me3, H3K27me3 and H3K27
methyltransferase EZH2, increase in H4K20me3
[9,16,19], reduced levels of heterochromatin protein 1
CONTACT Nicolas L
evy Nicolas.Levy@univ-amu.fr INSERM, U 1251, Marseille Medical Genetics, Facult
e de M
edecine de la Timone, 27 Boulevard Jean
Moulin, Marseille Cedex 05, 13385, France.
Ā© 2018 The Author(s). Published by Informa UK Limited, trading as Taylor  Francis Group
This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial License ( http://creativecommons.org/licenses/by-nc/4.0/), which per-
mits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
NUCLEUS
2018, VOL. 9, NO. 1, 265ā€“276
https://doi.org/10.1080/19491034.2018.1460045
REVIEW
(HP1) [20] and Lap2a [9]. Progerin also impairs the
formation of DNA repair foci due to recruitment deļ¬-
ciency of the DNA double-strand break (DSB) repair
factors p53-binding protein 1 (53BP1), Rad50 and
Rad51 at DNA damage sites [21,22]. On the other
hand, altered signaling pathways have been described in
HGPS cells [23]. Among them, altered extracellular
matrix synthesis caused by disturbed Wnt/b-catenin
signaling [24], affected Notch signaling [20], hyperacti-
vation of NF-Kb in response to inļ¬‚ammation [25] and
impaired NRF2 (Nuclear factor erythroid-2-Related Fac-
tor 2) transcriptional activity resulting in increased
chronic oxidative stress [26].
A wide spectrum of treatment strategies, targeting
several processes with different speciļ¬cities, has been
proposed to correct the defects in HGPS: (i) to directly
ā€œrepairā€ the disease -causing mutation; (ii) to inhibit
pre-mRNA aberrant splicing leading to progerin
mRNA production; (iii) to decrease the toxicity of iso-
prenylated and methylated progerin; (iv) to induce
progerin clearance; (v) to decrease the noxious down-
stream effects linked to progerin accumulation [27]
(Figure 1). These approaches will be presented below
and grouped following their individual action.
Prelamin A Isoprenylation and methylation
inhibitors: Lonafarnib, Zoledronate/Pravastatin,
Monoaminopyrimidines and isoprenylcysteine
carboxyl methyltransferase inhibitor
The aberrant splice event that gives rise to progerin
leads to the deletion of the ZMPSTE24 cleavage site
normally used to remove the farnesylated carboxy ter-
minus from prelamin A during posttranslational proc-
essing. Consequently, permanently farnesylated progerin
remains anchored to the inner nuclear membrane
resulting in dominant-negative disruption of the nuclear
scaffold upon progerin dimerization with wild-type
lamins [28]. Knowledge of these steps predicted that
blocking farnesylation using farnesyltransferase inhibitor
(FTI) drugs would decrease progerin production and
toxicity. FTI are small molecules which reversibly bind
to the farnesyltransferase CAAX binding site [29].
Blocking farnesylation of progerin with FTIs restored
normal nuclear architecture and resulted in signiļ¬cant
reductions in nuclear blebbing both in transiently trans-
fected HeLa, HEK 293, NIH 3T3 cells and human
HGPS ļ¬broblasts [8,30ā€“33]. In transgenic HGPS murine
models treated with FTIs, bone mineralization, and
weight are improved, lifespan is extended [34,35] and
cardiovascular defects are prevented [36]. In 2007, the
above studies led to the initiation of a prospective sin-
gle-arm clinical trial (ClinicalTrials.gov, NCT00425607),
using an FTI called lonafarnib, which was originally
developed for the treatment of cancer. A cohort of 25
HGPS patients between 3 and 16 years of age were
included in this trial and received lonafarnib for a mini-
mum of 2 years. In 2012, researchers reported that
some children with HGPS receiving lonafarnib showed
a modest improvement in weight gain: nine patientsā€™
rates of weight gain increased by 50%, six patientsā€™
rates of weight gain decreased by 50%, and 10
remained stable with respect to the rate of weight gain.
Other results from this trial showed improvements in
vascular stiffness including decreases in arterial pulse
wave velocity by a median of 35% in 18 subjects, in
addition to increases in skeletal rigidity (median percent
increases by 40ā€“50% in axial rigidity, 170ā€“228% in ļ¬‚ex-
ural rigidity, and 167ā€“229% in torsional rigidity) as well
as sensorineural hearing and bone mineral density
(3% increase at one or more sites in 76% of children
compared with 40% of the participants who exhibited
decreases at one or more sites) [37]. Although FTI treat-
ment increased mean survival by 1.6 years [38] it was
reported that progerin may become alternatively preny-
lated by geranylgeranyltransferase I when farnesyltrans-
ferases are inhibited; indeed, the simultaneous presence
of both farnesyltransferase inhibitor (FTI-277) and Ger-
anylGeranylTransferase I inhibitor (GGTI-2147) led to a
substantial amount of prelamin A accumulation com-
pared with the effect of each inhibitor alone [39],
thereby explaining the limited beneļ¬cial effects of FTI
monotherapy. Accordingly, authors hypothesized that
blocking both protein farnesylation and geranylgeranyla-
tion could minimize the possibility of alternative preny-
lation events that confer resistance to FTIs. Acting on
the synthetic pathway of farnesyl pyrophosphate, a co-
substrate of farnesyltransferase and a precursor of gera-
nylgeranyl pyrophosphate, we and our close collaborator
Carlos Lopez-Otin (Spain) demonstrated the synergistic
effect of a combination of ZOledronate (N-BisPhospho-
nate) and PRAvastatin (statin) (ZOPRA) and their effec-
tiveness to reduce prenylation and rescue HGPS cells
defects and the progeroid phenotypes of Zmpste24Ā”/Ā”
mice, including improvement of growth retardation, loss
of weight, lipodystrophy, hair loss and bone defects.
Likewise, the longevity of these mice was substantially
extended [39]. This preclinical work allowed our clinical
266 K. HARHOURI ET AL.
team to design and conduct a phase II, monocentric,
open-label, single arm clinical trial (ClinicalTrials.gov,
NCT00731016) using ZOPRA to evaluate its safety and
efļ¬cacy in 12 HGPS patients included for 3.5 years
each, with partially positive results including improve-
ment of weight gain and bone density defects, without
severe adverse effects (De Sandre Giovannoli, Sigaudy
et al, submitted). Although trials using FTIs (Lonafar-
nib) and ZOPRA showed some efļ¬cacy for given
parameters, these drugs could not be considered as a
cure, and further research was needed to implement
more effective therapeutic approaches for patients. A
single-arm triple therapy trial (ClinicalTrials.gov,
NCT00879034) was designed to administer pravastatin,
zoledronate and lonafarnib and sought to further
improve disease parameters by additionally inhibiting
progerin prenylation, given that each enzyme functions
along the protein prenylation pathway. The results of
this trial on 37 participants with HGPS which began in
2009 in Boston revealed additional bone mineral density
beneļ¬t but no major beneļ¬t beyond that seen with lona-
farnib alone [40].
However, caution should be taken when consider-
ing the long-term treatment effects of FTIs, as FTIs
have been shown to cause lethal cardiomyopathy in
mouse models of progeria because of non-farnesylated
prelamin A accumulation [41]. On the other hand,
among 21.608 small molecules tested in induced plu-
ripotent stem cell (iPSC) lines derived from HGPS
patients, Nissan and colleagues identiļ¬ed several com-
pounds, called monoaminopyrimidines (mono-APs),
as new modulators of farnesylation, resulting, in vitro,
Figure 1. Speciļ¬city of a wide spectrum of treatment strategies for progeria. Several therapeutic strategies for progeria are shown. In
decreasing order for their target speciļ¬city, treatment strategies can be mentioned as follows: the mutation-causing disease repair by
genome editing (CRISPR as a future possibility to be evaluated), the inhibition of pre-mRNA aberrant splicing (Antisense OligoNucleoti-
des: AONs, Metformin, MG132), the reduction of isoprenylated and methylated progerin levels (Farnesyl transferase inhibitors: FTIs,
ZOledronate-PRAvastatin: ZOPRA, Monoaminopyrimidines: Mono-APs, Isoprenylcysteine CarboxyMethyl Transferase (ICMT) inhibitor,
GeranylGeranylTransferase Inhibitor: GGTI-2147), the induction of progerin clearance (Rapamycin, Sulforaphane, Retinoids, MG132) and
the reduction of the noxious downstream effects due to progerin accumulation (Rock inhibitor: Y-27632, Nuclear factor erythroid-2-
Related Factor 2: NRF2 reactivation, Methylene blue, sodium salicylate, DOT1L inhibitor: epz-4777, Remodelin, Vitamin D, Resveratrol,
N-Acetyl-Cysteine: NAC, inorganic pyrophosphate: PPi and JH4).
NUCLEUS 267
in improved phenotypes associated with HGPS [42].
This class of protein farnesylation inhibitors, targeting
both farnesyl pyrophosphate synthase and farnesyl
transferase, acts like a combination of FTIs and
bisphosphonates.
In addition to isoprenylation, another consequence
of defective posttranslational modiļ¬cation to prelamin
A in progeria was reported implicating the carboxy
methylation that is mediated by isoprenylcysteine car-
boxyl methyltransferase (ICMT). In this context, Ibra-
him and colleagues revealed that, in vitro treatment
with ICMT inhibitor, increased proliferation and
delayed senescence in human HGPS ļ¬broblasts. Fur-
thermore, ICMT inhibition by lentiviral short hairpin
RNAs (shICMT) increased body weight, normalized
grip strength, as well as prevented bone fractures and
death in Zmpste24-deļ¬cient mice [43].
Autophagy-activating drugs: Rapamycin,
Sulforaphane, Retinoids and MG132
Rapamycin, an immunosuppressive agent that is used
to prevent the rejection of transplanted organs, pro-
moted progerin clearance through autophagy,
improved the abnormal nuclear shape, delayed the
onset of cellular senescence of HGPS ļ¬broblasts
[44,45] and rescued the chromatin phenotype of cul-
tured ļ¬broblasts, including histone methylation status
and BAF and LAP2alpha distribution patterns [46].
Otherwise, studies on LmnaĀ”/Ā”
mouse model for
dilated cardiomyopathy and muscular dystrophy,
showed that pharmacologic reversal of elevated
mTORC1 signaling by rapamycin improves cardiac
and skeletal muscle function and enhances survival
[47]. Using the same mouse model, Liao et al. showed
that rapamycin treatment results in lifespan extension
associated with increased body weight and fat content
[48]. Although these results show beneļ¬cial effects on
lmna KO mice, this model does not mimic the genet-
ics and pathophysiology of HGPS in patients. A phase
I/II monocentric trial (NCT02579044) of Everolimus,
an agent derived from rapamycin, in combination
with Lonafarnib, is currently conducted at the Clinical
and Translational Study Unit (CTSU) of Boston
Childrenā€™s Hospital. Rapamycin inhibits the activity of
mTOR (Mammalian Target Of Rapamycin), known
to regulate a large panel of cellular functions including
protein synthesis, cell growth, cytoskeleton rearrange-
ments, transcription, immune responses or autophagy.
Therefore, much caution should be used while per-
forming the direct translation of the in vitro results to
children affected with progeria. Furthermore, since
rapamycin is also known to inhibit adipogenesis
[49,50], precautions should be taken when using this
compound in HGPS patients who are affected with
generalized lipoatrophy and lipodystrophy.
Sulforaphane, an antioxidant derived from crucifer-
ous vegetables, has been also described to enhance pro-
gerin clearance by autophagy and to reverse the cellular
hallmarks of HGPS in vitro [51]. Intermittent treatment
with Sulforaphane and Lonafarnib separately rescued
the HGPS cellular phenotype [52]. Furthermore, since
the LMNA promoter contains a retinoic acid responsive
element, two recent studies suggested that either reti-
noids alone [53] or in a combination with rapamycin
[54] reduce the amount of progerin and reverse aging
defects in HGPS patient skin ļ¬broblasts. These drugs
may be beneļ¬cial in progeria treatment, but in vivo
extensive studies should be performed before translating
them into clinical trials.
We recently showed that progerin is sequestered into
abnormally shaped Promyelocytic-Nuclear Bodies
(PML-NB), identiļ¬ed as novel biomarkers in progeria.
We identiļ¬ed MG132 as being effective on progerin
degradation. Indeed, MG132 induces progerin nucleocy-
toplasmic translocation after a transition through the
nucleolus, and progerin clearance through macroau-
tophagy in HGPS patient ļ¬broblasts as well as in HGPS
patient iPSC-derived mesenchymal stem cells (MSCs)
and Vascular Smooth Muscle Cells (VSMCs). MG132
treatment improves cellular HGPS phenotypes, reduces
cellular senescence and enhances viability and prolifera-
tion in HGPS ļ¬broblasts. In vivo, through MG132 treat-
ment, progerin expression decreases in skeletal muscle
from LmnaG609G/G609G
mice. Altogether, we demonstrate
progerin reduction based on MG132 action and shed
light on a promising class of molecules towards a poten-
tial therapy for children with HGPS [55]. The observed
speciļ¬c effect of MG132 on progerin downregulation,
sparing normal A-type lamins, further supports the idea
that besides typical Progeria, other prelamin A-associ-
ated diseases might beneļ¬t from the same treatment.
Downregulation of Prelamin A aberrant splicing:
Antisense oligonucleotides, Metformin and MG132
The efļ¬ciency of an antisense therapeutic approach
using morpholino antisense oligonucleotides (AON)
268 K. HARHOURI ET AL.
in sterically blocking the aberrant LMNA splicing site
leading to progerin production has been previously
proven in vitro on human HGPS patientsā€™ cells and in
vivo on a knock-in LmnaG609G/G609G
mouse model
[56]. Indeed, Osorio et al. tested the combined admin-
istration of two AONs: ā€œMmEx11ā€ targeting the exon
11 aberrant splice site activated by the progeria muta-
tion in order to hamper its use, and ā€œMmEx10ā€, tar-
geting the physiological exon 10 splice site, in order to
reinforce the action of the ļ¬rst AON, by shifting splic-
ing events towards lamin C production. Similar results
were obtained by another group on the same
LmnaG609G/G609G
mouse model and in HGPS ļ¬bro-
blasts, but using another antisense oligonucleotide
(ASO) that reduces the binding of the splicing factor
SRSF-2 to exon 11 LMNA pre-mRNA, conļ¬rming the
efļ¬cacy of this approach [57].
Some patients carry other LMNA mutations affect-
ing exon 11 splicing and are named ā€œHGPS-likeā€
patients [58]. They also produce Progerin and/or
other truncated Prelamin A isoforms (D35 and D90).
Recently, we showed that downregulation of progerin
and other truncated or wild type Prelamin A isoforms
can be achieved via the antisense therapeutic approach
either in HGPS-like and Mandibuloacral Dysplasia
type B (MAD-B) patientsā€™ cells [59], establishing a
preclinical proof of principle for the use of antisense
morpholino oligonucleotides in HGPS, HGPS-like
and MAD-B syndromes. Nonetheless, the administra-
tion to children of the same molecules administered to
mice, i.e. vivo-morpholinos, could not be envisaged
due to their known toxicity. Therefore, the choice of
AON chemistry and route of administration will be
particularly important for future therapeutic
approaches involving splicing-modulation.
In human HGPS primary ļ¬broblasts and mouse
LmnaG609G/G609G 56
, the HGPS mutation induces the
use of an internal 5ā€™ cryptic splice site within exon 11
of the LMNA pre-mRNA, leading to an aberrant alter-
native splicing and production of a truncated form of
prelamin A (progerin). In 2011, the RNA-binding
protein SRSF-1 (for Serine/arginine-Rich Splicing Fac-
tor 1) was shown to favor this aberrant alternative
splicing [60]. On the other hand, it has been shown
that SRSF-1 expression is transcriptionally regulated
by the antidiabetic drug Metformin [61]. Based on
these ļ¬ndings, Egesipe et al. demonstrated that Met-
formin decreases SRSF-1 and progerin expression in
mesenchymal stem cells derived from HGPS induced
pluripotent stem cells (HGPS MSCs) and in several
other in vitro models of HGPS, i.e., human primary
HGPS ļ¬broblasts and LmnaG609G/G609G
mouse ļ¬bro-
blasts, resulting in improved nuclear shape abnormali-
ties and premature osteoblastic differentiation of
HGPS MSCs [62,63].
In HGPS ļ¬broblasts and in vivo via intramuscular
injections of MG132 in LmnaG609G/G609G
mice, we
recently demonstrated that, in addition to activating
macroautophagy leading to progerin degradation,
MG132 strongly reduces progerin production through
downregulation of SRSF-1, controlling prelamin A
mRNA aberrant splicing [55]. Further in vivo studies,
particularly on the LmnaG609G/G609G
mouse model,
will be needed to evaluate the effects of Metformin
and MG132 on HGPS phenotype improvement.
Reduction of progerin downstream toxic effects
Few approaches have been described to counteract the
altered downstream pathways caused by progerin
accumulation, including nuclear shape abnormalities,
ROS generation, accumulation of oxidized proteins,
mitochondrial dysfunction [14,64], cell senescence
and NF-kB activation, leading to the secretion of high
levels of the proinļ¬‚ammatory cytokines IL-6, CXCL1,
and TNF-a [25,65]. Treatment of progeroid ļ¬bro-
blasts with the ROS scavenger N-acetyl cysteine
(NAC) reduced the levels of un-repairable DSB and
improved their growth rates in culture [64]. Similarly,
it has also been shown that rho-associated protein
kinase (ROCK) regulates mitochondrial ROS genera-
tion by modulating the interaction between Rac1b and
cytochrome c [66]. Accordingly, in vitro treatment of
HGPS ļ¬broblasts with ROCK inhibitor (Y-27632)
resulted in decreased ROS levels and induced recovery
of mitochondrial function along with a reduction in
the frequency of abnormal nuclear morphology and
DNA double-strand breaks [66]. Elevated levels of
ROS and oxidative stress were also lowered by reacti-
vation of NRF2, whose transcriptional activity is
impaired in HGPS cells, resulting in improvements of
cellular HGPS defects [26]. Interestingly, MG132
seems to be a potentially effective drug to be used in
the prevention of oxidative damage in HGPS cells
through the activation of the NRF2 signaling pathway
[67ā€“69]. Mitochondrial dysfunction was reported in
HGPS ļ¬broblasts as well as in HGPS mouse models
[70,71]. To rescue mitochondrial defects, Xiong et al.
NUCLEUS 269
showed that treatment of HGPS cells with Methylene
Blue, an antioxidant compound known to stimulate
mitochondrial function, improves not only the mito-
chondrial morphology and function but also rescues
the premature aging phenotypes in HGPS cells includ-
ing nuclear morphology, perinuclear heterochromatin
loss and corrects misregulated gene expression [72].
In LmnaG609G/+
mice, mitochondrial dysfunction in
vascular smooth muscle cells (VSMCs) results in
increased tissue-nonspeciļ¬c alkaline phosphatase
activity and diminished ATP synthesis. Accordingly,
VSMCs have an impaired capacity to synthesize extra-
cellular pyrophosphate, a major inhibitor of vascular
calciļ¬cation [71]. Using LmnaG609G/G609G
, Villa-Bel-
losta et al. showed that aortic vascular calciļ¬cation
due to defective pyrophosphate production is counter-
balanced systemically by inorganic pyrophosphate
(PPi) treatment [71].
In order to explore the therapeutic potential of NF-
kB inhibition on HGPS disease parameters, Osorio et
al. showed that crossing Zmpste24Ā”/Ā”
mice with trans-
genic mice displaying reduced NF-kB signaling
extends longevity and prevents the development of
progeroid features. In the same study, and to the same
end, the authors also showed that sodium salicylate
treatment efļ¬ciently prevents NF-kB activation and
associated disease phenotypes in Zmpste24-deļ¬cient
mice, while also extending longevity in the
LmnaG609G/G609G
model [25]. Accordingly, NF-kB
activation impairs somatic cell reprogramming in
aging by eliciting the reprogramming repressor
DOT1L. The identiļ¬cation of this molecular mecha-
nism has allowed to translate this information into a
therapeutic approach, indeed, DOT1L inhibition by
epz-4777 extended longevity and prevented aging-
associated alterations in progeroid mice [73]. In the
same way, MG132 is also known to inhibit the secre-
tion of proinļ¬‚ammatory cytokines and attenuation of
IkB degradation, resulting in the abolition of NF-kB
activation [74-76]. These ļ¬ndings, together with the
fact that other models of normal and HGPS acceler-
ated aging show arteriosclerotic lesions with calciļ¬ca-
tion and inļ¬‚ammation [77], support the idea that
inļ¬‚ammation is a major regulator of the aging process
also in progeria and could be improved by several
inļ¬‚ammation-reducing treatments, including MG132.
Importantly, MG132 was reported to have a signiļ¬-
cant preventive and therapeutic effect on cardiovascu-
lar and renal injury [67,68,78], oxidative damage
[69,79] and on accelerated atherosclerosis in rabbits
[80]. These features are exhibited by HGPS patients
who might beneļ¬t from the same therapeutic effects.
On the other hand, compounds screening showed that
Remodelin, by inhibiting the lamina interacting
SUN1-associated acetyl-transferase protein NAT10,
improves nuclear shape and ļ¬tness of both progeric
and lamin A/Cā€“depleted cells, as observed by
decreased levels of the DNA double-strand break
markers gH2AX and autophosphorylated ATM
(ataxia telangiectasia mutated), decreased DNA dam-
age signaling and improved chromatin and nucleolar
organization [81].
Among the proteins affected by progerin accumula-
tion is the vitamin D receptor (VDR) whose levels are
reduced in HGPS cells. In addition, VDR knockout
mice develop a premature aging phenotype similar to
HGPS patients [82,83]. Interestingly, Kreienkamp et
al. recently showed that reconstituting VDR signaling
via 1a,25-dihydroxyvitamin D3 (1,25D), the active
hormonal form of vitamin D, improves HGPS pheno-
types, including nuclear morphological abnormalities,
DNA repair defects, and premature senescence [84].
Another important target of progerin is lamin A/C.
Indeed, progerin shows strong binding afļ¬nity for
lamin A/C exerting a negative dominant effect. On
this basis, Lee et al. identiļ¬ed new chemicals (JH4)
that can block the interaction between progerin and
lamin A/C through direct interaction with progerin.
Treatment with JH4 alleviated nuclear deformation
and reversed senescence and growth arrest markers.
Furthermore, administration of JH4 to LmnaG609G/
G609G
resulted in a marked improvement of several
progeria phenotypes including grip strength, body
weight, organ size and cell density, as well as lifespan
extension [85].
Resveratrol, a SIRT1 activator that interacts with
lamin A [86], has been proposed as an alternative
treatment for progeria. Indeed, in the premature aging
mouse model Zmpste24Ā”/Ā”
[87], due to the negative
dominant effect of progerin, SIRT-1 is weakly associ-
ated with the nuclear matrix: as a result deacetylase
activity is decreased, leading to a rapid depletion of
adult stem cells. By increasing SIRT1 interaction with
lamin A, Resveratrol rescues adult stem cell decline
and alleviates progeroid features in Zmpste24Ā”/Ā”
mice
[86]. Although this model reproduces the progeroid
syndromes phenotype, the underlying molecular
mechanism is different from that of progeria. Further
270 K. HARHOURI ET AL.
studies, using a model that produces progerin by the
same abnormal splicing mechanism observed in
humans, as the LmnaG609G/G609G
model, will be
needed to evaluate the impact of Resveratrol on the
disease phenotypes reversion. Another study revealed
that resveratrol treatment of an osteoblasts-speciļ¬c
progerin-expressing mouse model [88] does not show
overall beneļ¬cial effects [89]. Resveratrol efļ¬cacy has
been suboptimal due to poor bioavailability of this
compound and variable dose-linked effects, limiting
the full potential of SIRT1 activation treatment. A
novel formulation of micronized resveratrol, SRT501,
has been developed with improved bioavailability, tol-
erance and signiļ¬cant pharmacologic effect [90].
SRT501 open a new interesting research path on this
class of molecules towards the development of new
therapeutic approaches for progeria.
Recently, an in vivo study showed that partial
reprogramming by short-term cyclic expression of
Oct4, Sox2, Klf4, and c-Myc (OSKM) ameliorates cel-
lular and physiological hallmarks of aging and pro-
longs lifespan in LmnaG609G/G609G
mice [91,92].
Conclusion
Progress in progeria research led to a growing number
of promising therapeutic candidates (Figure 2). How-
ever, most of these approaches lack sufļ¬cient in vitro
and in vivo preclinical data to be transposed to
patients. Mostly, evidence of efļ¬cacy in an adapted
progeria animal model is required to exclude toxicity
and perform tentative determination of optimal doses,
dose frequencies, administration routes, and evalua-
tion endpoints. A speciļ¬c drug formulation requires
advanced preclinical toxicity testing to avoid serious
side effects that would prohibit long term use. It is
worth to note that, in HGPS preclinical studies, the
ability of a drug to preferentially target the cardiovas-
cular system should be evaluated, in that it represents
the major pathophysiological target in the disorder,
leading to premature death. Likewise, among treat-
ment efļ¬cacy readouts, cardiovascular measures
would be preferred. Increasing our understanding of
the disease biology will also be important to further
elucidate which impaired pathways are most relevant
to the disease, in order to identify both other treat-
ment pathophysiological targets and other outcome
measures. A limiting factor in advancing the develop-
ment of therapeutic approaches for progeria is that
studies are performed on primary cultures of patientā€™s
cells or animal models due to the limited number of
autopsy specimens from HGPS patients. To reproduce
key features of HGPS, iPSCs [93,94] and in vitro 3D
tissue model using human iPSC-derived cells [95]
thus provide relevant tools for drug screening.
To date, the amelioration of disease phenotypes in
HGPS mouse models by the different tested therapies
is limited, and this is even more true in HGPS
patients. One could speculate that the age of the
HGPS participant patients in the clinical trials could
determine the outcomes of the treatments, but accord-
ing to the clinical trial using lonafarnib, the authors
stated that no associations between age at time of
treatment and outcome measures that were improved
at end of study were identiļ¬ed. At the same time, it
should be noted that given the limited number of par-
ticipants, it is difļ¬cult to correlate age with outcomes
of the treatment.
As research progresses, it becomes clear that target-
ing the pathophysiology of progeria to a single level
will likely be insufļ¬cient to signiļ¬cantly alleviate the
signs and change the normal course of such a multi-
organ devastating disease. Altogether, there is a major
rationale in targeting progerin at different levels.
Addressing therapies in HGPS associated to progerin
accumulation may thus rely on multi-approaches
combination, including its decreased production,
increased degradation, or downstream noxious
cascades.
As a future therapeutic approach that still remains
to be evaluated for HGPS is the Clustered Regularly
Interspaced Short Palindromic Repeats/Cas protein
(CRISPR/Cas) for in vivo gene editing [96ā€“98] and
repair of the disease-causing mutation. On the other
hand, Adeno-Associated Virus (AAVs) have gained
popularity, in the last years, as safe gene delivery vec-
tors, due to their ability to mediate long-term expres-
sion in both non-dividing and dividing cells, with
speciļ¬c tissue tropism [99ā€“101]. In this regard, AAVs
constitute interesting CRISPR/Cas9 delivery candi-
dates to speciļ¬cally repair the progeria-causing muta-
tion. Preferentially, AAV serotype should target
VSMCs, since they are involved in heart attack and
stroke that represent the main cause of death in proge-
ria patients. To overcome the limited packaging
capacity of AAV (Ā» 4.8 kb), small Cas9 orthologues,
including Staphylococcus aureus-derived Cas9
(SaCas9, 3.16 kb) [102], has been used to package the
NUCLEUS 271
Cas9 and its gRNA into a single AAV delivery vehicle
for in vivo genome editing. CRISPR libraries should
be designed to ideally select those able to target the
mutation both in mouse and human genes with high
on-target activity and low off-target activity, so that
the effectiveness of the same gRNA could be studied
ļ¬rst on human HGPS ļ¬broblasts and then in vivo on
the Lmna G609G/G609G
mice.
Recent studies have shown the involvement of
microRNAs and in particular, mir-9 which prevents
progerin accumulation in HGPS neurons. Therefore,
another possibility for developing new therapeutic
approaches for progeria would be the modulation of
microRNAs, taking into consideration off-targets that,
again, must be evaluated in vitro and in vivo before
transposition to humans [103].
Figure 2. Treatment strategies for progeria and their targets. Envisaged treatments for progeria target several mechanisms triggering
the disease. By acting on the genetic cause, recent advances in genome editing using the CRISPR technique could be beneļ¬cial to repair
the mutation causing the disease. Other molecules target the aberrant splicing process leading to the production of a truncated protein,
by blocking the splice site with AONs or by inhibiting the splicing factor SRSF-1 (Metformin, MG132). During prelamin A maturation,
treatments inhibiting isoprenylation (farnesylation or geranylation) or the methylation have shown their effectiveness in reducing pro-
gerin toxicity (Statins, aminobisphonates, FTIs, GGTI-2147, monoaminopyrimidines, shICMT). To counter the accumulation of progerin,
several molecules have been described as activators of autophagy leading to progerin degradation (Rapamycin, Sulphoraphane, Reti-
noids and MG132). Finally, by acting on the reduction of the noxious downstream effects of progerin, some approaches have shown
their effectiveness against inļ¬‚ammation (sodium salicylate, DOT1L inhibition by epz-4777), oxidative stress (NAC, NRF2 reactivation),
nuclear shape abnormalities (Remodelin), mitochondrial abnormalities (Rock inhibitor: Y-27632, Methylene blue), impaired pyrophos-
phate metabolism (inorganic pyrophosphate: PPi), vitamin D receptor signaling (vitamin D), decreased deacetylase activity (Resveratrol),
Progerin/Lamin AC interaction (JH4) or aging hallmarks by reprogramming HGPS cells to pluripotency in vitro and in vivo.
272 K. HARHOURI ET AL.
Abbreviations
AON Antisense Oligonucleotides;
ATM Ataxia Telangiectasia Mutated;
BAF Barrier-to-Autointegration Factor;
CRISPR Clustered Regularly Interspaced Short Pal-
indromic Repeats;
DOT1L Disruptor Of Telomeric silencing 1-Like;
DSB Double-Strand Break;
EZH2 Enhancer of Zeste Homolog 2 polycomb
repressive complex 2 subunit;
FTase Farnesyltransferase;
FTI Farnesyl Transferase Inhibitor;
GGTI GeranylGeranylTransferase Inhibitor;
HDR Homology-Directed Repair;
HGPS Hutchinson-Gilford Progeria Syndrome;
HP1 Heterochromatin Protein 1;
ICMT Isoprenylcysteine
CarboxylMethylTransferase;
KLF4 Krā‚¬
uppel-Like Factor 4.
Lap2a Lamina-associated Polypeptide 2a;
MAD-B Mandibuloacral Dysplasia type B;
Mtor Mammalian Target Of Rapamycin;
MSC Mesenchymal Stem Cells;
NAC N-Acetyl Cysteine;
NAT10 N-acetyltransferase 10;
NFkB Nuclear Factor kappa B;
NHEJ Non-Homologous End Joining;
NRF2 Nuclear Factor-E2-related factor 2;
OCT4 Octamer-binding transcription factor 4;
PML-NB ProMyelocytic Leukemia-Nuclear Body;
ROS Reactive oxidative species;
SIRT-1 Sirtuin-1;
SOX2 Sex determining region Y-box 2;
SRSF-1 Serine/arginine-Rich Splicing Factor 1;
TNF Tumor Necrosis Factor;
VSMC Vascular Smooth Muscle Cells;
ZOPRA ZOledronate-PRAvastatin;
Disclosure of potential conļ¬‚icts of interest
No potential conļ¬‚icts of interest were disclosed.
Funding
This work was supported by grants from Institut National de la
Sant
e et de la Recherche M
edicale (INSERM), Aix-Marseille
University, A
Midex Foundation (VinTAGE Program) and
the Association FrancĢ§aise contre les Myopathies (AFM grant
MNH-Decrypt 2011ā€“2015 and TRIM-RD 2016ā€“2020 to NL).
This study is part of the FHU A
MIDEX project MARCHE n.
ANR-11-IDEX-001-02 funded by the ā€œInvestissement
dā€™avenirā€ French governmental program, managed by the
French National Research Agency (ANR).
ORCID
Karim Harhouri http://orcid.org/0000-0003-2904-5221
Diane Frankel http://orcid.org/0000-0003-3321-6951
Patrice Roll http://orcid.org/0000-0002-0045-5641
Annachiara De Sandre-Giovannoli http://orcid.org/0000-
0002-2324-2462
Nicolas L
evy http://orcid.org/0000-0001-5171-6365
References
[1] Hennekam RC. Hutchinson-Gilford progeria syndrome:
review of the phenotype. Am J Med Genet A.
2006;140:2603ā€“24.
[2] Merideth MA, Gordon LB, Clauss S, et al. Phenotype
and course of Hutchinson-Gilford progeria syndrome.
N Engl J Med. 2008;358:592ā€“604.
[3] Hutchinson J. Congenital absence of hair and mam-
mary glands with atrophic condition of the skin and its
appendages, in a boy whose mother had been almost
wholly bald from Alopecia Areata from the age of six.
Med Chir Trans. 1886;69:473ā€“7.
[4] Gilford H. On a condition of mixed premature and
immature development. Med Chir Trans. 1897;80:17-
46:25.
[5] De Sandre-Giovannoli A, Bernard R, Cau P, et al. Lamin
a truncation in Hutchinson-Gilford progeria. Science.
2003;300:2055.
[6] Eriksson M, Brown WT, Gordon LB, et al. Recurrent de
novo point mutations in lamin A cause Hutchinson-
Gilford progeria syndrome. Nature. 2003;423:293ā€“8.
[7] Goldman RD, Shumaker DK, Erdos MR, et al. Accumu-
lation of mutant lamin A causes progressive changes in
nuclear architecture in Hutchinson-Gilford progeria
syndrome. Proc Natl Acad Sci U S A. 2004;101:8963ā€“8.
[8] Capell BC, Erdos MR, Madigan JP, et al. Inhibiting far-
nesylation of progerin prevents the characteristic
nuclear blebbing of Hutchinson-Gilford progeria syn-
drome. Proc Natl Acad Sci U S A. 2005;102:12879ā€“84.
[9] Scafļ¬di P, Misteli T. Reversal of the cellular phenotype
in the premature aging disease Hutchinson-Gilford pro-
geria syndrome. Nat Med. 2005;11:440ā€“5.
[10] Huang S, Chen L, Libina N, et al. Correction of cellular
phenotypes of Hutchinson-Gilford Progeria cells by
RNA interference. Hum Genet. 2005;118:444ā€“50.
[11] Burtner CR, Kennedy BK. Progeria syndromes and age-
ing: What is the connection? Nat Rev Mol Cell Biol.
2010;11:567ā€“78.
[12] Liu B, Wang J, Chan KM, et al. Genomic instability in
laminopathy-based premature aging. Nat Med.
2005;11:780ā€“5.
[13] Hilton BA, Liu J, Cartwright BM, et al. Progerin seques-
tration of PCNA promotes replication fork collapse and
NUCLEUS 273
mislocalization of XPA in laminopathy-related proge-
roid syndromes. FASEB J. 2017;31:3882ā€“93.
[14] Viteri G, Chung YW, Stadtman ER. Effect of progerin
on the accumulation of oxidized proteins in ļ¬broblasts
from Hutchinson Gilford progeria patients. Mech Age-
ing Dev. 2010;131:2ā€“8.
[15] Rivera-Torres J, Acin-Perez R, Cabezas-Sanchez P, et al.
Identiļ¬cation of mitochondrial dysfunction in Hutchin-
son-Gilford progeria syndrome through use of stable
isotope labeling with amino acids in cell culture. J Prote-
omics. 2013;91:466ā€“77.
[16] Columbaro M, Capanni C, Mattioli E, et al. Rescue of
heterochromatin organization in Hutchinson-Gilford
progeria by drug treatment. Cell Mol Life Sci.
2005;62:2669ā€“78.
[17] Kubben N, Adriaens M, Meuleman W, et al. Mapping of
lamin A- and progerin-interacting genome regions.
Chromosoma. 2012;121:447ā€“64.
[18] McCord RP, Nazario-Toole A, Zhang H, et al. Corre-
lated alterations in genome organization, histone meth-
ylation, and DNA-lamin A/C interactions in
Hutchinson-Gilford progeria syndrome. Genome Res.
2013;23:260ā€“9.
[19] Shumaker DK, Dechat T, Kohlmaier A, et al. Mutant
nuclear lamin A leads to progressive alterations of epi-
genetic control in premature aging. Proc Natl Acad Sci
U S A. 2006;103:8703ā€“8.
[20] Scafļ¬di P, Misteli T. Lamin A-dependent misregulation
of adult stem cells associated with accelerated ageing.
Nat Cell Biol. 2008;10:452ā€“9.
[21] Liu Y, Wang Y, Rusinol AE, et al. Involvement of xero-
derma pigmentosum group A (XPA) in progeria arising
from defective maturation of prelamin A. Faseb J.
2008;22:603ā€“11.
[22] Manju K, Muralikrishna B, Parnaik VK. Expression of
disease-causing lamin A mutants impairs the formation
of DNA repair foci. J Cell Sci. 2006;119:2704ā€“14.
[23] Prokocimer M, Barkan R, Gruenbaum Y. Hutchinson-
Gilford progeria syndrome through the lens of tran-
scription. Aging Cell. 2013;12:533ā€“43.
[24] Hernandez L, Roux KJ, Wong ESM, et al. Functional
coupling between the extracellular matrix and nuclear
Lamina by wnt signaling in Progeria. Dev Cell.
2010;19:413ā€“25.
[25] Osorio FG, Barcena C, Soria-Valles C, et al. Nuclear
lamina defects cause ATM-dependent NF-kappaB acti-
vation and link accelerated aging to a systemic inļ¬‚am-
matory response. Genes Dev. 2012;26:2311ā€“24.
[26] Kubben N, Zhang W, Wang L, et al. Repression of the
Antioxidant NRF2 Pathway in Premature Aging. Cell.
2016;165:1361ā€“74.
[27] Cau P, Navarro C, Harhouri K, et al. Nuclear matrix,
nuclear envelope and premature aging syndromes in a
translational research perspective. Seminars Cell Dev
Biol. 2014;29:125ā€“47.
[28] Delbarre E, Tramier M, Coppey-Moisan M, et al. The
truncated prelamin A in Hutchinson-Gilford progeria
syndrome alters segregation of A-type and B-type lamin
homopolymers. Hum Mol Genetics. 2006;15:1113ā€“22.
[29] Basso AD, Kirschmeier P, Bishop WR. Lipid posttrans-
lational modiļ¬cations. Farnesyl transferase inhibitors. J
Lipid Res. 2006;47:15ā€“31.
[30] Glynn MW, Glover TW. Incomplete processing of mutant
lamin A in Hutchinson-Gilford progeria leads to nuclear
abnormalities, which are reversed by farnesyltransferase
inhibition. Hum Mol Genetics. 2005;14:2959ā€“69.
[31] Toth JI, Yang SH, Qiao X, et al. Blocking protein farne-
syltransferase improves nuclear shape in ļ¬broblasts
from humans with progeroid syndromes. Proc Natl
Acad Sci U S A. 2005;102:12873ā€“8.
[32] Yang SH, Bergo MO, Toth JI, et al. Blocking protein far-
nesyltransferase improves nuclear blebbing in mouse
ļ¬broblasts with a targeted Hutchinson-Gilford progeria
syndrome mutation. Proc Natl Acad Sci U S A.
2005;102:10291ā€“6.
[33] Mallampalli MP, Huyer G, Bendale P, et al. Inhibiting
farnesylation reverses the nuclear morphology defect in
a HeLa cell model for Hutchinson-Gilford progeria syn-
drome. Proc Natl Acad Sci U S A. 2005;102:14416ā€“21.
[34] Yang SH, Meta M, Qiao X, et al. A farnesyltransferase
inhibitor improves disease phenotypes in mice with a
Hutchinson-Gilford progeria syndrome mutation. J
Clin Invest. 2006;116:2115ā€“21.
[35] Yang SH, Andres DA, Spielmann HP, et al. Progerin elicits
disease phenotypes of progeria in mice whether or not it is
farnesylated. J Clin Invest. 2008;118:3291ā€“300.
[36] Capell BC, Olive M, Erdos MR, et al. A farnesyltransfer-
ase inhibitor prevents both the onset and late progres-
sion of cardiovascular disease in a progeria mouse
model. Proc Natl Acad Sci U S A. 2008;105:15902ā€“7.
[37] Gordon LB, Kleinman ME, Miller DT, et al. Clinical
trial of a farnesyltransferase inhibitor in children with
Hutchinson-Gilford progeria syndrome. Proc Natl Acad
Sci U S A. 2012;109:16666ā€“71.
[38] Gordon LB, Massaro J, Dā€™Agostino RB, et al. Impact of
Farnesylation inhibitors on survival in Hutchinson-Gil-
ford Progeria syndrome. Circulation. 2014;130:27
[39] Varela I, Pereira S, Ugalde AP, et al. Combined treat-
ment with statins and aminobisphosphonates extends
longevity in a mouse model of human premature aging.
Nat Med. 2008;14:767ā€“72.
[40] Gordon LB, Kleinman ME, Massaro J, et al. Clinical
Trial of the Protein Farnesylation inhibitors Lonafarnib,
Pravastatin, and Zoledronic Acid in Children With
Hutchinson-Gilford Progeria Syndrome. Circulation.
2016;134:114ā€“25.
[41] Davies BS, Barnes RH, 2nd, Tu Y, et al. An accumula-
tion of non-farnesylated prelamin A causes cardiomy-
opathy but not progeria. Hum Mol Genetics.
2010;19:2682ā€“94.
[42] Blondel S, Egesipe AL, Picardi P, et al. Drug screening
on Hutchinson Gilford progeria pluripotent stem cells
reveals aminopyrimidines as new modulators of farne-
sylation. Cell Death Dis. 2016;7:e2105.
274 K. HARHOURI ET AL.
[43] Ibrahim MX, Sayin VI, Akula MK, et al. Targeting iso-
prenylcysteine methylation ameliorates disease in a
mouse model of progeria. Science. 2013;340:1330ā€“3.
[44] Cao K, Graziotto JJ, Blair CD, et al. Rapamycin reverses
cellular phenotypes and enhances mutant protein clear-
ance in Hutchinson-Gilford progeria syndrome cells.
Sci Transl Med. 2011;3:89ra58.
[45] Graziotto JJ, Cao K, Collins FS, et al. Rapamycin acti-
vates autophagy in Hutchinson-Gilford progeria syn-
drome: implications for normal aging and age-
dependent neurodegenerative disorders. Autophagy.
2012;8:147ā€“51.
[46] Cenni V, Capanni C, Columbaro M, et al. Autophagic
degradation of farnesylated prelamin A as a therapeutic
approach to lamin-linked progeria (vol 55, e36, 2011).
Eur J Histochem. 2013;57:283ā€“6.
[47] Ramos FJ, Chen SC, Garelick MG, et al. Rapamycin
reverses elevated mTORC1 signaling in Lamin A/C-
deļ¬cient mice, rescues cardiac and skeletal muscle func-
tion, and extends survival. Sci Translational Med.
2012;4:144ra103
[48] Liao CY, Anderson SS, Chicoine NH, et al. Rapamycin
reverses metabolic deļ¬cits in Lamin A/C-Deļ¬cient
mice. Cell Rep. 2016;17:2542ā€“52.
[49] Cho HJ, Park J, Lee HW, et al. Regulation of adipocyte
differentiation and insulin action with rapamycin. Bio-
chem Bioph Res Co. 2004;321:942ā€“8.
[50] Yeh WC, Bierer BE, Mcknight SL. Rapamycin inhibits
clonal expansion and adipogenic differentiation of 3t3-
L1 cells. Proc Natl Acad Sci U S A. 1995;92:11086ā€“90.
[51] Gabriel D, Roedl D, Gordon LB, et al. Sulforaphane
enhances progerin clearance in Hutchinson-Gilford
progeria ļ¬broblasts. Aging Cell. 2015;14:78ā€“91.
[52] Gabriel D, Shafry DD, Gordon LB, et al. Intermittent treat-
ment with farnesyltransferase inhibitor and sulforaphane
improves cellular homeostasis in Hutchinson-Gilford pro-
geria ļ¬broblasts. Oncotarget. 2017;8:64809ā€“26.
[53] Kubben N, Brimacombe KR, Donegan M, et al. A high-
content imaging-based screening pipeline for the sys-
tematic identiļ¬cation of anti-progeroid compounds.
Methods. 2016;96:46ā€“58.
[54] Pellegrini C, Columbaro M, Capanni C, et al. All-trans reti-
noic acid and rapamycin normalize Hutchinson Gilford pro-
geria ļ¬broblast phenotype. Oncotarget. 2015;6:29914ā€“28.
[55] Harhouri K, Navarro C, Depetris D, et al. MG132-
induced progerin clearance is mediated by autophagy
activation and splicing regulation. EMBO Mol Med.
2017;9:1294ā€“313.
[56] Osorio FG, Navarro CL, Cadinanos J, et al. Splicing-
directed therapy in a new mouse model of human accel-
erated aging. Sci Transl Med. 2011;3:106ra7.
[57] Lee JM, Nobumori C, Tu Y, et al. Modulation of LMNA
splicing as a strategy to treat prelamin A diseases. J Clin
Invest. 2016;126:1592ā€“602.
[58] Barthelemy F, Navarro C, Fayek R, et al. Truncated prel-
amin A expression in HGPS-like patients: a transcrip-
tional study. Eur J Hum Genet. 2015;23:1051ā€“61.
[59] Harhouri K, Navarro C, Baquerre C, et al. Antisense-
based Progerin downregulation in HGPS-Like Patientsā€™
cells. Cells. 2016;5. pii: E31
[60] Lopez-Mejia IC, Vautrot V, De Toledo M, Behm-Ansm-
ant I, et al. A conserved splicing mechanism of the
LMNA gene controls premature aging. Hum Mol
Genetics. 2011;20:4540ā€“55.
[61] Larsson O, Morita M, Topisirovic I, et al. Distinct pertur-
bation of the translatome by the antidiabetic drug metfor-
min. Proc Natl Acad Sci U S A. 2012;109:8977ā€“82.
[62] Egesipe AL, Blondel S, Cicero AL, et al. Metformin
decreases progerin expression and alleviates pathologi-
cal defects of Hutchinson-Gilford progeria syndrome
cells. NPJ Aging Mechan Dis. 2016;2:16026.
[63] Park SK, Shin OS. Metformin alleviates ageing cellular
phenotypes in Hutchinson-Gilford progeria syndrome
dermal ļ¬broblasts. Exp Dermatol. 2017;26:889ā€“95.
[64] Richards SA, Muter J, Ritchie P, et al. The accumulation
of un-repairable DNA damage in laminopathy progeria
ļ¬broblasts is caused by ROS generation and is prevented
by treatment with N-acetyl cysteine. Hum Mol Genetics.
2011;20:3997ā€“4004.
[65] Tilstra JS, Clauson CL, Niedernhofer LJ, et al. NF-kap-
paB in aging and disease. Aging Dis. 2011;2:449ā€“65.
[66] Kang HT, Park JT, Choi K, et al. Chemical screening
identiļ¬es ROCK as a target for recovering mitochon-
drial function in Hutchinson-Gilford progeria syn-
drome. Aging Cell. 2017;16:541ā€“50.
[67] Cui W, Bai Y, Luo P, et al. Preventive and therapeutic
effects of MG132 by activating Nrf2-ARE signaling path-
way on oxidative stress-induced cardiovascular and renal
injury. Oxidative Med Cell Longevity. 2013;2013:306073.
[68] Wang Y, Sun W, Du B, et al. Therapeutic effect of MG-
132 on diabetic cardiomyopathy is associated with its
suppression of proteasomal activities: roles of Nrf2 and
NF-kappaB. Am J Physiol Heart Circulatory Physiol.
2013;304:H567ā€“78.
[69] Dreger H, Westphal K, Wilck N, et al. Protection of vas-
cular cells from oxidative stress by proteasome inhibition
depends on Nrf2. Cardiovascular Res. 2010;85:395ā€“403.
[70] Rivera-Torres J, Acin-Perez R, Cabezas-Sanchez P, et al.
Identiļ¬cation of mitochondrial dysfunction in Hutchin-
son-Gilford progeria syndrome through use of stable
isotope labeling with amino acids in cell culture. J Prote-
omics. 2013;91:466ā€“77.
[71] Villa-Bellosta R, Rivera-Torres J, Osorio FG, et al.
Defective extracellular pyrophosphate metabolism
promotes vascular calciļ¬cation in a mouse model
of Hutchinson-Gilford Progeria Syndrome that is Ame-
liorated on Pyrophosphate treatment. Circulation.
2013;127:2442ā€“51.
[72] Xiong ZM, Choi JY, Wang K, et al. Methylene blue alle-
viates nuclear and mitochondrial abnormalities in pro-
geria. Aging Cell. 2016;15:279ā€“90.
[73] Soria-Valles C, Osorio FG, Gutierrez-Fernandez A, et al.
NF-kappaB activation impairs somatic cell reprogram-
ming in ageing. Nat Cell Biol. 2015;17:1004ā€“13.
NUCLEUS 275
[74] Ortiz-Lazareno PC, Hernandez-Flores G, Dominguez-
Rodriguez JR, et al. MG132 proteasome inhibitor modu-
lates proinļ¬‚ammatory cytokines production and expres-
sion of their receptors in U937 cells: involvement of
nuclear factor-kappaB and activator protein-1. Immu-
nology. 2008;124:534ā€“41.
[75] Hozhabri NST, Kim H, Varanasi V. NF-kappa B inhibi-
tor MG132 enhances differentiation and collagen
expression of dental pulp stem cells. Faseb J. 2014;28
[76] Ahmed AS, Ahmed M, Li J, et al. Proteasome inhibitor
MG132 modulates inļ¬‚ammatory pain by central mecha-
nisms in adjuvant arthritis. Int J Rheum Dis. 2017;20:25ā€“32.
[77] Olive M, Harten I, Mitchell R, et al. Cardiovascular
pathology in Hutchinson-Gilford progeria: correlation
with the vascular pathology of aging. Arterioscler
Thromb Vasc Biol. 2010;30:2301ā€“9.
[78] Chatterjee PK, Yeboah MM, Dowling O, et al. Nicotinic
acetylcholine receptor agonists attenuate septic acute
kidney injury in mice by suppressing inļ¬‚ammation and
proteasome activity. PloS One. 2012;7:e35361.
[79] Miao X, Cui W, Sun W, et al. Therapeutic effect of
MG132 on the aortic oxidative damage and inļ¬‚amma-
tory response in OVE26 type 1 diabetic mice. Oxidative
Med Cell Longevity. 2013;2013:879516.
[80] Feng B, Zhang Y, Mu J, et al. Preventive effect of a pro-
teasome inhibitor on the formation of accelerated ath-
erosclerosis in rabbits with uremia. J Cardiovascular
Pharmacol. 2010;55:129ā€“38.
[81] Larrieu D, Britton S, Demir M, et al. Chemical inhibi-
tion of NAT10 corrects defects of laminopathic cells.
Science. 2014;344:527ā€“32.
[82] Bouillon R, Carmeliet G, Verlinden L, et al. Vitamin D
and human health: Lessons from Vitamin D receptor
null mice. Endocr Rev. 2008;29:726ā€“76.
[83] Plum LA, DeLuca HF. Vitamin D, disease and therapeutic
opportunities. Nat Rev Drug Discovery. 2010;9:941ā€“55.
[84] Kreienkamp R, Croke M, Neumann MA, et al. Vitamin D
receptor signaling improves Hutchinson-Gilford progeria
syndrome cellular phenotypes. Oncotarget. 2016;7:30018ā€“31.
[85] Lee SJ, Jung YS, Yoon MH, et al. Interruption of pro-
gerin-lamin A/C binding ameliorates Hutchinson-Gil-
ford progeria syndrome phenotype. J Clin Invest.
2016;126:3879ā€“93.
[86] Liu BH, Ghosh S, Yang X, et al. Resveratrol Rescues
SIRT1-Dependent Adult Stem Cell Decline and Allevi-
ates Progeroid Features in Laminopathy-Based Progeria.
Cell Metab. 2012;16:738ā€“50.
[87] Varela I, Pereira S, Ugalde AP, et al. Combined treat-
ment with statins and aminobisphosphonates extends
longevity in a mouse model of human premature aging.
Nat Med. 2008;14:767ā€“72.
[88] Schmidt E, Nilsson O, Koskela A, et al. Expression of
the Hutchinson-Gilford progeria mutation during oste-
oblast development results in loss of osteocytes,
irregular mineralization, and poor biomechanical prop-
erties. J Biol Chem. 2012;287:33512ā€“22.
[89] Strandgren C, Nasser HA, McKenna T, et al. Transgene
silencing of the Hutchinson-Gilford progeria syndrome
mutation results in a reversible bone phenotype,
whereas resveratrol treatment does not show overall
beneļ¬cial effects. FASEB J. 2015;29:3193ā€“205.
[90] Howells LM, Berry DP, Elliott PJ, et al. Phase I random-
ized, double-blind pilot study of micronized resveratrol
(SRT501) in patients with hepatic metastasesā€“safety,
pharmacokinetics, and pharmacodynamics. Cancer Pre-
vention Res. 2011;4:1419ā€“25.
[91] Ocampo A, Reddy P, Martinez-Redondo P, et al. In
Vivo Amelioration of age-associated Hallmarks by par-
tial reprogramming. Cell. 2016;167:1719ā€“33 e12.
[92] Ocampo A, Reddy P, Izpisua Belmonte JC. Anti-aging
strategies based on cellular reprogramming. Trends Mol
Med. 2016;22:725ā€“38.
[93] Liu GH, Barkho BZ, Ruiz S, et al. Recapitulation of pre-
mature ageing with iPSCs from Hutchinson-Gilford
progeria syndrome. Nature. 2011;472:221ā€“5.
[94] Zhang JQ, Lian QZ, Zhu GL, et al. A human iPSC model
of Hutchinson Gilford Progeria reveals vascular smooth
muscle and Mesenchymal stem cell defects. Cell Stem
Cell. 2011;8:31ā€“45.
[95] Atchison L, Zhang HY, Cao K, et al. A tissue engineered
blood vessel model of Hutchinson-Gilford Progeria
Syndrome using human iPSC-derived smooth muscle
cells. Sci Rep-Uk. 2017;7:8168.
[96] Sander JD, Joung JK. CRISPR-Cas systems for editing,
regulating and targeting genomes. Nat Biotechnol.
2014;32:347ā€“55.
[97] Doudna JA, Charpentier E. Genome editing. The new
frontier of genome engineering with CRISPR-Cas9. Sci-
ence. 2014;346:1258096.
[98] Hsu PD, Lander ES, Zhang F. Development and appli-
cations of CRISPR-Cas9 for genome engineering. Cell.
2014;157:1262ā€“78.
[99] Kotterman MA, Schaffer DV. Engineering adeno-asso-
ciated viruses for clinical gene therapy. Nat Rev Genet-
ics. 2014;15:445ā€“51.
[100] Yin H, Kauffman KJ, Anderson DG. Delivery technolo-
gies for genome editing. Nat Rev Drug Discovery.
2017;16:387ā€“99.
[101] Suzuki K, Tsunekawa Y, Hernandez-Benitez R, et al. In
vivo genome editing via CRISPR/Cas9 mediated homol-
ogy-independent targeted integration. Nature.
2016;540:144ā€“9.
[102] Ran FA, Cong L, Yan WX, et al. In vivo genome edit-
ing using Staphylococcus aureus Cas9. Nature.
2015;520:186ā€“U98.
[103] Frankel D, Delecourt V, Harhouri K, et al. MicroRNAs
in hereditary and sporadic premature aging syndromes
and other laminopathies. Aging Cell. 2018;e12766.
276 K. HARHOURI ET AL.

More Related Content

Similar to An Overview Of Treatment Strategies For Hutchinson-Gilford Progeria Syndrome

Topical hemostatic agents in surgical practice m. longoni e l. santoleri
Topical hemostatic agents in surgical practice   m. longoni e l. santoleriTopical hemostatic agents in surgical practice   m. longoni e l. santoleri
Topical hemostatic agents in surgical practice m. longoni e l. santolerianemo_site
Ā 
070125 chemotherapy for hn scc2
070125 chemotherapy for hn scc2070125 chemotherapy for hn scc2
070125 chemotherapy for hn scc2Asha Jangam
Ā 
FEOCROMOCITOMA PHYSIOLOGIA ENFERMEDAD ADRENAL
FEOCROMOCITOMA PHYSIOLOGIA ENFERMEDAD ADRENALFEOCROMOCITOMA PHYSIOLOGIA ENFERMEDAD ADRENAL
FEOCROMOCITOMA PHYSIOLOGIA ENFERMEDAD ADRENALAdictivitySong
Ā 
Gallbladder cancer patient analysis
Gallbladder cancer patient analysisGallbladder cancer patient analysis
Gallbladder cancer patient analysisAnton Yuryev
Ā 
Hemophagocytic lymphohistiocytosis (HLH)
Hemophagocytic lymphohistiocytosis (HLH)Hemophagocytic lymphohistiocytosis (HLH)
Hemophagocytic lymphohistiocytosis (HLH)Bincy Thankachan
Ā 
1 s2.0-s0006497120333553-main
1 s2.0-s0006497120333553-main1 s2.0-s0006497120333553-main
1 s2.0-s0006497120333553-mainjessicadesantiagoram
Ā 
Molecular Detection and Therapeutic Management of Feline Mycoplasmosis
Molecular Detection and Therapeutic Management of Feline MycoplasmosisMolecular Detection and Therapeutic Management of Feline Mycoplasmosis
Molecular Detection and Therapeutic Management of Feline MycoplasmosisIOSRJAVS
Ā 
Fisiopatologia y diagnoĢstico de cid
Fisiopatologia y diagnoĢstico de cidFisiopatologia y diagnoĢstico de cid
Fisiopatologia y diagnoĢstico de cidJose Alberto Vejar
Ā 
Antihistamines (medicinal chemistry) manik
Antihistamines (medicinal chemistry) manikAntihistamines (medicinal chemistry) manik
Antihistamines (medicinal chemistry) manikImran Nur Manik
Ā 
CHEMOTHERAPY_RDP_GENERAL PRINCIPLES OF CHEMOTHERAPY.pdf
CHEMOTHERAPY_RDP_GENERAL PRINCIPLES OF CHEMOTHERAPY.pdfCHEMOTHERAPY_RDP_GENERAL PRINCIPLES OF CHEMOTHERAPY.pdf
CHEMOTHERAPY_RDP_GENERAL PRINCIPLES OF CHEMOTHERAPY.pdfrishi2789
Ā 
Anti- leishmaniasis drugs.ppt.pptx
Anti- leishmaniasis drugs.ppt.pptxAnti- leishmaniasis drugs.ppt.pptx
Anti- leishmaniasis drugs.ppt.pptxTolDig
Ā 
Combining Old and New: Sensitising Drugs and Other Vaccines To Augment Effica...
Combining Old and New: Sensitising Drugs and Other Vaccines To Augment Effica...Combining Old and New: Sensitising Drugs and Other Vaccines To Augment Effica...
Combining Old and New: Sensitising Drugs and Other Vaccines To Augment Effica...NeuroAcademy
Ā 

Similar to An Overview Of Treatment Strategies For Hutchinson-Gilford Progeria Syndrome (20)

Topical hemostatic agents in surgical practice m. longoni e l. santoleri
Topical hemostatic agents in surgical practice   m. longoni e l. santoleriTopical hemostatic agents in surgical practice   m. longoni e l. santoleri
Topical hemostatic agents in surgical practice m. longoni e l. santoleri
Ā 
070125 chemotherapy for hn scc2
070125 chemotherapy for hn scc2070125 chemotherapy for hn scc2
070125 chemotherapy for hn scc2
Ā 
FEOCROMOCITOMA PHYSIOLOGIA ENFERMEDAD ADRENAL
FEOCROMOCITOMA PHYSIOLOGIA ENFERMEDAD ADRENALFEOCROMOCITOMA PHYSIOLOGIA ENFERMEDAD ADRENAL
FEOCROMOCITOMA PHYSIOLOGIA ENFERMEDAD ADRENAL
Ā 
Omara-Opyene et al 2004
Omara-Opyene et al 2004Omara-Opyene et al 2004
Omara-Opyene et al 2004
Ā 
TAXANES AND PODOPHYLLOTOXINS
TAXANES AND PODOPHYLLOTOXINSTAXANES AND PODOPHYLLOTOXINS
TAXANES AND PODOPHYLLOTOXINS
Ā 
NMT_Nature
NMT_NatureNMT_Nature
NMT_Nature
Ā 
Gallbladder cancer patient analysis
Gallbladder cancer patient analysisGallbladder cancer patient analysis
Gallbladder cancer patient analysis
Ā 
Hemophagocytic lymphohistiocytosis (HLH)
Hemophagocytic lymphohistiocytosis (HLH)Hemophagocytic lymphohistiocytosis (HLH)
Hemophagocytic lymphohistiocytosis (HLH)
Ā 
1 s2.0-s0006497120333553-main
1 s2.0-s0006497120333553-main1 s2.0-s0006497120333553-main
1 s2.0-s0006497120333553-main
Ā 
Molecular Detection and Therapeutic Management of Feline Mycoplasmosis
Molecular Detection and Therapeutic Management of Feline MycoplasmosisMolecular Detection and Therapeutic Management of Feline Mycoplasmosis
Molecular Detection and Therapeutic Management of Feline Mycoplasmosis
Ā 
M18 paper
M18 paperM18 paper
M18 paper
Ā 
Inflammation
InflammationInflammation
Inflammation
Ā 
Fisiopatologia y diagnoĢstico de cid
Fisiopatologia y diagnoĢstico de cidFisiopatologia y diagnoĢstico de cid
Fisiopatologia y diagnoĢstico de cid
Ā 
Antihistamines (medicinal chemistry) manik
Antihistamines (medicinal chemistry) manikAntihistamines (medicinal chemistry) manik
Antihistamines (medicinal chemistry) manik
Ā 
IJAA
IJAAIJAA
IJAA
Ā 
Fg5V2T
Fg5V2TFg5V2T
Fg5V2T
Ā 
CHEMOTHERAPY_RDP_GENERAL PRINCIPLES OF CHEMOTHERAPY.pdf
CHEMOTHERAPY_RDP_GENERAL PRINCIPLES OF CHEMOTHERAPY.pdfCHEMOTHERAPY_RDP_GENERAL PRINCIPLES OF CHEMOTHERAPY.pdf
CHEMOTHERAPY_RDP_GENERAL PRINCIPLES OF CHEMOTHERAPY.pdf
Ā 
Anti- leishmaniasis drugs.ppt.pptx
Anti- leishmaniasis drugs.ppt.pptxAnti- leishmaniasis drugs.ppt.pptx
Anti- leishmaniasis drugs.ppt.pptx
Ā 
Combining Old and New: Sensitising Drugs and Other Vaccines To Augment Effica...
Combining Old and New: Sensitising Drugs and Other Vaccines To Augment Effica...Combining Old and New: Sensitising Drugs and Other Vaccines To Augment Effica...
Combining Old and New: Sensitising Drugs and Other Vaccines To Augment Effica...
Ā 
Monoclonal antibodies
Monoclonal antibodiesMonoclonal antibodies
Monoclonal antibodies
Ā 

More from Angie Miller

Writing Poetry In The Upper Grades Poetry Lessons,
Writing Poetry In The Upper Grades Poetry Lessons,Writing Poetry In The Upper Grades Poetry Lessons,
Writing Poetry In The Upper Grades Poetry Lessons,Angie Miller
Ā 
ReMarkable 2 Is A 10.3-Inch E-Paper Tablet With A Stylus, Starts At
ReMarkable 2 Is A 10.3-Inch E-Paper Tablet With A Stylus, Starts AtReMarkable 2 Is A 10.3-Inch E-Paper Tablet With A Stylus, Starts At
ReMarkable 2 Is A 10.3-Inch E-Paper Tablet With A Stylus, Starts AtAngie Miller
Ā 
Printable Lined Paper For Kids That Are Soft Harper Blog
Printable Lined Paper For Kids That Are Soft Harper BlogPrintable Lined Paper For Kids That Are Soft Harper Blog
Printable Lined Paper For Kids That Are Soft Harper BlogAngie Miller
Ā 
Writing Your Introduction, Transitions, And Conclusion
Writing Your Introduction, Transitions, And ConclusionWriting Your Introduction, Transitions, And Conclusion
Writing Your Introduction, Transitions, And ConclusionAngie Miller
Ā 
Groundhog Day Writing Paper
Groundhog Day Writing PaperGroundhog Day Writing Paper
Groundhog Day Writing PaperAngie Miller
Ā 
5 Writing Tips To Help Overcome Anxiety Youn
5 Writing Tips To Help Overcome Anxiety Youn5 Writing Tips To Help Overcome Anxiety Youn
5 Writing Tips To Help Overcome Anxiety YounAngie Miller
Ā 
How To Write An Essay In 6 Simple Steps ScoolWork
How To Write An Essay In 6 Simple Steps ScoolWorkHow To Write An Essay In 6 Simple Steps ScoolWork
How To Write An Essay In 6 Simple Steps ScoolWorkAngie Miller
Ā 
Scroll Paper - Cliparts.Co
Scroll Paper - Cliparts.CoScroll Paper - Cliparts.Co
Scroll Paper - Cliparts.CoAngie Miller
Ā 
Hnh Nh Bn, S Tay, Vit, Cng Vic, Ang Lm Vic, Sch, Ngi
Hnh Nh Bn, S Tay, Vit, Cng Vic, Ang Lm Vic, Sch, NgiHnh Nh Bn, S Tay, Vit, Cng Vic, Ang Lm Vic, Sch, Ngi
Hnh Nh Bn, S Tay, Vit, Cng Vic, Ang Lm Vic, Sch, NgiAngie Miller
Ā 
Recycling Essay Essay On Re
Recycling Essay Essay On ReRecycling Essay Essay On Re
Recycling Essay Essay On ReAngie Miller
Ā 
Pin On PAPER SHEETS
Pin On PAPER SHEETSPin On PAPER SHEETS
Pin On PAPER SHEETSAngie Miller
Ā 
Pin By Cloe Einam On Referencing Harvard Referencing, Essay, Essa
Pin By Cloe Einam On Referencing Harvard Referencing, Essay, EssaPin By Cloe Einam On Referencing Harvard Referencing, Essay, Essa
Pin By Cloe Einam On Referencing Harvard Referencing, Essay, EssaAngie Miller
Ā 
Pin Von Carmen Perez De La Cruz Auf German-BRIEF,
Pin Von Carmen Perez De La Cruz Auf German-BRIEF,Pin Von Carmen Perez De La Cruz Auf German-BRIEF,
Pin Von Carmen Perez De La Cruz Auf German-BRIEF,Angie Miller
Ā 
Powerful Quotes To Start Essays. QuotesGram
Powerful Quotes To Start Essays. QuotesGramPowerful Quotes To Start Essays. QuotesGram
Powerful Quotes To Start Essays. QuotesGramAngie Miller
Ā 
Can Essay Writing Services Be Trusted - UK Writing Experts Blog
Can Essay Writing Services Be Trusted - UK Writing Experts BlogCan Essay Writing Services Be Trusted - UK Writing Experts Blog
Can Essay Writing Services Be Trusted - UK Writing Experts BlogAngie Miller
Ā 
The SmARTteacher Resource Writing An Essa
The SmARTteacher Resource Writing An EssaThe SmARTteacher Resource Writing An Essa
The SmARTteacher Resource Writing An EssaAngie Miller
Ā 
Order Paper Writing Help 24
Order Paper Writing Help 24Order Paper Writing Help 24
Order Paper Writing Help 24Angie Miller
Ā 
How To Format A College Application Essay
How To Format A College Application EssayHow To Format A College Application Essay
How To Format A College Application EssayAngie Miller
Ā 
Thanksgiving Printable Worksheets Colorful Fall,
Thanksgiving Printable Worksheets Colorful Fall,Thanksgiving Printable Worksheets Colorful Fall,
Thanksgiving Printable Worksheets Colorful Fall,Angie Miller
Ā 
Writing Paper, Notebook Paper, , (2)
Writing Paper, Notebook Paper, ,  (2)Writing Paper, Notebook Paper, ,  (2)
Writing Paper, Notebook Paper, , (2)Angie Miller
Ā 

More from Angie Miller (20)

Writing Poetry In The Upper Grades Poetry Lessons,
Writing Poetry In The Upper Grades Poetry Lessons,Writing Poetry In The Upper Grades Poetry Lessons,
Writing Poetry In The Upper Grades Poetry Lessons,
Ā 
ReMarkable 2 Is A 10.3-Inch E-Paper Tablet With A Stylus, Starts At
ReMarkable 2 Is A 10.3-Inch E-Paper Tablet With A Stylus, Starts AtReMarkable 2 Is A 10.3-Inch E-Paper Tablet With A Stylus, Starts At
ReMarkable 2 Is A 10.3-Inch E-Paper Tablet With A Stylus, Starts At
Ā 
Printable Lined Paper For Kids That Are Soft Harper Blog
Printable Lined Paper For Kids That Are Soft Harper BlogPrintable Lined Paper For Kids That Are Soft Harper Blog
Printable Lined Paper For Kids That Are Soft Harper Blog
Ā 
Writing Your Introduction, Transitions, And Conclusion
Writing Your Introduction, Transitions, And ConclusionWriting Your Introduction, Transitions, And Conclusion
Writing Your Introduction, Transitions, And Conclusion
Ā 
Groundhog Day Writing Paper
Groundhog Day Writing PaperGroundhog Day Writing Paper
Groundhog Day Writing Paper
Ā 
5 Writing Tips To Help Overcome Anxiety Youn
5 Writing Tips To Help Overcome Anxiety Youn5 Writing Tips To Help Overcome Anxiety Youn
5 Writing Tips To Help Overcome Anxiety Youn
Ā 
How To Write An Essay In 6 Simple Steps ScoolWork
How To Write An Essay In 6 Simple Steps ScoolWorkHow To Write An Essay In 6 Simple Steps ScoolWork
How To Write An Essay In 6 Simple Steps ScoolWork
Ā 
Scroll Paper - Cliparts.Co
Scroll Paper - Cliparts.CoScroll Paper - Cliparts.Co
Scroll Paper - Cliparts.Co
Ā 
Hnh Nh Bn, S Tay, Vit, Cng Vic, Ang Lm Vic, Sch, Ngi
Hnh Nh Bn, S Tay, Vit, Cng Vic, Ang Lm Vic, Sch, NgiHnh Nh Bn, S Tay, Vit, Cng Vic, Ang Lm Vic, Sch, Ngi
Hnh Nh Bn, S Tay, Vit, Cng Vic, Ang Lm Vic, Sch, Ngi
Ā 
Recycling Essay Essay On Re
Recycling Essay Essay On ReRecycling Essay Essay On Re
Recycling Essay Essay On Re
Ā 
Pin On PAPER SHEETS
Pin On PAPER SHEETSPin On PAPER SHEETS
Pin On PAPER SHEETS
Ā 
Pin By Cloe Einam On Referencing Harvard Referencing, Essay, Essa
Pin By Cloe Einam On Referencing Harvard Referencing, Essay, EssaPin By Cloe Einam On Referencing Harvard Referencing, Essay, Essa
Pin By Cloe Einam On Referencing Harvard Referencing, Essay, Essa
Ā 
Pin Von Carmen Perez De La Cruz Auf German-BRIEF,
Pin Von Carmen Perez De La Cruz Auf German-BRIEF,Pin Von Carmen Perez De La Cruz Auf German-BRIEF,
Pin Von Carmen Perez De La Cruz Auf German-BRIEF,
Ā 
Powerful Quotes To Start Essays. QuotesGram
Powerful Quotes To Start Essays. QuotesGramPowerful Quotes To Start Essays. QuotesGram
Powerful Quotes To Start Essays. QuotesGram
Ā 
Can Essay Writing Services Be Trusted - UK Writing Experts Blog
Can Essay Writing Services Be Trusted - UK Writing Experts BlogCan Essay Writing Services Be Trusted - UK Writing Experts Blog
Can Essay Writing Services Be Trusted - UK Writing Experts Blog
Ā 
The SmARTteacher Resource Writing An Essa
The SmARTteacher Resource Writing An EssaThe SmARTteacher Resource Writing An Essa
The SmARTteacher Resource Writing An Essa
Ā 
Order Paper Writing Help 24
Order Paper Writing Help 24Order Paper Writing Help 24
Order Paper Writing Help 24
Ā 
How To Format A College Application Essay
How To Format A College Application EssayHow To Format A College Application Essay
How To Format A College Application Essay
Ā 
Thanksgiving Printable Worksheets Colorful Fall,
Thanksgiving Printable Worksheets Colorful Fall,Thanksgiving Printable Worksheets Colorful Fall,
Thanksgiving Printable Worksheets Colorful Fall,
Ā 
Writing Paper, Notebook Paper, , (2)
Writing Paper, Notebook Paper, ,  (2)Writing Paper, Notebook Paper, ,  (2)
Writing Paper, Notebook Paper, , (2)
Ā 

Recently uploaded

Introduction to Nonprofit Accounting: The Basics
Introduction to Nonprofit Accounting: The BasicsIntroduction to Nonprofit Accounting: The Basics
Introduction to Nonprofit Accounting: The BasicsTechSoup
Ā 
Ecological Succession. ( ECOSYSTEM, B. Pharmacy, 1st Year, Sem-II, Environmen...
Ecological Succession. ( ECOSYSTEM, B. Pharmacy, 1st Year, Sem-II, Environmen...Ecological Succession. ( ECOSYSTEM, B. Pharmacy, 1st Year, Sem-II, Environmen...
Ecological Succession. ( ECOSYSTEM, B. Pharmacy, 1st Year, Sem-II, Environmen...Shubhangi Sonawane
Ā 
Web & Social Media Analytics Previous Year Question Paper.pdf
Web & Social Media Analytics Previous Year Question Paper.pdfWeb & Social Media Analytics Previous Year Question Paper.pdf
Web & Social Media Analytics Previous Year Question Paper.pdfJayanti Pande
Ā 
Basic Civil Engineering first year Notes- Chapter 4 Building.pptx
Basic Civil Engineering first year Notes- Chapter 4 Building.pptxBasic Civil Engineering first year Notes- Chapter 4 Building.pptx
Basic Civil Engineering first year Notes- Chapter 4 Building.pptxDenish Jangid
Ā 
How to Give a Domain for a Field in Odoo 17
How to Give a Domain for a Field in Odoo 17How to Give a Domain for a Field in Odoo 17
How to Give a Domain for a Field in Odoo 17Celine George
Ā 
Advanced Views - Calendar View in Odoo 17
Advanced Views - Calendar View in Odoo 17Advanced Views - Calendar View in Odoo 17
Advanced Views - Calendar View in Odoo 17Celine George
Ā 
Presentation by Andreas Schleicher Tackling the School Absenteeism Crisis 30 ...
Presentation by Andreas Schleicher Tackling the School Absenteeism Crisis 30 ...Presentation by Andreas Schleicher Tackling the School Absenteeism Crisis 30 ...
Presentation by Andreas Schleicher Tackling the School Absenteeism Crisis 30 ...EduSkills OECD
Ā 
PROCESS RECORDING FORMAT.docx
PROCESS      RECORDING        FORMAT.docxPROCESS      RECORDING        FORMAT.docx
PROCESS RECORDING FORMAT.docxPoojaSen20
Ā 
Food Chain and Food Web (Ecosystem) EVS, B. Pharmacy 1st Year, Sem-II
Food Chain and Food Web (Ecosystem) EVS, B. Pharmacy 1st Year, Sem-IIFood Chain and Food Web (Ecosystem) EVS, B. Pharmacy 1st Year, Sem-II
Food Chain and Food Web (Ecosystem) EVS, B. Pharmacy 1st Year, Sem-IIShubhangi Sonawane
Ā 
Unit-IV- Pharma. Marketing Channels.pptx
Unit-IV- Pharma. Marketing Channels.pptxUnit-IV- Pharma. Marketing Channels.pptx
Unit-IV- Pharma. Marketing Channels.pptxVishalSingh1417
Ā 
ICT Role in 21st Century Education & its Challenges.pptx
ICT Role in 21st Century Education & its Challenges.pptxICT Role in 21st Century Education & its Challenges.pptx
ICT Role in 21st Century Education & its Challenges.pptxAreebaZafar22
Ā 
On National Teacher Day, meet the 2024-25 Kenan Fellows
On National Teacher Day, meet the 2024-25 Kenan FellowsOn National Teacher Day, meet the 2024-25 Kenan Fellows
On National Teacher Day, meet the 2024-25 Kenan FellowsMebane Rash
Ā 
Russian Escort Service in Delhi 11k Hotel Foreigner Russian Call Girls in Delhi
Russian Escort Service in Delhi 11k Hotel Foreigner Russian Call Girls in DelhiRussian Escort Service in Delhi 11k Hotel Foreigner Russian Call Girls in Delhi
Russian Escort Service in Delhi 11k Hotel Foreigner Russian Call Girls in Delhikauryashika82
Ā 
Role Of Transgenic Animal In Target Validation-1.pptx
Role Of Transgenic Animal In Target Validation-1.pptxRole Of Transgenic Animal In Target Validation-1.pptx
Role Of Transgenic Animal In Target Validation-1.pptxNikitaBankoti2
Ā 
ICT role in 21st century education and it's challenges.
ICT role in 21st century education and it's challenges.ICT role in 21st century education and it's challenges.
ICT role in 21st century education and it's challenges.MaryamAhmad92
Ā 
Unit-IV; Professional Sales Representative (PSR).pptx
Unit-IV; Professional Sales Representative (PSR).pptxUnit-IV; Professional Sales Representative (PSR).pptx
Unit-IV; Professional Sales Representative (PSR).pptxVishalSingh1417
Ā 
Seal of Good Local Governance (SGLG) 2024Final.pptx
Seal of Good Local Governance (SGLG) 2024Final.pptxSeal of Good Local Governance (SGLG) 2024Final.pptx
Seal of Good Local Governance (SGLG) 2024Final.pptxnegromaestrong
Ā 
General Principles of Intellectual Property: Concepts of Intellectual Proper...
General Principles of Intellectual Property: Concepts of Intellectual  Proper...General Principles of Intellectual Property: Concepts of Intellectual  Proper...
General Principles of Intellectual Property: Concepts of Intellectual Proper...Poonam Aher Patil
Ā 
Beyond the EU: DORA and NIS 2 Directive's Global Impact
Beyond the EU: DORA and NIS 2 Directive's Global ImpactBeyond the EU: DORA and NIS 2 Directive's Global Impact
Beyond the EU: DORA and NIS 2 Directive's Global ImpactPECB
Ā 

Recently uploaded (20)

Introduction to Nonprofit Accounting: The Basics
Introduction to Nonprofit Accounting: The BasicsIntroduction to Nonprofit Accounting: The Basics
Introduction to Nonprofit Accounting: The Basics
Ā 
Ecological Succession. ( ECOSYSTEM, B. Pharmacy, 1st Year, Sem-II, Environmen...
Ecological Succession. ( ECOSYSTEM, B. Pharmacy, 1st Year, Sem-II, Environmen...Ecological Succession. ( ECOSYSTEM, B. Pharmacy, 1st Year, Sem-II, Environmen...
Ecological Succession. ( ECOSYSTEM, B. Pharmacy, 1st Year, Sem-II, Environmen...
Ā 
Web & Social Media Analytics Previous Year Question Paper.pdf
Web & Social Media Analytics Previous Year Question Paper.pdfWeb & Social Media Analytics Previous Year Question Paper.pdf
Web & Social Media Analytics Previous Year Question Paper.pdf
Ā 
Basic Civil Engineering first year Notes- Chapter 4 Building.pptx
Basic Civil Engineering first year Notes- Chapter 4 Building.pptxBasic Civil Engineering first year Notes- Chapter 4 Building.pptx
Basic Civil Engineering first year Notes- Chapter 4 Building.pptx
Ā 
How to Give a Domain for a Field in Odoo 17
How to Give a Domain for a Field in Odoo 17How to Give a Domain for a Field in Odoo 17
How to Give a Domain for a Field in Odoo 17
Ā 
Advanced Views - Calendar View in Odoo 17
Advanced Views - Calendar View in Odoo 17Advanced Views - Calendar View in Odoo 17
Advanced Views - Calendar View in Odoo 17
Ā 
Presentation by Andreas Schleicher Tackling the School Absenteeism Crisis 30 ...
Presentation by Andreas Schleicher Tackling the School Absenteeism Crisis 30 ...Presentation by Andreas Schleicher Tackling the School Absenteeism Crisis 30 ...
Presentation by Andreas Schleicher Tackling the School Absenteeism Crisis 30 ...
Ā 
PROCESS RECORDING FORMAT.docx
PROCESS      RECORDING        FORMAT.docxPROCESS      RECORDING        FORMAT.docx
PROCESS RECORDING FORMAT.docx
Ā 
Food Chain and Food Web (Ecosystem) EVS, B. Pharmacy 1st Year, Sem-II
Food Chain and Food Web (Ecosystem) EVS, B. Pharmacy 1st Year, Sem-IIFood Chain and Food Web (Ecosystem) EVS, B. Pharmacy 1st Year, Sem-II
Food Chain and Food Web (Ecosystem) EVS, B. Pharmacy 1st Year, Sem-II
Ā 
Unit-IV- Pharma. Marketing Channels.pptx
Unit-IV- Pharma. Marketing Channels.pptxUnit-IV- Pharma. Marketing Channels.pptx
Unit-IV- Pharma. Marketing Channels.pptx
Ā 
ICT Role in 21st Century Education & its Challenges.pptx
ICT Role in 21st Century Education & its Challenges.pptxICT Role in 21st Century Education & its Challenges.pptx
ICT Role in 21st Century Education & its Challenges.pptx
Ā 
On National Teacher Day, meet the 2024-25 Kenan Fellows
On National Teacher Day, meet the 2024-25 Kenan FellowsOn National Teacher Day, meet the 2024-25 Kenan Fellows
On National Teacher Day, meet the 2024-25 Kenan Fellows
Ā 
Russian Escort Service in Delhi 11k Hotel Foreigner Russian Call Girls in Delhi
Russian Escort Service in Delhi 11k Hotel Foreigner Russian Call Girls in DelhiRussian Escort Service in Delhi 11k Hotel Foreigner Russian Call Girls in Delhi
Russian Escort Service in Delhi 11k Hotel Foreigner Russian Call Girls in Delhi
Ā 
Mehran University Newsletter Vol-X, Issue-I, 2024
Mehran University Newsletter Vol-X, Issue-I, 2024Mehran University Newsletter Vol-X, Issue-I, 2024
Mehran University Newsletter Vol-X, Issue-I, 2024
Ā 
Role Of Transgenic Animal In Target Validation-1.pptx
Role Of Transgenic Animal In Target Validation-1.pptxRole Of Transgenic Animal In Target Validation-1.pptx
Role Of Transgenic Animal In Target Validation-1.pptx
Ā 
ICT role in 21st century education and it's challenges.
ICT role in 21st century education and it's challenges.ICT role in 21st century education and it's challenges.
ICT role in 21st century education and it's challenges.
Ā 
Unit-IV; Professional Sales Representative (PSR).pptx
Unit-IV; Professional Sales Representative (PSR).pptxUnit-IV; Professional Sales Representative (PSR).pptx
Unit-IV; Professional Sales Representative (PSR).pptx
Ā 
Seal of Good Local Governance (SGLG) 2024Final.pptx
Seal of Good Local Governance (SGLG) 2024Final.pptxSeal of Good Local Governance (SGLG) 2024Final.pptx
Seal of Good Local Governance (SGLG) 2024Final.pptx
Ā 
General Principles of Intellectual Property: Concepts of Intellectual Proper...
General Principles of Intellectual Property: Concepts of Intellectual  Proper...General Principles of Intellectual Property: Concepts of Intellectual  Proper...
General Principles of Intellectual Property: Concepts of Intellectual Proper...
Ā 
Beyond the EU: DORA and NIS 2 Directive's Global Impact
Beyond the EU: DORA and NIS 2 Directive's Global ImpactBeyond the EU: DORA and NIS 2 Directive's Global Impact
Beyond the EU: DORA and NIS 2 Directive's Global Impact
Ā 

An Overview Of Treatment Strategies For Hutchinson-Gilford Progeria Syndrome

  • 1. HAL Id: hal-01774301 https://hal.archives-ouvertes.fr/hal-01774301 Submitted on 13 Nov 2018 HAL is a multi-disciplinary open access archive for the deposit and dissemination of sci- entific research documents, whether they are pub- lished or not. The documents may come from teaching and research institutions in France or abroad, or from public or private research centers. Lā€™archive ouverte pluridisciplinaire HAL, est destinĆ©e au dĆ©pĆ“t et Ć  la diffusion de documents scientifiques de niveau recherche, publiĆ©s ou non, Ć©manant des Ć©tablissements dā€™enseignement et de recherche franƧais ou Ć©trangers, des laboratoires publics ou privĆ©s. An overview of treatment strategies for Hutchinson-Gilford Progeria Syndrome Karim Harhouri, Diane Frankel, Catherine Bartoli, Patrice Roll, Annachiara de Sandre-Giovannoli, Nicolas LĆ©vy To cite this version: Karim Harhouri, Diane Frankel, Catherine Bartoli, Patrice Roll, Annachiara de Sandre-Giovannoli, et al.. An overview of treatment strategies for Hutchinson-Gilford Progeria Syndrome. Nucleus, European Nuclear Society, 2018, 9 (1), pp.265-276. ąø€10.1080/19491034.2018.1460045ąø€. ąø€hal-01774301ąø€
  • 2. Full Terms & Conditions of access and use can be found at http://www.tandfonline.com/action/journalInformation?journalCode=kncl20 Nucleus ISSN: 1949-1034 (Print) 1949-1042 (Online) Journal homepage: http://www.tandfonline.com/loi/kncl20 An overview of treatment strategies for Hutchinson-Gilford Progeria syndrome Karim Harhouri, Diane Frankel, Catherine Bartoli, Patrice Roll, Annachiara De Sandre-Giovannoli & Nicolas LĆ©vy To cite this article: Karim Harhouri, Diane Frankel, Catherine Bartoli, Patrice Roll, Annachiara De Sandre-Giovannoli & Nicolas LĆ©vy (2018) An overview of treatment strategies for Hutchinson- Gilford Progeria syndrome, Nucleus, 9:1, 265-276, DOI: 10.1080/19491034.2018.1460045 To link to this article: https://doi.org/10.1080/19491034.2018.1460045 Ā© 2018 The Author(s). Published by Informa UK Limited, trading as Taylor & Francis Group Accepted author version posted online: 05 Apr 2018. Published online: 05 Apr 2018. Submit your article to this journal Article views: 1423 View Crossmark data Citing articles: 3 View citing articles
  • 3. An overview of treatment strategies for Hutchinson-Gilford Progeria syndrome Karim Harhouri a , Diane Frankel a,b , Catherine Bartolia , Patrice Roll a,b , Annachiara De Sandre-Giovannoli a,c and Nicolas L evy a,c a Aix Marseille Univ, INSERM, MMG ā€“ U1251, Marseille, France; b APHM, H^ opital la Timone, Service de Biologie Cellulaire, Marseille, France; c APHM, H^ opital la Timone, D epartement de G en etique M edicale, Marseille, France ARTICLE HISTORY Received 16 January 2018 Revised 21 March 2018 Accepted 27 March 2018 ABSTRACT Hutchinson-Gilford progeria syndrome (HGPS) is a sporadic, autosomal dominant disorder characterized by premature and accelerated aging symptoms leading to death at the mean age of 14.6 years usually due to cardiovascular complications. HGPS is caused by a de novo point mutation in the LMNA gene encoding the intermediate ļ¬lament proteins lamins A and C which are structural components of the nuclear lamina. This mutation leads to the production of a truncated toxic form of lamin A, issued from aberrant splicing and called progerin. Progerin accumulates in HGPS cellsā€™ nuclei and is a hallmark of the disease. Small amounts of progerin are also produced during normal aging. HGPS cells and animal preclinical models have provided insights into the molecular and cellular pathways that underlie the disease and have also highlighted possible mechanisms involved in normal aging. This review reports recent medical advances and treatment approaches for patients affected with HGPS. KEYWORDS HGPS; Progerin; FTI; ZOPRA; AON; Rapamycin; Metformin; MG132 Introduction Hutchinson-Gilford progeria syndrome (HGPS; OMIM #176670) is a rare genetic disorder which affects 1 in 4ā€“8 million children with symptoms resembling physio- logical aging that include growth impairment, very thin skin, loss of subcutaneous fat, alopecia, osteoporosis and heart disease leading to shortened life span and death at about 14.6 years [1,2]. HGPS was ļ¬rst described at the end of the XIX century by Jonathan Hutchinson and Hastings Gilford [3,4]. It was only more than 100 years later, in 2003, that the heterozy- gous, de novo mutation c.1824CT, p.G608G (NM_170707.3) responsible for this accelerated-aging disease was found to be located within exon 11 of the LMNA gene that encodes lamins A and C [5,6]. Lamin A is a nuclear protein belonging to type V intermediate ļ¬laments. It is synthetized as a precursor called prela- min A. Prelamin A undergoes a multistep post-transla- tional processing, including cysteine farnesylation by farnesyl transferase (FTase) on its C-terminal CaaX motif, then cleavage of the remaining 3 amino acids by the metallopeptidase ZMPSTE24. The C-terminal cyste- ine is then carboxymethylated by the isoprenylcysteine carboxylmethyltransferase (ICMT). Finally, the last 15 amino acids are cleaved again by ZMPSTE24 to pro- duce the unfarnesylated, mature lamin A. HGPS muta- tion activates a cryptic donor splice site in exon 11 of the LMNA gene that leads to deletion of 50 amino acids near the C terminus, abrogating the second ZMPSTE24 cleavage site and resulting in accumulation of a trun- cated and permanently farnesylated prelamin A called progerin that incorporates abnormally into the nuclear lamina and exerts multiple toxic effects [7]. At the cellu- lar level, HGPS is characterized by dramatic defects in nuclear envelope structure and function [8,9]. Primary ļ¬broblasts from HGPS patients exhibit reduced prolifer- ation as well as premature senescence [10], impaired DNA repair mechanisms [2,11ā€“13], increased reactive oxygen species production [14], mitochondrial dysfunc- tion [15], loss of peripheral heterochromatin [16ā€“18] and telomere attrition [18]. Through these alterations, progerin accumulation results in loss of peripheral het- erochromatin caused by the decrease in the repressive histone marks H3K9me3, H3K27me3 and H3K27 methyltransferase EZH2, increase in H4K20me3 [9,16,19], reduced levels of heterochromatin protein 1 CONTACT Nicolas L evy Nicolas.Levy@univ-amu.fr INSERM, U 1251, Marseille Medical Genetics, Facult e de M edecine de la Timone, 27 Boulevard Jean Moulin, Marseille Cedex 05, 13385, France. Ā© 2018 The Author(s). Published by Informa UK Limited, trading as Taylor Francis Group This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial License ( http://creativecommons.org/licenses/by-nc/4.0/), which per- mits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited. NUCLEUS 2018, VOL. 9, NO. 1, 265ā€“276 https://doi.org/10.1080/19491034.2018.1460045 REVIEW
  • 4. (HP1) [20] and Lap2a [9]. Progerin also impairs the formation of DNA repair foci due to recruitment deļ¬- ciency of the DNA double-strand break (DSB) repair factors p53-binding protein 1 (53BP1), Rad50 and Rad51 at DNA damage sites [21,22]. On the other hand, altered signaling pathways have been described in HGPS cells [23]. Among them, altered extracellular matrix synthesis caused by disturbed Wnt/b-catenin signaling [24], affected Notch signaling [20], hyperacti- vation of NF-Kb in response to inļ¬‚ammation [25] and impaired NRF2 (Nuclear factor erythroid-2-Related Fac- tor 2) transcriptional activity resulting in increased chronic oxidative stress [26]. A wide spectrum of treatment strategies, targeting several processes with different speciļ¬cities, has been proposed to correct the defects in HGPS: (i) to directly ā€œrepairā€ the disease -causing mutation; (ii) to inhibit pre-mRNA aberrant splicing leading to progerin mRNA production; (iii) to decrease the toxicity of iso- prenylated and methylated progerin; (iv) to induce progerin clearance; (v) to decrease the noxious down- stream effects linked to progerin accumulation [27] (Figure 1). These approaches will be presented below and grouped following their individual action. Prelamin A Isoprenylation and methylation inhibitors: Lonafarnib, Zoledronate/Pravastatin, Monoaminopyrimidines and isoprenylcysteine carboxyl methyltransferase inhibitor The aberrant splice event that gives rise to progerin leads to the deletion of the ZMPSTE24 cleavage site normally used to remove the farnesylated carboxy ter- minus from prelamin A during posttranslational proc- essing. Consequently, permanently farnesylated progerin remains anchored to the inner nuclear membrane resulting in dominant-negative disruption of the nuclear scaffold upon progerin dimerization with wild-type lamins [28]. Knowledge of these steps predicted that blocking farnesylation using farnesyltransferase inhibitor (FTI) drugs would decrease progerin production and toxicity. FTI are small molecules which reversibly bind to the farnesyltransferase CAAX binding site [29]. Blocking farnesylation of progerin with FTIs restored normal nuclear architecture and resulted in signiļ¬cant reductions in nuclear blebbing both in transiently trans- fected HeLa, HEK 293, NIH 3T3 cells and human HGPS ļ¬broblasts [8,30ā€“33]. In transgenic HGPS murine models treated with FTIs, bone mineralization, and weight are improved, lifespan is extended [34,35] and cardiovascular defects are prevented [36]. In 2007, the above studies led to the initiation of a prospective sin- gle-arm clinical trial (ClinicalTrials.gov, NCT00425607), using an FTI called lonafarnib, which was originally developed for the treatment of cancer. A cohort of 25 HGPS patients between 3 and 16 years of age were included in this trial and received lonafarnib for a mini- mum of 2 years. In 2012, researchers reported that some children with HGPS receiving lonafarnib showed a modest improvement in weight gain: nine patientsā€™ rates of weight gain increased by 50%, six patientsā€™ rates of weight gain decreased by 50%, and 10 remained stable with respect to the rate of weight gain. Other results from this trial showed improvements in vascular stiffness including decreases in arterial pulse wave velocity by a median of 35% in 18 subjects, in addition to increases in skeletal rigidity (median percent increases by 40ā€“50% in axial rigidity, 170ā€“228% in ļ¬‚ex- ural rigidity, and 167ā€“229% in torsional rigidity) as well as sensorineural hearing and bone mineral density (3% increase at one or more sites in 76% of children compared with 40% of the participants who exhibited decreases at one or more sites) [37]. Although FTI treat- ment increased mean survival by 1.6 years [38] it was reported that progerin may become alternatively preny- lated by geranylgeranyltransferase I when farnesyltrans- ferases are inhibited; indeed, the simultaneous presence of both farnesyltransferase inhibitor (FTI-277) and Ger- anylGeranylTransferase I inhibitor (GGTI-2147) led to a substantial amount of prelamin A accumulation com- pared with the effect of each inhibitor alone [39], thereby explaining the limited beneļ¬cial effects of FTI monotherapy. Accordingly, authors hypothesized that blocking both protein farnesylation and geranylgeranyla- tion could minimize the possibility of alternative preny- lation events that confer resistance to FTIs. Acting on the synthetic pathway of farnesyl pyrophosphate, a co- substrate of farnesyltransferase and a precursor of gera- nylgeranyl pyrophosphate, we and our close collaborator Carlos Lopez-Otin (Spain) demonstrated the synergistic effect of a combination of ZOledronate (N-BisPhospho- nate) and PRAvastatin (statin) (ZOPRA) and their effec- tiveness to reduce prenylation and rescue HGPS cells defects and the progeroid phenotypes of Zmpste24Ā”/Ā” mice, including improvement of growth retardation, loss of weight, lipodystrophy, hair loss and bone defects. Likewise, the longevity of these mice was substantially extended [39]. This preclinical work allowed our clinical 266 K. HARHOURI ET AL.
  • 5. team to design and conduct a phase II, monocentric, open-label, single arm clinical trial (ClinicalTrials.gov, NCT00731016) using ZOPRA to evaluate its safety and efļ¬cacy in 12 HGPS patients included for 3.5 years each, with partially positive results including improve- ment of weight gain and bone density defects, without severe adverse effects (De Sandre Giovannoli, Sigaudy et al, submitted). Although trials using FTIs (Lonafar- nib) and ZOPRA showed some efļ¬cacy for given parameters, these drugs could not be considered as a cure, and further research was needed to implement more effective therapeutic approaches for patients. A single-arm triple therapy trial (ClinicalTrials.gov, NCT00879034) was designed to administer pravastatin, zoledronate and lonafarnib and sought to further improve disease parameters by additionally inhibiting progerin prenylation, given that each enzyme functions along the protein prenylation pathway. The results of this trial on 37 participants with HGPS which began in 2009 in Boston revealed additional bone mineral density beneļ¬t but no major beneļ¬t beyond that seen with lona- farnib alone [40]. However, caution should be taken when consider- ing the long-term treatment effects of FTIs, as FTIs have been shown to cause lethal cardiomyopathy in mouse models of progeria because of non-farnesylated prelamin A accumulation [41]. On the other hand, among 21.608 small molecules tested in induced plu- ripotent stem cell (iPSC) lines derived from HGPS patients, Nissan and colleagues identiļ¬ed several com- pounds, called monoaminopyrimidines (mono-APs), as new modulators of farnesylation, resulting, in vitro, Figure 1. Speciļ¬city of a wide spectrum of treatment strategies for progeria. Several therapeutic strategies for progeria are shown. In decreasing order for their target speciļ¬city, treatment strategies can be mentioned as follows: the mutation-causing disease repair by genome editing (CRISPR as a future possibility to be evaluated), the inhibition of pre-mRNA aberrant splicing (Antisense OligoNucleoti- des: AONs, Metformin, MG132), the reduction of isoprenylated and methylated progerin levels (Farnesyl transferase inhibitors: FTIs, ZOledronate-PRAvastatin: ZOPRA, Monoaminopyrimidines: Mono-APs, Isoprenylcysteine CarboxyMethyl Transferase (ICMT) inhibitor, GeranylGeranylTransferase Inhibitor: GGTI-2147), the induction of progerin clearance (Rapamycin, Sulforaphane, Retinoids, MG132) and the reduction of the noxious downstream effects due to progerin accumulation (Rock inhibitor: Y-27632, Nuclear factor erythroid-2- Related Factor 2: NRF2 reactivation, Methylene blue, sodium salicylate, DOT1L inhibitor: epz-4777, Remodelin, Vitamin D, Resveratrol, N-Acetyl-Cysteine: NAC, inorganic pyrophosphate: PPi and JH4). NUCLEUS 267
  • 6. in improved phenotypes associated with HGPS [42]. This class of protein farnesylation inhibitors, targeting both farnesyl pyrophosphate synthase and farnesyl transferase, acts like a combination of FTIs and bisphosphonates. In addition to isoprenylation, another consequence of defective posttranslational modiļ¬cation to prelamin A in progeria was reported implicating the carboxy methylation that is mediated by isoprenylcysteine car- boxyl methyltransferase (ICMT). In this context, Ibra- him and colleagues revealed that, in vitro treatment with ICMT inhibitor, increased proliferation and delayed senescence in human HGPS ļ¬broblasts. Fur- thermore, ICMT inhibition by lentiviral short hairpin RNAs (shICMT) increased body weight, normalized grip strength, as well as prevented bone fractures and death in Zmpste24-deļ¬cient mice [43]. Autophagy-activating drugs: Rapamycin, Sulforaphane, Retinoids and MG132 Rapamycin, an immunosuppressive agent that is used to prevent the rejection of transplanted organs, pro- moted progerin clearance through autophagy, improved the abnormal nuclear shape, delayed the onset of cellular senescence of HGPS ļ¬broblasts [44,45] and rescued the chromatin phenotype of cul- tured ļ¬broblasts, including histone methylation status and BAF and LAP2alpha distribution patterns [46]. Otherwise, studies on LmnaĀ”/Ā” mouse model for dilated cardiomyopathy and muscular dystrophy, showed that pharmacologic reversal of elevated mTORC1 signaling by rapamycin improves cardiac and skeletal muscle function and enhances survival [47]. Using the same mouse model, Liao et al. showed that rapamycin treatment results in lifespan extension associated with increased body weight and fat content [48]. Although these results show beneļ¬cial effects on lmna KO mice, this model does not mimic the genet- ics and pathophysiology of HGPS in patients. A phase I/II monocentric trial (NCT02579044) of Everolimus, an agent derived from rapamycin, in combination with Lonafarnib, is currently conducted at the Clinical and Translational Study Unit (CTSU) of Boston Childrenā€™s Hospital. Rapamycin inhibits the activity of mTOR (Mammalian Target Of Rapamycin), known to regulate a large panel of cellular functions including protein synthesis, cell growth, cytoskeleton rearrange- ments, transcription, immune responses or autophagy. Therefore, much caution should be used while per- forming the direct translation of the in vitro results to children affected with progeria. Furthermore, since rapamycin is also known to inhibit adipogenesis [49,50], precautions should be taken when using this compound in HGPS patients who are affected with generalized lipoatrophy and lipodystrophy. Sulforaphane, an antioxidant derived from crucifer- ous vegetables, has been also described to enhance pro- gerin clearance by autophagy and to reverse the cellular hallmarks of HGPS in vitro [51]. Intermittent treatment with Sulforaphane and Lonafarnib separately rescued the HGPS cellular phenotype [52]. Furthermore, since the LMNA promoter contains a retinoic acid responsive element, two recent studies suggested that either reti- noids alone [53] or in a combination with rapamycin [54] reduce the amount of progerin and reverse aging defects in HGPS patient skin ļ¬broblasts. These drugs may be beneļ¬cial in progeria treatment, but in vivo extensive studies should be performed before translating them into clinical trials. We recently showed that progerin is sequestered into abnormally shaped Promyelocytic-Nuclear Bodies (PML-NB), identiļ¬ed as novel biomarkers in progeria. We identiļ¬ed MG132 as being effective on progerin degradation. Indeed, MG132 induces progerin nucleocy- toplasmic translocation after a transition through the nucleolus, and progerin clearance through macroau- tophagy in HGPS patient ļ¬broblasts as well as in HGPS patient iPSC-derived mesenchymal stem cells (MSCs) and Vascular Smooth Muscle Cells (VSMCs). MG132 treatment improves cellular HGPS phenotypes, reduces cellular senescence and enhances viability and prolifera- tion in HGPS ļ¬broblasts. In vivo, through MG132 treat- ment, progerin expression decreases in skeletal muscle from LmnaG609G/G609G mice. Altogether, we demonstrate progerin reduction based on MG132 action and shed light on a promising class of molecules towards a poten- tial therapy for children with HGPS [55]. The observed speciļ¬c effect of MG132 on progerin downregulation, sparing normal A-type lamins, further supports the idea that besides typical Progeria, other prelamin A-associ- ated diseases might beneļ¬t from the same treatment. Downregulation of Prelamin A aberrant splicing: Antisense oligonucleotides, Metformin and MG132 The efļ¬ciency of an antisense therapeutic approach using morpholino antisense oligonucleotides (AON) 268 K. HARHOURI ET AL.
  • 7. in sterically blocking the aberrant LMNA splicing site leading to progerin production has been previously proven in vitro on human HGPS patientsā€™ cells and in vivo on a knock-in LmnaG609G/G609G mouse model [56]. Indeed, Osorio et al. tested the combined admin- istration of two AONs: ā€œMmEx11ā€ targeting the exon 11 aberrant splice site activated by the progeria muta- tion in order to hamper its use, and ā€œMmEx10ā€, tar- geting the physiological exon 10 splice site, in order to reinforce the action of the ļ¬rst AON, by shifting splic- ing events towards lamin C production. Similar results were obtained by another group on the same LmnaG609G/G609G mouse model and in HGPS ļ¬bro- blasts, but using another antisense oligonucleotide (ASO) that reduces the binding of the splicing factor SRSF-2 to exon 11 LMNA pre-mRNA, conļ¬rming the efļ¬cacy of this approach [57]. Some patients carry other LMNA mutations affect- ing exon 11 splicing and are named ā€œHGPS-likeā€ patients [58]. They also produce Progerin and/or other truncated Prelamin A isoforms (D35 and D90). Recently, we showed that downregulation of progerin and other truncated or wild type Prelamin A isoforms can be achieved via the antisense therapeutic approach either in HGPS-like and Mandibuloacral Dysplasia type B (MAD-B) patientsā€™ cells [59], establishing a preclinical proof of principle for the use of antisense morpholino oligonucleotides in HGPS, HGPS-like and MAD-B syndromes. Nonetheless, the administra- tion to children of the same molecules administered to mice, i.e. vivo-morpholinos, could not be envisaged due to their known toxicity. Therefore, the choice of AON chemistry and route of administration will be particularly important for future therapeutic approaches involving splicing-modulation. In human HGPS primary ļ¬broblasts and mouse LmnaG609G/G609G 56 , the HGPS mutation induces the use of an internal 5ā€™ cryptic splice site within exon 11 of the LMNA pre-mRNA, leading to an aberrant alter- native splicing and production of a truncated form of prelamin A (progerin). In 2011, the RNA-binding protein SRSF-1 (for Serine/arginine-Rich Splicing Fac- tor 1) was shown to favor this aberrant alternative splicing [60]. On the other hand, it has been shown that SRSF-1 expression is transcriptionally regulated by the antidiabetic drug Metformin [61]. Based on these ļ¬ndings, Egesipe et al. demonstrated that Met- formin decreases SRSF-1 and progerin expression in mesenchymal stem cells derived from HGPS induced pluripotent stem cells (HGPS MSCs) and in several other in vitro models of HGPS, i.e., human primary HGPS ļ¬broblasts and LmnaG609G/G609G mouse ļ¬bro- blasts, resulting in improved nuclear shape abnormali- ties and premature osteoblastic differentiation of HGPS MSCs [62,63]. In HGPS ļ¬broblasts and in vivo via intramuscular injections of MG132 in LmnaG609G/G609G mice, we recently demonstrated that, in addition to activating macroautophagy leading to progerin degradation, MG132 strongly reduces progerin production through downregulation of SRSF-1, controlling prelamin A mRNA aberrant splicing [55]. Further in vivo studies, particularly on the LmnaG609G/G609G mouse model, will be needed to evaluate the effects of Metformin and MG132 on HGPS phenotype improvement. Reduction of progerin downstream toxic effects Few approaches have been described to counteract the altered downstream pathways caused by progerin accumulation, including nuclear shape abnormalities, ROS generation, accumulation of oxidized proteins, mitochondrial dysfunction [14,64], cell senescence and NF-kB activation, leading to the secretion of high levels of the proinļ¬‚ammatory cytokines IL-6, CXCL1, and TNF-a [25,65]. Treatment of progeroid ļ¬bro- blasts with the ROS scavenger N-acetyl cysteine (NAC) reduced the levels of un-repairable DSB and improved their growth rates in culture [64]. Similarly, it has also been shown that rho-associated protein kinase (ROCK) regulates mitochondrial ROS genera- tion by modulating the interaction between Rac1b and cytochrome c [66]. Accordingly, in vitro treatment of HGPS ļ¬broblasts with ROCK inhibitor (Y-27632) resulted in decreased ROS levels and induced recovery of mitochondrial function along with a reduction in the frequency of abnormal nuclear morphology and DNA double-strand breaks [66]. Elevated levels of ROS and oxidative stress were also lowered by reacti- vation of NRF2, whose transcriptional activity is impaired in HGPS cells, resulting in improvements of cellular HGPS defects [26]. Interestingly, MG132 seems to be a potentially effective drug to be used in the prevention of oxidative damage in HGPS cells through the activation of the NRF2 signaling pathway [67ā€“69]. Mitochondrial dysfunction was reported in HGPS ļ¬broblasts as well as in HGPS mouse models [70,71]. To rescue mitochondrial defects, Xiong et al. NUCLEUS 269
  • 8. showed that treatment of HGPS cells with Methylene Blue, an antioxidant compound known to stimulate mitochondrial function, improves not only the mito- chondrial morphology and function but also rescues the premature aging phenotypes in HGPS cells includ- ing nuclear morphology, perinuclear heterochromatin loss and corrects misregulated gene expression [72]. In LmnaG609G/+ mice, mitochondrial dysfunction in vascular smooth muscle cells (VSMCs) results in increased tissue-nonspeciļ¬c alkaline phosphatase activity and diminished ATP synthesis. Accordingly, VSMCs have an impaired capacity to synthesize extra- cellular pyrophosphate, a major inhibitor of vascular calciļ¬cation [71]. Using LmnaG609G/G609G , Villa-Bel- losta et al. showed that aortic vascular calciļ¬cation due to defective pyrophosphate production is counter- balanced systemically by inorganic pyrophosphate (PPi) treatment [71]. In order to explore the therapeutic potential of NF- kB inhibition on HGPS disease parameters, Osorio et al. showed that crossing Zmpste24Ā”/Ā” mice with trans- genic mice displaying reduced NF-kB signaling extends longevity and prevents the development of progeroid features. In the same study, and to the same end, the authors also showed that sodium salicylate treatment efļ¬ciently prevents NF-kB activation and associated disease phenotypes in Zmpste24-deļ¬cient mice, while also extending longevity in the LmnaG609G/G609G model [25]. Accordingly, NF-kB activation impairs somatic cell reprogramming in aging by eliciting the reprogramming repressor DOT1L. The identiļ¬cation of this molecular mecha- nism has allowed to translate this information into a therapeutic approach, indeed, DOT1L inhibition by epz-4777 extended longevity and prevented aging- associated alterations in progeroid mice [73]. In the same way, MG132 is also known to inhibit the secre- tion of proinļ¬‚ammatory cytokines and attenuation of IkB degradation, resulting in the abolition of NF-kB activation [74-76]. These ļ¬ndings, together with the fact that other models of normal and HGPS acceler- ated aging show arteriosclerotic lesions with calciļ¬ca- tion and inļ¬‚ammation [77], support the idea that inļ¬‚ammation is a major regulator of the aging process also in progeria and could be improved by several inļ¬‚ammation-reducing treatments, including MG132. Importantly, MG132 was reported to have a signiļ¬- cant preventive and therapeutic effect on cardiovascu- lar and renal injury [67,68,78], oxidative damage [69,79] and on accelerated atherosclerosis in rabbits [80]. These features are exhibited by HGPS patients who might beneļ¬t from the same therapeutic effects. On the other hand, compounds screening showed that Remodelin, by inhibiting the lamina interacting SUN1-associated acetyl-transferase protein NAT10, improves nuclear shape and ļ¬tness of both progeric and lamin A/Cā€“depleted cells, as observed by decreased levels of the DNA double-strand break markers gH2AX and autophosphorylated ATM (ataxia telangiectasia mutated), decreased DNA dam- age signaling and improved chromatin and nucleolar organization [81]. Among the proteins affected by progerin accumula- tion is the vitamin D receptor (VDR) whose levels are reduced in HGPS cells. In addition, VDR knockout mice develop a premature aging phenotype similar to HGPS patients [82,83]. Interestingly, Kreienkamp et al. recently showed that reconstituting VDR signaling via 1a,25-dihydroxyvitamin D3 (1,25D), the active hormonal form of vitamin D, improves HGPS pheno- types, including nuclear morphological abnormalities, DNA repair defects, and premature senescence [84]. Another important target of progerin is lamin A/C. Indeed, progerin shows strong binding afļ¬nity for lamin A/C exerting a negative dominant effect. On this basis, Lee et al. identiļ¬ed new chemicals (JH4) that can block the interaction between progerin and lamin A/C through direct interaction with progerin. Treatment with JH4 alleviated nuclear deformation and reversed senescence and growth arrest markers. Furthermore, administration of JH4 to LmnaG609G/ G609G resulted in a marked improvement of several progeria phenotypes including grip strength, body weight, organ size and cell density, as well as lifespan extension [85]. Resveratrol, a SIRT1 activator that interacts with lamin A [86], has been proposed as an alternative treatment for progeria. Indeed, in the premature aging mouse model Zmpste24Ā”/Ā” [87], due to the negative dominant effect of progerin, SIRT-1 is weakly associ- ated with the nuclear matrix: as a result deacetylase activity is decreased, leading to a rapid depletion of adult stem cells. By increasing SIRT1 interaction with lamin A, Resveratrol rescues adult stem cell decline and alleviates progeroid features in Zmpste24Ā”/Ā” mice [86]. Although this model reproduces the progeroid syndromes phenotype, the underlying molecular mechanism is different from that of progeria. Further 270 K. HARHOURI ET AL.
  • 9. studies, using a model that produces progerin by the same abnormal splicing mechanism observed in humans, as the LmnaG609G/G609G model, will be needed to evaluate the impact of Resveratrol on the disease phenotypes reversion. Another study revealed that resveratrol treatment of an osteoblasts-speciļ¬c progerin-expressing mouse model [88] does not show overall beneļ¬cial effects [89]. Resveratrol efļ¬cacy has been suboptimal due to poor bioavailability of this compound and variable dose-linked effects, limiting the full potential of SIRT1 activation treatment. A novel formulation of micronized resveratrol, SRT501, has been developed with improved bioavailability, tol- erance and signiļ¬cant pharmacologic effect [90]. SRT501 open a new interesting research path on this class of molecules towards the development of new therapeutic approaches for progeria. Recently, an in vivo study showed that partial reprogramming by short-term cyclic expression of Oct4, Sox2, Klf4, and c-Myc (OSKM) ameliorates cel- lular and physiological hallmarks of aging and pro- longs lifespan in LmnaG609G/G609G mice [91,92]. Conclusion Progress in progeria research led to a growing number of promising therapeutic candidates (Figure 2). How- ever, most of these approaches lack sufļ¬cient in vitro and in vivo preclinical data to be transposed to patients. Mostly, evidence of efļ¬cacy in an adapted progeria animal model is required to exclude toxicity and perform tentative determination of optimal doses, dose frequencies, administration routes, and evalua- tion endpoints. A speciļ¬c drug formulation requires advanced preclinical toxicity testing to avoid serious side effects that would prohibit long term use. It is worth to note that, in HGPS preclinical studies, the ability of a drug to preferentially target the cardiovas- cular system should be evaluated, in that it represents the major pathophysiological target in the disorder, leading to premature death. Likewise, among treat- ment efļ¬cacy readouts, cardiovascular measures would be preferred. Increasing our understanding of the disease biology will also be important to further elucidate which impaired pathways are most relevant to the disease, in order to identify both other treat- ment pathophysiological targets and other outcome measures. A limiting factor in advancing the develop- ment of therapeutic approaches for progeria is that studies are performed on primary cultures of patientā€™s cells or animal models due to the limited number of autopsy specimens from HGPS patients. To reproduce key features of HGPS, iPSCs [93,94] and in vitro 3D tissue model using human iPSC-derived cells [95] thus provide relevant tools for drug screening. To date, the amelioration of disease phenotypes in HGPS mouse models by the different tested therapies is limited, and this is even more true in HGPS patients. One could speculate that the age of the HGPS participant patients in the clinical trials could determine the outcomes of the treatments, but accord- ing to the clinical trial using lonafarnib, the authors stated that no associations between age at time of treatment and outcome measures that were improved at end of study were identiļ¬ed. At the same time, it should be noted that given the limited number of par- ticipants, it is difļ¬cult to correlate age with outcomes of the treatment. As research progresses, it becomes clear that target- ing the pathophysiology of progeria to a single level will likely be insufļ¬cient to signiļ¬cantly alleviate the signs and change the normal course of such a multi- organ devastating disease. Altogether, there is a major rationale in targeting progerin at different levels. Addressing therapies in HGPS associated to progerin accumulation may thus rely on multi-approaches combination, including its decreased production, increased degradation, or downstream noxious cascades. As a future therapeutic approach that still remains to be evaluated for HGPS is the Clustered Regularly Interspaced Short Palindromic Repeats/Cas protein (CRISPR/Cas) for in vivo gene editing [96ā€“98] and repair of the disease-causing mutation. On the other hand, Adeno-Associated Virus (AAVs) have gained popularity, in the last years, as safe gene delivery vec- tors, due to their ability to mediate long-term expres- sion in both non-dividing and dividing cells, with speciļ¬c tissue tropism [99ā€“101]. In this regard, AAVs constitute interesting CRISPR/Cas9 delivery candi- dates to speciļ¬cally repair the progeria-causing muta- tion. Preferentially, AAV serotype should target VSMCs, since they are involved in heart attack and stroke that represent the main cause of death in proge- ria patients. To overcome the limited packaging capacity of AAV (Ā» 4.8 kb), small Cas9 orthologues, including Staphylococcus aureus-derived Cas9 (SaCas9, 3.16 kb) [102], has been used to package the NUCLEUS 271
  • 10. Cas9 and its gRNA into a single AAV delivery vehicle for in vivo genome editing. CRISPR libraries should be designed to ideally select those able to target the mutation both in mouse and human genes with high on-target activity and low off-target activity, so that the effectiveness of the same gRNA could be studied ļ¬rst on human HGPS ļ¬broblasts and then in vivo on the Lmna G609G/G609G mice. Recent studies have shown the involvement of microRNAs and in particular, mir-9 which prevents progerin accumulation in HGPS neurons. Therefore, another possibility for developing new therapeutic approaches for progeria would be the modulation of microRNAs, taking into consideration off-targets that, again, must be evaluated in vitro and in vivo before transposition to humans [103]. Figure 2. Treatment strategies for progeria and their targets. Envisaged treatments for progeria target several mechanisms triggering the disease. By acting on the genetic cause, recent advances in genome editing using the CRISPR technique could be beneļ¬cial to repair the mutation causing the disease. Other molecules target the aberrant splicing process leading to the production of a truncated protein, by blocking the splice site with AONs or by inhibiting the splicing factor SRSF-1 (Metformin, MG132). During prelamin A maturation, treatments inhibiting isoprenylation (farnesylation or geranylation) or the methylation have shown their effectiveness in reducing pro- gerin toxicity (Statins, aminobisphonates, FTIs, GGTI-2147, monoaminopyrimidines, shICMT). To counter the accumulation of progerin, several molecules have been described as activators of autophagy leading to progerin degradation (Rapamycin, Sulphoraphane, Reti- noids and MG132). Finally, by acting on the reduction of the noxious downstream effects of progerin, some approaches have shown their effectiveness against inļ¬‚ammation (sodium salicylate, DOT1L inhibition by epz-4777), oxidative stress (NAC, NRF2 reactivation), nuclear shape abnormalities (Remodelin), mitochondrial abnormalities (Rock inhibitor: Y-27632, Methylene blue), impaired pyrophos- phate metabolism (inorganic pyrophosphate: PPi), vitamin D receptor signaling (vitamin D), decreased deacetylase activity (Resveratrol), Progerin/Lamin AC interaction (JH4) or aging hallmarks by reprogramming HGPS cells to pluripotency in vitro and in vivo. 272 K. HARHOURI ET AL.
  • 11. Abbreviations AON Antisense Oligonucleotides; ATM Ataxia Telangiectasia Mutated; BAF Barrier-to-Autointegration Factor; CRISPR Clustered Regularly Interspaced Short Pal- indromic Repeats; DOT1L Disruptor Of Telomeric silencing 1-Like; DSB Double-Strand Break; EZH2 Enhancer of Zeste Homolog 2 polycomb repressive complex 2 subunit; FTase Farnesyltransferase; FTI Farnesyl Transferase Inhibitor; GGTI GeranylGeranylTransferase Inhibitor; HDR Homology-Directed Repair; HGPS Hutchinson-Gilford Progeria Syndrome; HP1 Heterochromatin Protein 1; ICMT Isoprenylcysteine CarboxylMethylTransferase; KLF4 Krā‚¬ uppel-Like Factor 4. Lap2a Lamina-associated Polypeptide 2a; MAD-B Mandibuloacral Dysplasia type B; Mtor Mammalian Target Of Rapamycin; MSC Mesenchymal Stem Cells; NAC N-Acetyl Cysteine; NAT10 N-acetyltransferase 10; NFkB Nuclear Factor kappa B; NHEJ Non-Homologous End Joining; NRF2 Nuclear Factor-E2-related factor 2; OCT4 Octamer-binding transcription factor 4; PML-NB ProMyelocytic Leukemia-Nuclear Body; ROS Reactive oxidative species; SIRT-1 Sirtuin-1; SOX2 Sex determining region Y-box 2; SRSF-1 Serine/arginine-Rich Splicing Factor 1; TNF Tumor Necrosis Factor; VSMC Vascular Smooth Muscle Cells; ZOPRA ZOledronate-PRAvastatin; Disclosure of potential conļ¬‚icts of interest No potential conļ¬‚icts of interest were disclosed. Funding This work was supported by grants from Institut National de la Sant e et de la Recherche M edicale (INSERM), Aix-Marseille University, A Midex Foundation (VinTAGE Program) and the Association FrancĢ§aise contre les Myopathies (AFM grant MNH-Decrypt 2011ā€“2015 and TRIM-RD 2016ā€“2020 to NL). This study is part of the FHU A MIDEX project MARCHE n. ANR-11-IDEX-001-02 funded by the ā€œInvestissement dā€™avenirā€ French governmental program, managed by the French National Research Agency (ANR). ORCID Karim Harhouri http://orcid.org/0000-0003-2904-5221 Diane Frankel http://orcid.org/0000-0003-3321-6951 Patrice Roll http://orcid.org/0000-0002-0045-5641 Annachiara De Sandre-Giovannoli http://orcid.org/0000- 0002-2324-2462 Nicolas L evy http://orcid.org/0000-0001-5171-6365 References [1] Hennekam RC. Hutchinson-Gilford progeria syndrome: review of the phenotype. Am J Med Genet A. 2006;140:2603ā€“24. [2] Merideth MA, Gordon LB, Clauss S, et al. Phenotype and course of Hutchinson-Gilford progeria syndrome. N Engl J Med. 2008;358:592ā€“604. [3] Hutchinson J. Congenital absence of hair and mam- mary glands with atrophic condition of the skin and its appendages, in a boy whose mother had been almost wholly bald from Alopecia Areata from the age of six. Med Chir Trans. 1886;69:473ā€“7. [4] Gilford H. On a condition of mixed premature and immature development. Med Chir Trans. 1897;80:17- 46:25. [5] De Sandre-Giovannoli A, Bernard R, Cau P, et al. Lamin a truncation in Hutchinson-Gilford progeria. Science. 2003;300:2055. [6] Eriksson M, Brown WT, Gordon LB, et al. Recurrent de novo point mutations in lamin A cause Hutchinson- Gilford progeria syndrome. Nature. 2003;423:293ā€“8. [7] Goldman RD, Shumaker DK, Erdos MR, et al. Accumu- lation of mutant lamin A causes progressive changes in nuclear architecture in Hutchinson-Gilford progeria syndrome. Proc Natl Acad Sci U S A. 2004;101:8963ā€“8. [8] Capell BC, Erdos MR, Madigan JP, et al. Inhibiting far- nesylation of progerin prevents the characteristic nuclear blebbing of Hutchinson-Gilford progeria syn- drome. Proc Natl Acad Sci U S A. 2005;102:12879ā€“84. [9] Scafļ¬di P, Misteli T. Reversal of the cellular phenotype in the premature aging disease Hutchinson-Gilford pro- geria syndrome. Nat Med. 2005;11:440ā€“5. [10] Huang S, Chen L, Libina N, et al. Correction of cellular phenotypes of Hutchinson-Gilford Progeria cells by RNA interference. Hum Genet. 2005;118:444ā€“50. [11] Burtner CR, Kennedy BK. Progeria syndromes and age- ing: What is the connection? Nat Rev Mol Cell Biol. 2010;11:567ā€“78. [12] Liu B, Wang J, Chan KM, et al. Genomic instability in laminopathy-based premature aging. Nat Med. 2005;11:780ā€“5. [13] Hilton BA, Liu J, Cartwright BM, et al. Progerin seques- tration of PCNA promotes replication fork collapse and NUCLEUS 273
  • 12. mislocalization of XPA in laminopathy-related proge- roid syndromes. FASEB J. 2017;31:3882ā€“93. [14] Viteri G, Chung YW, Stadtman ER. Effect of progerin on the accumulation of oxidized proteins in ļ¬broblasts from Hutchinson Gilford progeria patients. Mech Age- ing Dev. 2010;131:2ā€“8. [15] Rivera-Torres J, Acin-Perez R, Cabezas-Sanchez P, et al. Identiļ¬cation of mitochondrial dysfunction in Hutchin- son-Gilford progeria syndrome through use of stable isotope labeling with amino acids in cell culture. J Prote- omics. 2013;91:466ā€“77. [16] Columbaro M, Capanni C, Mattioli E, et al. Rescue of heterochromatin organization in Hutchinson-Gilford progeria by drug treatment. Cell Mol Life Sci. 2005;62:2669ā€“78. [17] Kubben N, Adriaens M, Meuleman W, et al. Mapping of lamin A- and progerin-interacting genome regions. Chromosoma. 2012;121:447ā€“64. [18] McCord RP, Nazario-Toole A, Zhang H, et al. Corre- lated alterations in genome organization, histone meth- ylation, and DNA-lamin A/C interactions in Hutchinson-Gilford progeria syndrome. Genome Res. 2013;23:260ā€“9. [19] Shumaker DK, Dechat T, Kohlmaier A, et al. Mutant nuclear lamin A leads to progressive alterations of epi- genetic control in premature aging. Proc Natl Acad Sci U S A. 2006;103:8703ā€“8. [20] Scafļ¬di P, Misteli T. Lamin A-dependent misregulation of adult stem cells associated with accelerated ageing. Nat Cell Biol. 2008;10:452ā€“9. [21] Liu Y, Wang Y, Rusinol AE, et al. Involvement of xero- derma pigmentosum group A (XPA) in progeria arising from defective maturation of prelamin A. Faseb J. 2008;22:603ā€“11. [22] Manju K, Muralikrishna B, Parnaik VK. Expression of disease-causing lamin A mutants impairs the formation of DNA repair foci. J Cell Sci. 2006;119:2704ā€“14. [23] Prokocimer M, Barkan R, Gruenbaum Y. Hutchinson- Gilford progeria syndrome through the lens of tran- scription. Aging Cell. 2013;12:533ā€“43. [24] Hernandez L, Roux KJ, Wong ESM, et al. Functional coupling between the extracellular matrix and nuclear Lamina by wnt signaling in Progeria. Dev Cell. 2010;19:413ā€“25. [25] Osorio FG, Barcena C, Soria-Valles C, et al. Nuclear lamina defects cause ATM-dependent NF-kappaB acti- vation and link accelerated aging to a systemic inļ¬‚am- matory response. Genes Dev. 2012;26:2311ā€“24. [26] Kubben N, Zhang W, Wang L, et al. Repression of the Antioxidant NRF2 Pathway in Premature Aging. Cell. 2016;165:1361ā€“74. [27] Cau P, Navarro C, Harhouri K, et al. Nuclear matrix, nuclear envelope and premature aging syndromes in a translational research perspective. Seminars Cell Dev Biol. 2014;29:125ā€“47. [28] Delbarre E, Tramier M, Coppey-Moisan M, et al. The truncated prelamin A in Hutchinson-Gilford progeria syndrome alters segregation of A-type and B-type lamin homopolymers. Hum Mol Genetics. 2006;15:1113ā€“22. [29] Basso AD, Kirschmeier P, Bishop WR. Lipid posttrans- lational modiļ¬cations. Farnesyl transferase inhibitors. J Lipid Res. 2006;47:15ā€“31. [30] Glynn MW, Glover TW. Incomplete processing of mutant lamin A in Hutchinson-Gilford progeria leads to nuclear abnormalities, which are reversed by farnesyltransferase inhibition. Hum Mol Genetics. 2005;14:2959ā€“69. [31] Toth JI, Yang SH, Qiao X, et al. Blocking protein farne- syltransferase improves nuclear shape in ļ¬broblasts from humans with progeroid syndromes. Proc Natl Acad Sci U S A. 2005;102:12873ā€“8. [32] Yang SH, Bergo MO, Toth JI, et al. Blocking protein far- nesyltransferase improves nuclear blebbing in mouse ļ¬broblasts with a targeted Hutchinson-Gilford progeria syndrome mutation. Proc Natl Acad Sci U S A. 2005;102:10291ā€“6. [33] Mallampalli MP, Huyer G, Bendale P, et al. Inhibiting farnesylation reverses the nuclear morphology defect in a HeLa cell model for Hutchinson-Gilford progeria syn- drome. Proc Natl Acad Sci U S A. 2005;102:14416ā€“21. [34] Yang SH, Meta M, Qiao X, et al. A farnesyltransferase inhibitor improves disease phenotypes in mice with a Hutchinson-Gilford progeria syndrome mutation. J Clin Invest. 2006;116:2115ā€“21. [35] Yang SH, Andres DA, Spielmann HP, et al. Progerin elicits disease phenotypes of progeria in mice whether or not it is farnesylated. J Clin Invest. 2008;118:3291ā€“300. [36] Capell BC, Olive M, Erdos MR, et al. A farnesyltransfer- ase inhibitor prevents both the onset and late progres- sion of cardiovascular disease in a progeria mouse model. Proc Natl Acad Sci U S A. 2008;105:15902ā€“7. [37] Gordon LB, Kleinman ME, Miller DT, et al. Clinical trial of a farnesyltransferase inhibitor in children with Hutchinson-Gilford progeria syndrome. Proc Natl Acad Sci U S A. 2012;109:16666ā€“71. [38] Gordon LB, Massaro J, Dā€™Agostino RB, et al. Impact of Farnesylation inhibitors on survival in Hutchinson-Gil- ford Progeria syndrome. Circulation. 2014;130:27 [39] Varela I, Pereira S, Ugalde AP, et al. Combined treat- ment with statins and aminobisphosphonates extends longevity in a mouse model of human premature aging. Nat Med. 2008;14:767ā€“72. [40] Gordon LB, Kleinman ME, Massaro J, et al. Clinical Trial of the Protein Farnesylation inhibitors Lonafarnib, Pravastatin, and Zoledronic Acid in Children With Hutchinson-Gilford Progeria Syndrome. Circulation. 2016;134:114ā€“25. [41] Davies BS, Barnes RH, 2nd, Tu Y, et al. An accumula- tion of non-farnesylated prelamin A causes cardiomy- opathy but not progeria. Hum Mol Genetics. 2010;19:2682ā€“94. [42] Blondel S, Egesipe AL, Picardi P, et al. Drug screening on Hutchinson Gilford progeria pluripotent stem cells reveals aminopyrimidines as new modulators of farne- sylation. Cell Death Dis. 2016;7:e2105. 274 K. HARHOURI ET AL.
  • 13. [43] Ibrahim MX, Sayin VI, Akula MK, et al. Targeting iso- prenylcysteine methylation ameliorates disease in a mouse model of progeria. Science. 2013;340:1330ā€“3. [44] Cao K, Graziotto JJ, Blair CD, et al. Rapamycin reverses cellular phenotypes and enhances mutant protein clear- ance in Hutchinson-Gilford progeria syndrome cells. Sci Transl Med. 2011;3:89ra58. [45] Graziotto JJ, Cao K, Collins FS, et al. Rapamycin acti- vates autophagy in Hutchinson-Gilford progeria syn- drome: implications for normal aging and age- dependent neurodegenerative disorders. Autophagy. 2012;8:147ā€“51. [46] Cenni V, Capanni C, Columbaro M, et al. Autophagic degradation of farnesylated prelamin A as a therapeutic approach to lamin-linked progeria (vol 55, e36, 2011). Eur J Histochem. 2013;57:283ā€“6. [47] Ramos FJ, Chen SC, Garelick MG, et al. Rapamycin reverses elevated mTORC1 signaling in Lamin A/C- deļ¬cient mice, rescues cardiac and skeletal muscle func- tion, and extends survival. Sci Translational Med. 2012;4:144ra103 [48] Liao CY, Anderson SS, Chicoine NH, et al. Rapamycin reverses metabolic deļ¬cits in Lamin A/C-Deļ¬cient mice. Cell Rep. 2016;17:2542ā€“52. [49] Cho HJ, Park J, Lee HW, et al. Regulation of adipocyte differentiation and insulin action with rapamycin. Bio- chem Bioph Res Co. 2004;321:942ā€“8. [50] Yeh WC, Bierer BE, Mcknight SL. Rapamycin inhibits clonal expansion and adipogenic differentiation of 3t3- L1 cells. Proc Natl Acad Sci U S A. 1995;92:11086ā€“90. [51] Gabriel D, Roedl D, Gordon LB, et al. Sulforaphane enhances progerin clearance in Hutchinson-Gilford progeria ļ¬broblasts. Aging Cell. 2015;14:78ā€“91. [52] Gabriel D, Shafry DD, Gordon LB, et al. Intermittent treat- ment with farnesyltransferase inhibitor and sulforaphane improves cellular homeostasis in Hutchinson-Gilford pro- geria ļ¬broblasts. Oncotarget. 2017;8:64809ā€“26. [53] Kubben N, Brimacombe KR, Donegan M, et al. A high- content imaging-based screening pipeline for the sys- tematic identiļ¬cation of anti-progeroid compounds. Methods. 2016;96:46ā€“58. [54] Pellegrini C, Columbaro M, Capanni C, et al. All-trans reti- noic acid and rapamycin normalize Hutchinson Gilford pro- geria ļ¬broblast phenotype. Oncotarget. 2015;6:29914ā€“28. [55] Harhouri K, Navarro C, Depetris D, et al. MG132- induced progerin clearance is mediated by autophagy activation and splicing regulation. EMBO Mol Med. 2017;9:1294ā€“313. [56] Osorio FG, Navarro CL, Cadinanos J, et al. Splicing- directed therapy in a new mouse model of human accel- erated aging. Sci Transl Med. 2011;3:106ra7. [57] Lee JM, Nobumori C, Tu Y, et al. Modulation of LMNA splicing as a strategy to treat prelamin A diseases. J Clin Invest. 2016;126:1592ā€“602. [58] Barthelemy F, Navarro C, Fayek R, et al. Truncated prel- amin A expression in HGPS-like patients: a transcrip- tional study. Eur J Hum Genet. 2015;23:1051ā€“61. [59] Harhouri K, Navarro C, Baquerre C, et al. Antisense- based Progerin downregulation in HGPS-Like Patientsā€™ cells. Cells. 2016;5. pii: E31 [60] Lopez-Mejia IC, Vautrot V, De Toledo M, Behm-Ansm- ant I, et al. A conserved splicing mechanism of the LMNA gene controls premature aging. Hum Mol Genetics. 2011;20:4540ā€“55. [61] Larsson O, Morita M, Topisirovic I, et al. Distinct pertur- bation of the translatome by the antidiabetic drug metfor- min. Proc Natl Acad Sci U S A. 2012;109:8977ā€“82. [62] Egesipe AL, Blondel S, Cicero AL, et al. Metformin decreases progerin expression and alleviates pathologi- cal defects of Hutchinson-Gilford progeria syndrome cells. NPJ Aging Mechan Dis. 2016;2:16026. [63] Park SK, Shin OS. Metformin alleviates ageing cellular phenotypes in Hutchinson-Gilford progeria syndrome dermal ļ¬broblasts. Exp Dermatol. 2017;26:889ā€“95. [64] Richards SA, Muter J, Ritchie P, et al. The accumulation of un-repairable DNA damage in laminopathy progeria ļ¬broblasts is caused by ROS generation and is prevented by treatment with N-acetyl cysteine. Hum Mol Genetics. 2011;20:3997ā€“4004. [65] Tilstra JS, Clauson CL, Niedernhofer LJ, et al. NF-kap- paB in aging and disease. Aging Dis. 2011;2:449ā€“65. [66] Kang HT, Park JT, Choi K, et al. Chemical screening identiļ¬es ROCK as a target for recovering mitochon- drial function in Hutchinson-Gilford progeria syn- drome. Aging Cell. 2017;16:541ā€“50. [67] Cui W, Bai Y, Luo P, et al. Preventive and therapeutic effects of MG132 by activating Nrf2-ARE signaling path- way on oxidative stress-induced cardiovascular and renal injury. Oxidative Med Cell Longevity. 2013;2013:306073. [68] Wang Y, Sun W, Du B, et al. Therapeutic effect of MG- 132 on diabetic cardiomyopathy is associated with its suppression of proteasomal activities: roles of Nrf2 and NF-kappaB. Am J Physiol Heart Circulatory Physiol. 2013;304:H567ā€“78. [69] Dreger H, Westphal K, Wilck N, et al. Protection of vas- cular cells from oxidative stress by proteasome inhibition depends on Nrf2. Cardiovascular Res. 2010;85:395ā€“403. [70] Rivera-Torres J, Acin-Perez R, Cabezas-Sanchez P, et al. Identiļ¬cation of mitochondrial dysfunction in Hutchin- son-Gilford progeria syndrome through use of stable isotope labeling with amino acids in cell culture. J Prote- omics. 2013;91:466ā€“77. [71] Villa-Bellosta R, Rivera-Torres J, Osorio FG, et al. Defective extracellular pyrophosphate metabolism promotes vascular calciļ¬cation in a mouse model of Hutchinson-Gilford Progeria Syndrome that is Ame- liorated on Pyrophosphate treatment. Circulation. 2013;127:2442ā€“51. [72] Xiong ZM, Choi JY, Wang K, et al. Methylene blue alle- viates nuclear and mitochondrial abnormalities in pro- geria. Aging Cell. 2016;15:279ā€“90. [73] Soria-Valles C, Osorio FG, Gutierrez-Fernandez A, et al. NF-kappaB activation impairs somatic cell reprogram- ming in ageing. Nat Cell Biol. 2015;17:1004ā€“13. NUCLEUS 275
  • 14. [74] Ortiz-Lazareno PC, Hernandez-Flores G, Dominguez- Rodriguez JR, et al. MG132 proteasome inhibitor modu- lates proinļ¬‚ammatory cytokines production and expres- sion of their receptors in U937 cells: involvement of nuclear factor-kappaB and activator protein-1. Immu- nology. 2008;124:534ā€“41. [75] Hozhabri NST, Kim H, Varanasi V. NF-kappa B inhibi- tor MG132 enhances differentiation and collagen expression of dental pulp stem cells. Faseb J. 2014;28 [76] Ahmed AS, Ahmed M, Li J, et al. Proteasome inhibitor MG132 modulates inļ¬‚ammatory pain by central mecha- nisms in adjuvant arthritis. Int J Rheum Dis. 2017;20:25ā€“32. [77] Olive M, Harten I, Mitchell R, et al. Cardiovascular pathology in Hutchinson-Gilford progeria: correlation with the vascular pathology of aging. Arterioscler Thromb Vasc Biol. 2010;30:2301ā€“9. [78] Chatterjee PK, Yeboah MM, Dowling O, et al. Nicotinic acetylcholine receptor agonists attenuate septic acute kidney injury in mice by suppressing inļ¬‚ammation and proteasome activity. PloS One. 2012;7:e35361. [79] Miao X, Cui W, Sun W, et al. Therapeutic effect of MG132 on the aortic oxidative damage and inļ¬‚amma- tory response in OVE26 type 1 diabetic mice. Oxidative Med Cell Longevity. 2013;2013:879516. [80] Feng B, Zhang Y, Mu J, et al. Preventive effect of a pro- teasome inhibitor on the formation of accelerated ath- erosclerosis in rabbits with uremia. J Cardiovascular Pharmacol. 2010;55:129ā€“38. [81] Larrieu D, Britton S, Demir M, et al. Chemical inhibi- tion of NAT10 corrects defects of laminopathic cells. Science. 2014;344:527ā€“32. [82] Bouillon R, Carmeliet G, Verlinden L, et al. Vitamin D and human health: Lessons from Vitamin D receptor null mice. Endocr Rev. 2008;29:726ā€“76. [83] Plum LA, DeLuca HF. Vitamin D, disease and therapeutic opportunities. Nat Rev Drug Discovery. 2010;9:941ā€“55. [84] Kreienkamp R, Croke M, Neumann MA, et al. Vitamin D receptor signaling improves Hutchinson-Gilford progeria syndrome cellular phenotypes. Oncotarget. 2016;7:30018ā€“31. [85] Lee SJ, Jung YS, Yoon MH, et al. Interruption of pro- gerin-lamin A/C binding ameliorates Hutchinson-Gil- ford progeria syndrome phenotype. J Clin Invest. 2016;126:3879ā€“93. [86] Liu BH, Ghosh S, Yang X, et al. Resveratrol Rescues SIRT1-Dependent Adult Stem Cell Decline and Allevi- ates Progeroid Features in Laminopathy-Based Progeria. Cell Metab. 2012;16:738ā€“50. [87] Varela I, Pereira S, Ugalde AP, et al. Combined treat- ment with statins and aminobisphosphonates extends longevity in a mouse model of human premature aging. Nat Med. 2008;14:767ā€“72. [88] Schmidt E, Nilsson O, Koskela A, et al. Expression of the Hutchinson-Gilford progeria mutation during oste- oblast development results in loss of osteocytes, irregular mineralization, and poor biomechanical prop- erties. J Biol Chem. 2012;287:33512ā€“22. [89] Strandgren C, Nasser HA, McKenna T, et al. Transgene silencing of the Hutchinson-Gilford progeria syndrome mutation results in a reversible bone phenotype, whereas resveratrol treatment does not show overall beneļ¬cial effects. FASEB J. 2015;29:3193ā€“205. [90] Howells LM, Berry DP, Elliott PJ, et al. Phase I random- ized, double-blind pilot study of micronized resveratrol (SRT501) in patients with hepatic metastasesā€“safety, pharmacokinetics, and pharmacodynamics. Cancer Pre- vention Res. 2011;4:1419ā€“25. [91] Ocampo A, Reddy P, Martinez-Redondo P, et al. In Vivo Amelioration of age-associated Hallmarks by par- tial reprogramming. Cell. 2016;167:1719ā€“33 e12. [92] Ocampo A, Reddy P, Izpisua Belmonte JC. Anti-aging strategies based on cellular reprogramming. Trends Mol Med. 2016;22:725ā€“38. [93] Liu GH, Barkho BZ, Ruiz S, et al. Recapitulation of pre- mature ageing with iPSCs from Hutchinson-Gilford progeria syndrome. Nature. 2011;472:221ā€“5. [94] Zhang JQ, Lian QZ, Zhu GL, et al. A human iPSC model of Hutchinson Gilford Progeria reveals vascular smooth muscle and Mesenchymal stem cell defects. Cell Stem Cell. 2011;8:31ā€“45. [95] Atchison L, Zhang HY, Cao K, et al. A tissue engineered blood vessel model of Hutchinson-Gilford Progeria Syndrome using human iPSC-derived smooth muscle cells. Sci Rep-Uk. 2017;7:8168. [96] Sander JD, Joung JK. CRISPR-Cas systems for editing, regulating and targeting genomes. Nat Biotechnol. 2014;32:347ā€“55. [97] Doudna JA, Charpentier E. Genome editing. The new frontier of genome engineering with CRISPR-Cas9. Sci- ence. 2014;346:1258096. [98] Hsu PD, Lander ES, Zhang F. Development and appli- cations of CRISPR-Cas9 for genome engineering. Cell. 2014;157:1262ā€“78. [99] Kotterman MA, Schaffer DV. Engineering adeno-asso- ciated viruses for clinical gene therapy. Nat Rev Genet- ics. 2014;15:445ā€“51. [100] Yin H, Kauffman KJ, Anderson DG. Delivery technolo- gies for genome editing. Nat Rev Drug Discovery. 2017;16:387ā€“99. [101] Suzuki K, Tsunekawa Y, Hernandez-Benitez R, et al. In vivo genome editing via CRISPR/Cas9 mediated homol- ogy-independent targeted integration. Nature. 2016;540:144ā€“9. [102] Ran FA, Cong L, Yan WX, et al. In vivo genome edit- ing using Staphylococcus aureus Cas9. Nature. 2015;520:186ā€“U98. [103] Frankel D, Delecourt V, Harhouri K, et al. MicroRNAs in hereditary and sporadic premature aging syndromes and other laminopathies. Aging Cell. 2018;e12766. 276 K. HARHOURI ET AL.