SlideShare a Scribd company logo
1 of 11
Download to read offline
CALL FOR PAPERS Novel Aspects of Adipocyte Biology
Zinc transporter Slc39a14 regulates inflammatory signaling associated
with hypertrophic adiposity
Catalina Troche, Tolunay Beker Aydemir, and Robert J. Cousins
Food Science and Human Nutrition Department and Center for Nutritional Sciences, College of Agricultural and Life
Sciences, University of Florida, Gainesville, Florida
Submitted 23 September 2015; accepted in final form 2 December 2015
Troche C, Beker Aydemir T, Cousins RJ. Zinc transporter Slc39a14
regulates inflammatory signaling associated with hypertrophic adiposity.
Am J Physiol Endocrinol Metab 310: E258–E268, 2016. First published
December 8, 2015; doi:10.1152/ajpendo.00421.2015.—Zinc is a signal-
ing molecule in numerous metabolic pathways, the coordination of which
occurs through activity of zinc transporters. The expression of zinc
transporter Zip14 (Slc39a14), a zinc importer of the solute carrier 39
family, is stimulated under proinflammatory conditions. Adipose tissue
upregulates Zip14 during lipopolysaccharide-induced endotoxemia. A
null mutation of Zip14 (KO) revealed that phenotypic changes in
adipose include increased cytokine production, increased plasma lep-
tin, hypertrophied adipocytes, and dampened insulin signaling. Adi-
pose tissue from KO mice had increased levels of preadipocyte
markers, lower expression of the differentiation marker (PPAR␥), and
activation of NF-␬B and STAT3 pathways. Our overall hypothesis
was that ZIP14 would play a role in adipocyte differentiation and
inflammatory obesity. Global Zip14 KO causes systemic endotox-
emia. The observed metabolic changes in adipose metabolism were
reversed when oral antibiotics were administrated, indicating that
circulating levels of endotoxin were in part responsible for the adipose
phenotype. To evaluate a mechanism, 3T3-L1 cells were differenti-
ated into adipocytes and treated with siRNA to knock down Zip14.
These cells had an impaired ability to mobilize zinc, which caused
dysregulation of inflammatory pathways (JAK2/STAT3 and NF-␬B).
The Zip14 deletion may limit the availability of intracellular zinc,
yielding the unique phenotype of inflammation coupled with hyper-
trophy. Taken together, these results suggest that aberrant zinc distri-
bution observed with Zip14 ablation impacts adipose cytokine pro-
duction and metabolism, ultimately increasing fat deposition when
exposed to endotoxin. To our knowledge, this is the first investigation
into the mechanistic role of ZIP14 in adipose tissue regulation and
metabolism.
adipose; endotoxemia; zinc signaling; hypertrophy; inflammation
WHITE ADIPOSE TISSUE (WAT) contains a wide array of cell types
that are characterized as connective tissue, nervous tissue,
stromovascular cells, and immune cells. Together, these cells
produce unique substances, i.e., leptin, adiponectin, and resis-
tin, which contribute to the para- and autoendocrine regulation
of lipid metabolism (21, 28). Limited epidemiological studies
have linked aberrant trace mineral metabolism with adipose
pathology; e.g., obesity is clinically correlated to deficiencies
in iron, calcium, and zinc (18, 19, 49, 50). Zinc was first
thought to be involved in metabolic activity of adipocytes
through insulin-like effects (36) and through its inherent anti-
oxidant properties (16). Human adipose tissue expresses many
zinc transporters that may respond to the metabolic status of
the patients, e.g., lean vs. obese (47). Despite the apparent
correlation between zinc and adipose metabolism, a mecha-
nism of action has yet to be defined.
Zinc partitioning within mammalian cells is due to ZIP and
ZnT transporter activity, which control influx and efflux, re-
spectively, of cytosolic zinc (27, 31). ZIP14 is a zinc influx
transporter that is known to be upregulated during inflamma-
tion (3–5, 34, 40). We have focused on ZIP14 since it was
found in our initial experiments to be the most responsive zinc
transporter in mouse liver post-lipopolysaccharide administra-
tion (34). Knockout (KO) of ZIP14 results in a variety of
unique phenotypes. We have reported previously that mice
lacking ZIP14 have impaired liver zinc uptake (3) and elevated
levels of serum endotoxin (25). Previously, Aydemir et al. (3)
reported an increase in fat/lean ratios in Zip14-KO mice, a
finding that led to our further investigation of ZIP14 in adipose
function. The elevated level of serum endotoxin is particularly
relevant to adipose in that chronic exposure to endotoxin can
initiate obesity and insulin resistance (10). Inflammatory cyto-
kines have a proliferative effect on adipocytes, leading to
expansion of cell mass through both hypertrophy and hyper-
plasia (14, 20). Collectively, the marked induction of ZIP14 in
WAT during inflammation along with the KO phenotype of
increased adiposity, and metabolic endotoxemia suggests that
this transporter alters zinc trafficking in adipocytes with func-
tional outcomes.
Based on these previous findings, we hypothesized that
ZIP14 would be critical to the inflammatory response and
ultimately metabolic activity of WAT. Here, we report that
WAT from KO mice appears to be insulin insensitive with
hypertrophied adipocytes and dampened insulin signaling.
Insulin resistance was linked to chronic inflammation within
KO adipose tissue through upregulated cytokine expression
and the Toll-like receptor 4 (TLR4) accessory protein my-
eloid differentiation primary response gene 88 (MyD88).
Finally, we show that aberrant zinc signaling within the KO
adipocyte is linked to enhanced JAK/STAT and NF-␬B
signaling, leading to impaired differentiation. Both findings
tie directly to dyslipidemia and hypertrophy. The involve-
ment of ZIP14 in major inflammatory pathways impacts
adipocyte development and makes it a potential therapeutic
target for inflammatory disorders in adipose, e.g., obesity
and insulin resistance.
Address for reprint requests and other correspondence: R. J. Cousins, Univ.
of Florida, 572 Newell Dr., P. O. Box 110370, Gainesville, FL 32611-0370
(e-mail: cousins@ufl.edu).
Am J Physiol Endocrinol Metab 310: E258–E268, 2016.
First published December 8, 2015; doi:10.1152/ajpendo.00421.2015.
0193-1849/16 Copyright © 2016 the American Physiological Society http://www.ajpendo.orgE258
MATERIALS AND METHODS
Animals and diets. Design and genomic validation of the Zip14-KO
(Zip14Ϫ/Ϫ
) mice have been described previously (3, 5, 33). Briefly,
Zip14 heterozygous mice were used to establish a breeding colony to
generate wild-type (WT; Zip14ϩ/ϩ
) and KO mice. For the experi-
ments described herein, female KO and WT mice were used between
8 and 16 wk of age. Mice were provided with ad libitum access to a
commercial chow rodent diet (7912, with 60 mg zinc/kg provided by
ZnO; Harlan-Teklad, Indianapolis, IN) and tap water.
Animal treatments. In experiments modeling acute inflammation,
lipopolysaccharide (LPS; E. coli serotype 055:B5; Sigma, St. Louis,
MO) in phosphate-buffered saline (PBS) was administered at 2.0
mg/kg via intraperitoneal (ip) injection. Mice received LPS injections
up to 18 h prior to euthanasia. Control animals received ip injections
of PBS. In other experiments, fasted mice were gavaged with 65
Zn (2
␮Ci/mouse in 250 ␮l of saline) and euthanized 3 h postgavage to
determine the tissue distribution. Specific activity of the 65
Zn (Perkin-
Elmer, Waltham, MA) when used was 4.4 mCi/mg. Tissue accumu-
lation of 65
Zn was measured via ␥-scintillation spectrometry. For the
antibiotic experiment, ultrapure (MiliQ, Billerica, MA) drinking water
was supplemented with neomycin (0.5 mg/ml) and ampicillin (1
mg/ml) for 4 wk prior to tissue collection. Mice were anesthetized
using isoflurane (Baxter, Deerfield, IL) prior to injection or gavage.
Euthanasia was conducted via cardiac puncture. Blood from cardiac
puncture was collected into a clot activator microgel barrier collection
tube (Capiject; Terumo Medical, Somerset, NJ). Serum was separated
via centrifugation and stored at Ϫ80°C prior to further analysis. All
harvested tissues (intestine, muscle, liver, and adipose) were snap-
frozen in liquid nitrogen and stored at Ϫ80°C prior to further pro-
cessing. All studies described herein used intra-abdominal white fat
pads (parameterial fat pads) that lay along the uterine horn. Unless
otherwise specified, tissues were homogenized in assay-specific buf-
fers using a Bullet blender with zirconium oxide beads (Next Ad-
vance, Averill Park, NY). All animal protocols were approved by the
University of Florida Institutional Animal Care and Use Committee.
Cell culture. A well-established embryonic mouse fibroblast cell
line (3T3-L1; ATCC, Manassas, VA) was used to model adipocyte
growth and differentiation. Cells were cultured in growth medium
(DMEM;4.5 g/l glucose, 15% FBS, and 1% pencillin-streptomycin;
all from Corning, Manassas, VA). After two passages, subconfluent
primary cultures were trypsinized and plated into 12-well plates for
experimentation. Transient knockdown of ZIP14 was carried out with
HiPerfect (Qiagen, Valencia, CA). 3T3-L1 cells were grown to 90%
confluence, and cells were treated with 25 nM siRNA (Darmacon,
Pittsburgh, PA) for 48 h prior to incubation with differentiation
medium with or without LPS (10 ng/ml). Thereafter, cells were
exposed to differentiation medium (DMEM; 4.5 g/l glucose, 10%
FBS, and 1% pencillin-streptomycin supplemented with 1.7 ␮M
insulin, 1 ␮M dexamethasone, and 0.5 mM isobutylmethylxanthine).
Cells were exposed to differentiation medium for three days prior to
postdifferentiation medium (DMEM; 4.5 g/l glucose, 10% FBS, and
1% pencillin-streptomycin). The entire experimental period from
differentiation to collection was 8 days. For experiments which
involved LPS, cells were plated into their experimental dishes (pas-
sage 3), and 10 ng/ml LPS was supplemented to the medium the next
day. Thereafter, cells were exposed to LPS throughout the entire
experimental course through differentiation; medium was changed
every 2 days.
To confirm our findings in 3T3L1 cells, ear mesenchymal stem
cells (EMSC) were collected from WT and KO mice, as described
previously by Rim et al. (42). Briefly, ears were minced and digested
in medium containing collagenase type I (Worthington, Lakewood,
NJ). Prior to the cell suspension being filtered (100 ␮m; Fisher
Scientific, Suwanee, GA) minced tissue and collagenase medium were
placed in a 37°C shaking water bath for 1 h. Filtered cells were
pelleted through centrifugation (360 g, 5 min, room temperature), and
red blood cells were lysed. Isolated cells were plated in 100-mm petri
dishes in the previously described growth medium. Cells were al-
lowed to expand for 3–5 days, and then these subconfluent primary
cultures were trypsinized and plated into 24-well plates for experi-
mentation. At visually confirmed confluence the EMSC were stimu-
lated into adipocytes with previously described differentiation me-
dium. After 3 days in differentiation medium, cells were maintained in
postdifferentiation media. The entire experimental period from differ-
entiation to collection was 10 days.
Analytical procedures. Serum was diluted 1:3 using ultrapure
water, and zinc levels were determined using flame atomic absorption
spectrophotometry (AAS). Adipose tissue was weighed prior to HNO3
digestion (90°C for 3 h) and diluted 1:1 with ultrapure water prior to
AAS analysis. In experiments where total zinc was determined in
cells, 3T3-L1 cells were first cultured in 150-mm plates (Corning).
Cells were collected in ice-cold PBS, and an aliquot was obtained for
total protein determination (Pierce BCA assay; Thermo Fisher Scien-
tific, Waltham, MA). The remaining cell pellet was digested in HNO3
(80°C for 3 h) and diluted 1:1 with ultrapure water. Total zinc was
measured by AAS, and values were normalized to total protein, as
described above. Serum endotoxin was measured as reported previ-
ously (25) using the LAL Chromogenic Endotoxin Quantitation Kit
(Thermo Fisher Scientific), with absorbance read at 407 nm. Serum
and tissue leptin were measured using a mouse/rat-specific ELISA kit
(Alpco, Salem, NH). Tissue was first homogenized in cell extraction
buffer (20 mM Tris·HCl, 1 mM EDTA, and 254 mM sucrose, pH 7.4),
and absorbance was measured at 450 nm (1). Leptin values were
normalized to total protein, as described above. Adipose glucose
content was determined enzymatically with a total glucose assay kit
(Sigma-Aldridge, St. Louis, MO). Prior to analysis, tissues were
homogenized in ultrapure water and diluted 1:2. Absorbance was
measured at 540 nm.
RNA quantification. For RNA isolation, a portion of frozen fat pad
was placed into TRIzol reagent (Life Technologies, Thermo Fisher
Scientific). Early experiments (Fig. 1, A–C) utilized a Polytron
blender to lyse tissue. Subsequent tissues were homogenized as
described previously using the Bullet Blender. cDNA was generated
using the iScript reagents (Bio-Rad, Hercules, CA). Quantitative
real-time PCR was performed using the Real-time PCR Fast SYBR
Green Master Mix and a StepOnePlus Fast Thermocycler (Applied
Biosystems, Thermo Fisher Scientific). Primers to detect specific
mRNAs were designed to span intron/exon boundaries: peroxisome
proliferator-activated receptor-␥ (Ppar␥), 5=-GGAAGACCACTC-
GCATTCCTT-3= and 5=-GTAATCAGCAACCATTGGGTCA-3=;
plasminogen activator inhibitor-1 (Pai-1), 5=-AGGGCTTCATGC-
CCCACTTCTTCA-3= and 5=-GTAGAGGGCATTCACCAGCA-
CCA-3=. All other primer sequences were selected using the qPrim-
erDepot database, http://mouseprimerdepot.nci.nih.gov/, as described
in Cui et al. (17). TBP mRNA was the normalizer for relative
expression, as described previously (3, 5).
Western blotting. Western analysis was performed using polyclonal
rabbit antibodies against ZIP14 developed and affinity purified (Life
Technologies, Thermo Fisher Scientific) as described previously
(3, 5). Akt, phosphorylated Akt (p-Ser473
), hormone-sensitive lipase
(HSL), phosphorylated HSL (Ser660
), I␬B, phosphorylated I␬B (p-
Ser32/36
), IR1␤, phosphorylated IR1␤ (p-Y1150/1151
), mammalian tar-
get of rapamycin (mTOR), phosphorylated mTOR (Ser2448
), MyD88,
NF-␬B, phosphorylated NF-␬B (p65), PPAR␥, preadipocyte factor-1
(PREF-1/DLK-1), SOCS3, STAT3, and phosphorylated STAT3 (p-
Y705
) antibodies were purchased from Cell Signaling Technology
(Boston, MA). F4/80 and zinc ␣2-glycoprotein (ZAG) antibody were
purchased from Santa Cruz Biotechnology (Dallas, TX). Frozen
tissues were processed over liquid nitrogen to prevent freeze thaw and
protein degradation. Tissues were homogenized in RIPA lysis buffer
(Santa Cruz Biotechnology) with 100ϫ protease and phosphatase
inhibitors (Thermo Fisher Scientific), along with phenylmethanesul-
fonyl fluoride (Sigma-Aldrich). Proteins were separated using a 10%
E259ZINC TRANSPORTER Zip14 AND ADIPOSE TISSUE REGULATION
AJP-Endocrinol Metab • doi:10.1152/ajpendo.00421.2015 • www.ajpendo.org
acrylamide gel for SDS-PAGE and transferred to nitrocellulose mem-
branes. Transfer was verified through Ponceau Red staining, and
proteins were visualized through chemiluminescence (SuperSignal
West Pico, Thermo Fisher Scientific) and digital imaging (Protein
Simple, San Jose, CA). Tubulin (Abcam, Cambridge, MA) abundance
was used as the loading control.
Adipose histology. Collected fat pads (parametrial fat pads) were
fixed in 10% neutral buffered formalin for 24 h prior to paraffin
embedding and sectioning. Slides were hematoxylin and eosin stained
prior to microscopy using a Zeiss Axiovert 100 microscope at ϫ10
magnification. Adipocyte area was measured using ImageJ software
(National Institute of Mental Health, Bethesda, MD). Cell areas were
determined on cells with contiguous borders using the Adiposoft
open-source ImageJ plugin http://fiji.sc/Adiposoft (22). Between 600
and 1,500 cells/image were used. Ten full images per genotype (5
mice, 2 images/mouse) were used to generate cell area images. In cell
experiments where Oil Red O was used, cells were first fixed in 3.3%
formaldehyde and stained in 3 ␮g/ml Oil Red O. Images were
collected using the previously described Zeiss Axiovert 100 at ϫ10
magnification. To quantify staining, Oil Red O was extracted with
isopropanol containing 4% NP-40, and absorbance was measured at
520 nm (62).
Confocal laser-scanning microscopy. Imaging was done with a
Leica TCS SP5 laser-scanning confocal microscope with LAS-AF
imaging software, using a ϫ40 oil objective. For detection of labile
zinc, cells were incubated with FluoZin-3 AM (Invitrogen, Waltham,
MA), as described previously (4). Briefly, cells were incubated in 5
␮M FluoZin-3 for 30 min, followed by a 30-min incubation in
Locke’s buffer (154 mM NaCl, 5.6 mM KCl, 3.6 mM NaHCO3, 1.2
mM MgSO4, 5.6 mM glucose, 2.5 mM CaCl2, and 10 mM HEPES).
Cells were then stimulated with 40 ␮M ZnCl2, and fluorescence was
measured at 516 nm with excitation at 494 nm. A similar method was
used to quantify FluoZin-3 AM in 12-well plates, with fluorescence
being determined as above. Immediately after fluorescence was read,
the cells were trypsanized and diluted 1:1 with trypan blue to deter-
mine cell number. FluoZin-3 AM relative fluorescent units were
Fig. 2. LPS-induced Zip14 expression is correlated with cytokine expression. A: mice received LPS (2 mg/kg ip) or the same volume (250 ␮l) of saline [control
(CTRL)] 1–18 h before being euthanized. Relative transcript levels of Zip14 and specific cytokines; n ϭ 3 mice/treatment Ϯ SE. B: serum hypozincemia confirms
the acute-phase response. Zinc concentrations in serum and WAT were measured by atomic absorption spectrophotometry (AAS). Adipose zinc values were
normalized to wet tissue weight; n ϭ 3 mice/treatment Ϯ SE. C: Western analysis of ZIP14, lipolytic marker hormone-sensitive lipase (HSL), and differentiation
marker PPAR␥ (peroxisome proliferator-activated receptor-␥) during the acute-phase response in WAT. Each lane is a pooled lysate of 3 mice/treatment. *P Ͻ
0.05; **P Ͻ 0.01; ***P Ͻ 0.0001. P-IR, phosphorylated insulin receptor.
Fig. 1. Expression of Zip14 in white adipose tissue (WAT) is induced by lipopolysaccharide (LPS). A: tissue expression profile of Zip14 transcripts. Transcript
abundance was determined by quantitative PCR, starting with an equal amount of total RNA from each tissue. B: fold change in Zip14 tissue expression 18 h
post-intraperitoneal (ip) injection of LPS (2 mg/kg). C: of the WAT zinc transporters significantly (P Յ 0.05) impacted by LPS, Zip14 had the highest induction.
For all graphs, n ϭ 5 mice/genotype Ϯ SE. DUO, duodenum.
E260 ZINC TRANSPORTER Zip14 AND ADIPOSE TISSUE REGULATION
AJP-Endocrinol Metab • doi:10.1152/ajpendo.00421.2015 • www.ajpendo.org
normalized to cell number. To generate confocal images, 3T3-L1 cells
were passaged into two-well chambered coverglass (Thermo Fisher
Scientific) prior to siRNA treatment and a subsequent 8-day differ-
entiation period. Imaging was performed at the University of Florida
Cell & Tissue Analysis Core with a Leica TCS SP5 laser-scanning
confocal microscope, using a ϫ40 oil objective. LAS-AF imaging
software was used for image analysis.
Statistics. Statistical analyses was performed using SAS verison 9.2
(SAS Institute, Cary, NC). The effects of WT vs. KO genotype were
compared using Student’s t-tests. The independent effects of genotype
and antibiotic were analyzed using the Proc Mixed procedure (SAS)
with mouse within treatment as the random effect. Multiple compar-
ison significance was analyzed using the Tukey adjustment. Reported
values represent means Ϯ SE.
RESULTS
Zip14 is highly expressed in WAT during acute inflammation.
Relative Zip14 mRNA abundance in WAT of WT mice is low
compared with that found in duodenum or liver but greater than
that found in muscle (Fig. 1A). Aydemir et al. (3) demonstrated
that the highest induction of liver Zip14 mRNA occurred 18 h
post-ip injection of LPS. Therefore, the 18-h time point was
used to examine the comparative expression of Zip14 across
tissues during LPS challenge (Fig. 1B). Expression of Zip14
mRNA increased two- and 32-fold in liver and WAT, respec-
tively, with LPS administration. A transcript screening of 24
zinc transporters was performed. The transporter mRNAs that
were significantly (P Յ 0.05) altered by LPS are shown (Fig.
1C). With the exception of ZnT3 and ZnT8, all zinc transporter
genes were expressed in WAT. Zip14 had the greatest LPS
induction of the zinc transporters.
LPS-induced Zip14 expression is correlated with upregu-
lated cytokine expression. To compare WAT with our previous
liver findings (3), we next sought to evaluate the induction of
WAT ZIP14 during acute LPS challenge. The endotoxin-
induced acute phase response was shown through increased
expression of Il-6, Tnf␣, and Il-1␤ mRNA (Fig. 2A). The
induction of Zip14 expression peaked at 6 h post-ip injection
and preceded the 9-h peaks of cytokine transcripts (Fig. 2A).
Fig. 3. Zip14-knockout (KO) mice have enhanced levels of circulating endotoxin, predicating an inflammatory state characterized by adipose hypertrophy and
dampened insulin signaling. A: colorimetrically determined that serum endotoxin was higher in Zip14-KO mice; n ϭ 3/genotype Ϯ SE (left). Proinflammatory
signaling pathways in wild-type (WT) and KO mice are shown in WAT (right). Pooled lysates are depicted; lanes are triplicate repeats. B: expression of cytokines
Il-6, Tnf␣, Il-1␤, and adiponectin (Adpn) in WT and KO mice. Preadipocyte marker [plasminogen activator inhibitor-1 (Pai-1) and preadipocyte factor-1 (Pref-1)]
and differentiation marker (Ppar␥) are also depicted; n ϭ 28 mice/genotype Ϯ SE. C: serum and WAT leptin concentrations in WT and KO mice; n ϭ 3
mice/genotype Ϯ SE. ELISA values from WAT were normalized to total protein. D: representative hematoxylin and eosin (H & E) images of WAT from WT
and KO adipose. Cell areas are from n ϭ 10 images/genotype Ϯ SE. Bars, 150 ␮m. E: transcripts for key adipogenic enzymes (n ϭ 28 mice/genotype Ϯ SE)
and Western analysis of lipolytic markers (n ϭ 3 mice/genotype). F: Western analysis of insulin signaling (n ϭ 3 mice/genotype). MyD88, myeloid
differentiation primary response gene; mTOR, mammalian target of rapamycin; ZAG, zinc ␣2-glycoprotein.
E261ZINC TRANSPORTER Zip14 AND ADIPOSE TISSUE REGULATION
AJP-Endocrinol Metab • doi:10.1152/ajpendo.00421.2015 • www.ajpendo.org
Serum hypozincemia occurred as expected (3, 5); however,
adipose levels of zinc fluctuated over the course of the LPS
challenge (Fig. 2B). This finding suggests that a redistribution
of zinc occurs within adipose during inflammation. The up-
regulation of cytokine mRNA transcripts coincided with in-
creased ZIP14 protein levels, as shown in the Western blots
from adipose lysates (Fig. 2C). In adipose, the acute-phase
response is associated with increased markers of lipolysis and
decreased markers of differentiation (14, 23, 64). Our data
confirmed findings of others in that the phosphorylation of
HSL (HSL660) was upregulated during endotoxemia. In con-
trast, phosphorylated insulin receptor and PPAR␥ were down-
regulated, which suggested a loss of adipocyte differentiation.
Zip14 KO mice have hypertrophy of adipocytes and greater
leptin production. It is apparent that ZIP14 is highly induced
by endotoxin in WAT. Previously, we established that global
Zip14-KO impaired intestinal barrier function, which precipi-
tates systemic endotoxemia (25). Therefore, we hypothesized
that systemic endotoxemia would enhance the inflammatory
status of adipose tissue. In confirmation of our previous find-
ings, plasma endotoxin levels were greater in KO mice (Fig.
3A). We first sought to characterize the phenotypic profile of
KO adipose. Enhanced expression of MyD88 (an adaptor
protein that facilitates LPS induction of IL-6), NF-␬B, and
STAT3 is indicative of WAT inflammation in the KO mice
(Fig. 3A, right). In agreement, the steady-state levels of Il-6,
Tnf␣, and Il-1␤ mRNAs were greater in WAT from the KO
mice (Fig. 3B). Similarly, upregulation of Pref-1 (29, 58) and
Pai-1 (35) mRNAs combined with downregulation of Ppar␥ in
the WAT suggests a proinflammatory state, a characteristic of
preadipocytic cells (Fig. 3B). Adiponectin (Adpn) expression, a
hormone positively correlated with insulin sensitivity, was
reduced with KO. Leptin protein levels were significantly (P Ͻ
0.05) greater in both KO serum and WAT (Fig. 3C). Moreover,
cell areas of KO adipocytes were on average 40% larger than
WT adipocytes (Fig. 3D). KO adipose had increased expres-
sion of key adipogenic enzyme transcripts and altered lipid
homeostasis (Fig. 3E). Compared with WT mice, steady-state
lipolysis markers, p-HSL (HSL660), and ZAG (39) were re-
duced in WAT from KO mice. The observed dyslipidemia was
accompanied by reduced PPAR␥ levels (Fig. 3E). Overall, the
KO mutation appeared to enhance adiposity and depress adi-
pocyte differentiation. An evaluation of steady-state insulin
signaling revealed that KO WAT had decreased phosphoryla-
Fig. 4. Zip14 KO enhances LPS-induced inflammation. The effects of the KO are reversed with oral antibiotics (AB). A: WT and KO mice were administered
LPS (2 mg/kg ip) for 6 h. Zip14, Tnf␣, and Il-1␤ mRNAs were measured by quantitative PCR. Serum and WAT IL-6 concentrations were measured by ELISA;
n ϭ 3 mice/treatment Ϯ SE. In the absence of LPS, baseline IL-6 levels in either genotype were not detected (ND). B: WT and KO mice were provided AB
in drinking water (0.5 mg/ml neomycin and 1 mg/ml ampicillin) for 4 wk. WAT expression of Il-6, Tnf␣, and Il-1␤ mRNAs in WT and KO mice with AB
treatment; n ϭ 5 mice/treatment Ϯ SE. C: H & E images of WAT were used to measure cell areas; n ϭ 10 images/treatment Ϯ SE. Bars, 150 ␮m. D: Western
analysis of WAT insulin signaling in WT and KO mice with AB treatment; n ϭ 3 mice/treatment.
E262 ZINC TRANSPORTER Zip14 AND ADIPOSE TISSUE REGULATION
AJP-Endocrinol Metab • doi:10.1152/ajpendo.00421.2015 • www.ajpendo.org
tion of the insulin receptor (IR), protein kinase B (Akt), and
mTOR (Fig. 3F) (9). These data show that KO adipose dem-
onstrates a phenotype of dampened insulin signaling. Further-
more, data in Fig. 3F also show that the macrophage marker
(F4/80) is not different between genotypes, suggesting minimal
influence of macrophage infiltration (59) on our observed KO
phenotype.
The Zip14 phenotype is enhanced with LPS challenge and
minimized with oral antibiotics. Given the systemic endotox-
emia in the KO, we hypothesized that the phenotype would be
exacerbated by acute LPS and improved with antibiotics (AB).
The highest induction of Zip14 mRNA during our LPS time
course occurred 6 h post-ip injection (Fig. 1C). Therefore, we
used the 6-h LPS response as the point of comparison between
genotypes. The loss of Zip14 caused higher induction of WAT
cytokine transcripts (Fig. 4A, left) along with serum IL-6 (Fig.
4A, right). These findings suggest that ZIP14 may serve as a
negative regulator of cytokine induction. In contrast to the
exacerbating effects of LPS, the impact of endotoxin-induced
inflammation was reduced with AB. Expression of Il-6 and
Tnf␣ mRNAs in KO WAT was reduced with AB (Fig. 4B).
Furthermore, AB prevented the adipocyte hypertrophy seen in
KO WAT (Fig. 4C). The AB treatment also normalized insulin
signaling between the genotypes (Fig. 4D).
Upregulated cytokine pathways due to the KO mutation lead
to impaired capacity to differentiate. In an effort to eliminate
the effect of circulating endotoxin on KO adipose, mesenchy-
mal stem cells were cultured in the absence of LPS. Stem cells
were derived from WT and KO ears and differentiated into
adipocytes. Zip14-KO was confirmed with quantitative PCR
(Fig. 5A). We hypothesized that KO cells would exhibit en-
hanced steady-state cytokine signaling in culture. Cytokine
mRNA expression (Il-6 and Il-1␤) was higher in fully differ-
entiated KO cells. Mt1 expression was not significantly differ-
ent between genotype but tended to be lower in KO cells.
Additionally, KO preadipocytic markers Pref-1 (29, 58) and
Pai-1 (35) were upregulated (Fig. 5A). Western analysis of
both PPAR␥ and PREF-1 (Fig. 5B) confirmed our KO tissue
findings (Figs. 3, B and E, and 4D). Although the same
antibody was used for cells and tissue, persistent bands appear
in the Western blots of primary mesenchymal stem cells (Fig.
5B). However, this same persistent banding was again noted in
siRNA-treated 3T3-L1 cells, which may serve as our negative
control for cellular ZIP14 expression (Fig. 6F). Oil Red O
staining also demonstrated that cultured KO cells are more
preadipocytic and have a lower capacity to differentiate and
accumulate lipids (Fig. 5C, top). It was not until cells were
cultured in LPS that KO cells began to overproduce lipids (Fig.
Fig. 5. Stem cells from Zip14-KO mice are inflammatory and preadipocytic in culture. Primary stem cells from WT and KO mice were cultured in 3T3-L1 growth
media. Upon confluence, cells were placed in differentiation medium and cultured for 10 days. A: transcript levels of Zip14, cytokine (Il-6, Tnf␣, and Il-1␤), Mt1,
and preadipocytic markers (Pref-1 and Pai-1) were measured; n ϭ 3 pooled wells Ϯ SE. B: Western analysis of the preadipocytic markers in WT and KO cells;
n ϭ 3 pooled wells Ϯ SE. C: accumulation of lipids in WT- and KO-derived cells, as measured by Oil Red O (Ϯ 10 ng/ml LPS for 10 days). D: Oil Red O
was eluted from cell cultures and measured colorimetrically and normalized to protein; n ϭ 3 wells/treatment Ϯ SE.
E263ZINC TRANSPORTER Zip14 AND ADIPOSE TISSUE REGULATION
AJP-Endocrinol Metab • doi:10.1152/ajpendo.00421.2015 • www.ajpendo.org
5C, bottom). Quantification of Oil Red O confirmed our visual
observations (Fig. 5D).
ZIP14-KO alters intracellular zinc, which enhances key
inflammatory pathways. Since the focus of the molecular site
responsible for the KO phenotype is likely related to metal
transport, it was necessary to place those findings within a
function of dyshomeostasis of cellular zinc metabolism. 65
Zn
was administered orally to measure zinc uptake/retention. KO
mice accumulated more zinc (65
Zn) in WAT. Figure 6A shows
that Mt1 mRNA expression, a surrogate measure of cytosolic
zinc, is reduced. Adipogenic 3T3-L1 cells transfected with
siRNA had a pattern of zinc distribution similar to KO tissue.
Specifically Zip14 knockdown increased total zinc in cells,
whereas Mt1 mRNA was decreased (Fig. 6B). Intracellular
labile zinc was visualized with a zinc probe, fluozin-3 AM.
Fluorescence was detected using laser-scanning confocal mi-
croscopy (Fig. 6C). The silencing of Zip14 with siRNA clearly
resulted in increased (P Ͻ 0.0001) vesicular zinc. Transfection
with siRNA increased expression of ZIP8, a transporter that is
a homologue of ZIP14 (Fig. 6D) (31). These in vitro data are
compatible with the hypothesis that less functional cellular zinc
is available in adipocytes of KO mice. This finding is indica-
tive of the “zinc trap” hypothesis associated with Zip14 abla-
tion that we advanced earlier (25) in mouse intestine. Zip14
siRNA clearly resulted in increased expression of Il-6 and Tnf␣
(P Ͻ 0.05) and to a lesser extent Il-1␤ (Fig. 6E).
Zinc’s ability to inhibit key cytokine pathways is one way in
which zinc may regulate cytokine expression. We hypothe-
sized that zinc trapped within vesicles, through either Zip14
ablation in mice or with siRNA transfection in cells, would
limit the inhibition of key proinflammatory signaling cascades
by zinc. With the exception of A20 (11), Zip14 knockdown
enhanced activation of both the JAK2/STAT3 and NF-␬B
pathways in 3T3-L1 cells (Fig. 6F). Upregulation of these
pathways depressed the ability of cells to differentiate (upregu-
lation of PREF-1 expression) and increase cytokine expression.
As shown in the model presented in Fig. 7, zinc transported by
ZIP14 appears to impact the TLR4 pathway as early as MyD88
and also influences signaling further downstream, e.g., phos-
phorylated I␬B␣ (p-I␬B␣) and NF-␬B. Zip14 knockdown
Fig. 6. Zip14-KO adipose and 3T3-L1 adipocytes treated with Zip14 siRNA display altered zinc homeostasis and enhanced proinflammatory signaling though
the NF-␬B and JAK2/STAT3 pathways. A: WT and KO mice received 65
Zn by gavage, and WAT zinc accumulation was calculated from 65
Zn-specific activity.
Mt1 mRNA was measured as an index of available intracellular zinc; n ϭ 10 mice/genotype Ϯ SE. B: 3T3L1 cells were transfected with Zip14 siRNA. Total
zinc (AAS) and Mt1 mRNA; n ϭ 3 plates/group replicated 3 times. C: representative laser-scanning confocal images used to detect labile zinc pools using
FluoZin-3 AM fluorescence. FluoZin-3 AM was quantified and normalized to cell count; n ϭ 24 wells/group replicated twice. D: representative Western blots
show the effect of Zip14 siRNA on ZIP14 and ZIP8 (n ϭ 2 replicated 3 times). E: relative mRNA expression of Zip14, Il-6, Tnf␣, and Il-1␤ in Zip14
siRNA-transfected 3T3-L1 cells Ϯ 10 ng/ml LPS (n ϭ 3 wells/group Ϯ SE). F: representative Western blots show the effect of Zip14 siRNA on proinflammatory
signaling pathways (n ϭ 4 replicated 3 times). SCR, scrambled; RFU, relative fluorescence units.
E264 ZINC TRANSPORTER Zip14 AND ADIPOSE TISSUE REGULATION
AJP-Endocrinol Metab • doi:10.1152/ajpendo.00421.2015 • www.ajpendo.org
limits cytosolic availability of zinc, and inhibition of NF-␬B is
lifted, allowing enhanced induction of proinflammatory genes
Il-6, Tnf␣, and Il-1␤ (Fig. 6E). Similarly, Zip14 deficiency
enhanced JAK2 and STAT3 activity (Fig. 3A, right) with a
concomitant stimulation of the Il-6 transcript (Fig. 3B) that
could in turn execute an autocrine response leading to in-
creased leptin production and secretion (Fig. 3C).
DISCUSSION
Here we show for the first time that deletion of Zip14
impacts adipose function. Although Zip14 was first cloned
from adipogenic 3T3-L1 cells (53), little has been reported
regarding its expression/function in adipose metabolism. In
this report, it is shown that KO alters zinc signaling pathways,
intracellular zinc trafficking, and ultimately adipocyte metab-
olism.
Inflammation, expansion of tissue mass, and recruitment of
inflammatory cells are normal functions of healthy adipose (49,
60). However, a prolonged inflammatory response will even-
tually lead to metabolic alterations within WAT. Acute inflam-
mation impacts ZIP14 expression in a tissue-specific manner
(3, 25). From the experiments described to this point, it has
been established that adipose ZIP14 is highly responsive to
LPS. Zip14 transcript abundance was upregulated 30-fold by
18 h after LPS injection, suggesting a critical role for ZIP14 in
adipose inflammatory response. These data suggest that up-
regulation of Zip14 expression in WAT is kinetically more
rapid than liver expression (3). Furthermore, Zip14 ablation
drastically influences cytokine and leptin production along
with signaling pathway activity in WAT. Upregulated Il-6
production with KO may be responsible for the induction of
leptin secretion (55). We interpret these responses as being
indicative of a greater requirement for ZIP14 and/or zinc
during inflammatory challenge (43). The reversal of the KO
phenotype with antibiotics supports this hypothesis.
The findings with antibiotics are noteworthy in that the
treatment was a cocktail of neomycin and ampicillin. Neomy-
cin in particular, is poorly absorbed through the gastrointestinal
tract (15). Therefore, these antibiotics target intestinal micro-
flora populations directly, a finding that suggests that intestinal
microflora (along with any treatment that alters the microfloral
load, dietary or otherwise) impacts adipose development and
function. In fact, the tie between microflora and adipose has
been established previously by Ley et al. (30). The impact of
KO on the intestinal microfloral load is certainly relevant
considering the proposed links between the microbiome and
metabolic diseases, a finding that we plan to evaluate in further
studies. Relevant to this report is that the in vivo phenotype
was successfully modeled in vitro using LPS alone, confirming
that KO predisposes cells to an inflammatory phenotype that is
exacerbated with LPS.
Increased intestinal permeability increased serum endotoxin
in KO mice (25). This finding was of particular interest, as
chronic exposure to endotoxin leads to metabolic dysfunction.
Metabolic endotoxemia is characterized by atypically high
levels of circulating endotoxin, which produces low-grade,
systemic inflammation (10, 34, 46). This inflammation has
far-reaching physiological implications, including increased
weight gain, hepatic insulin resistance (12, 26, 37), and mac-
rophage recruitment (52, 59), all of which precede the devel-
opment of type 2 diabetes upon the consumption of a high-fat
diet. Luche et al. (35) found that a 28-day LPS pretreatment
caused mice consuming high-fat diets to gain more weight than
cohorts who had not received the LPS conditioning. The
reported increase in body weight was coupled with an in-
creased fat/lean ratio, an influx of small inflammatory preadi-
Fig. 7. Proposed model for increased IL-6
and leptin production caused by global
Zip14 deletion. Increased cytosolic zinc due
to ZIP14 inhibits cytokine signaling. Sys-
temic endotoxemia and/or LPS administration
leads to Toll-like receptor 4 (TLR4) activation
and induction of the NF-␬B pathway. Without
surface expression of ZIP14, zinc is trapped in
intracellular compartments, where it is unable
to inhibit activities of the NF-␬B and JAK2/
STAT3 pathways, thereby increasing IL-6 and
leptin production.
E265ZINC TRANSPORTER Zip14 AND ADIPOSE TISSUE REGULATION
AJP-Endocrinol Metab • doi:10.1152/ajpendo.00421.2015 • www.ajpendo.org
pocytes, and an overall higher glycemic index (35). Unique to
our KO model, dampened insulin signaling and low-grade
inflammation were coupled with hypertrophy. Enhanced mark-
ers of lipogenesis and decreased markers of lipolysis (8, 38)
were also noted with KO. These markers are suggestive of a
chronic (vs. acute; see Ref. 61) inflammatory state in KO
WAT. Furthermore, these physiological differences occur in
the absence of a dietary intervention such as the feeding of a
high-fat diet.
Inflammatory cytokines are well-known mediators of altered
adipose function and development (12, 48). Furthermore, prea-
dipocytes themselves are able to acquire phagocytic activity
and express macrophage-specific antigens under acute inflam-
matory conditions (13). Using cultures at tiered stages of
differentiation (0, 50, and 90%), Chung et al. (14) elegantly
demonstrated that preadipocytes were the primary producers of
the endogenous cytokines responsible for endotoxin-induced
suppression of insulin-stimulated glucose uptake. It has been
shown previously that Zip14 mRNA is induced during the early
stages of adipocyte differentiation (53). Here, we show that KO
increased preadipocytic markers and enhanced cytokine ex-
pression. The differentiation marker PPAR␥ was downregu-
lated with KO. This finding is relevant, as PPAR␥ is zinc
responsive and has the ability to inactivate STAT3 in myeloma
cells (38, 56, 57). Zip14 KO limits the ability of mesenchymal
stem cells to differentiate and accumulate lipids (Fig. 4).
Although this may initially seem counterintuitive, limiting a
preadipocyte’s ability to differentiate precipitates hypertrophic
obesity in humans (24, 49). Hypertrophic obesity is the likely
cause of our previous finding where KO mice had enhanced
liver lipids (3).
Aberrant zinc signaling with Zip14 KO impacts the
growth and development of adipocytes. Here, we report a
phenotype characterized by high tissue zinc but low Mt1
expression. Trayhurn et al. (54) found that adipose expres-
sion of Mt1 was not influenced by subcutaneous zinc injec-
tion. However, experiments conducted with metallothion-
ein-KO (7) revealed that MtϪ/Ϫ
mice had increased visceral
WAT weight coupled with adipocyte hypertrophy and
higher levels of circulating leptin (7). Furthermore, this
phenotype was exacerbated by high-fat feeding (44). Previ-
ous literature has reported conflicting relationships between
serum leptin and fat pad zinc content (2, 41, 51). In humans,
obesity has long been associated with hypozincemia. In
terms of zinc transporters, obesity is correlated with down-
regulated ZnT (Ϫ2, Ϫ3, Ϫ6, and Ϫ8) and ZIP (Ϫ1, Ϫ2, Ϫ3,
Ϫ4, Ϫ5, Ϫ6, and Ϫ7) expression in subcutaneous adipose
(47). Our data shows that Zip14 deletion causes adipose to
act as if it were experiencing a zinc deficiency. Evidence for
that includes the lower Mt1 mRNA levels in WAT of the KO
mice (Fig. 6A). This zinc-deficient status apparently cannot
be overcome by the concomitant upregulation of zinc trans-
porters, most notably ZIP8 (Fig. 6D). The ability of adi-
pocytes to regulate zinc trafficking may determine how
adipose is remodeled during tissue expansion. Furthermore,
ZIP14 appears to lie at the plastic interface of adipose
metabolism and inflammation.
As shown in Fig. 7, our data show that ZIP14 influences
adipocyte cytokine expression through two well-established
signaling pathways, NF-␬B and JAK2/STAT3 (6, 34, 55, 63).
The adipose mass expresses TLR4 (45) and is a major secre-
tory organ of both leptin and IL-6 (28). Hence, the demonstra-
tion here that both are overproduced with Zip14 deletion points
to ZIP14 acting to provide control over the production of both
mediators. Zip14 deletion enhances phosphorylation of these
signaling pathways and is a likely cause for the upregulation of
cytokines and leptin observed with KO. It should be noted that
the ZIP14 homolog ZIP8 has been shown to inhibit the TLR4
pathway though inhibition of I␬B kinase (IKK) (32). In this
report, ZIP8 levels are greater with Zip14 ablation, demonstrat-
ing that ZIP8 is unable to compensate for the upregulation of
the NF-␬B pathway. Therefore, ZIP8-mediated inhibition of
IKK may be of minor significance in our Zip14-KO adipose
model.
In summary, adipocytes with a Zip14 deletion appear to have
an impaired ability to mobilize intracellular zinc. In our model,
limited availability of intracellular zinc disinhibits key cyto-
kine pathways. Adipocytes have enhanced cytokine signaling
and are more preadipocytic with KO. ZIP14 knockdown cells
did not produce excess lipids until they were cultured in LPS
media. These results in part model hypertrophic WAT ob-
served with KO in vivo. Furthermore, the KO phenotype of
hypertrophy was diminished with oral antibiotics, indicating
that low-grade inflammation is necessary to induce enhanced
adiposity. Our results demonstrate that ZIP14 is critical to
cytokine production in adipose tissue and that targeted zinc
transport is critical to adipocyte development.
ACKNOWLEDGMENTS
Scanning confocal microscopy was performed at the University of Florida
Core. We thank Dr. Joseph L. Purswell, US Department of Agriculture/
Agricultural Research Service at Mississippi State University, for assistance
with the ImageJ analysis.
GRANTS
This project was supported by National Institute of Diabetes and Digestive
and Kidney Diseases Grant R01-DK-094244 and the Boston Family Endow-
ment Funds of the University of Florida to R. J. Cousins.
DISCLOSURES
The authors report no conflicts of interest, financial or otherwise.
AUTHOR CONTRIBUTIONS
C.T., T.B.A., and R.J.C. conception and design of research; C.T. and T.B.A.
performed experiments; C.T. analyzed data; C.T., T.B.A., and R.J.C. inter-
preted results of experiments; C.T. prepared figures; C.T., T.B.A., and R.J.C.
drafted manuscript; C.T., T.B.A., and R.J.C. edited and revised manuscript;
R.J.C. approved final version of manuscript.
REFERENCES
1. Allred CC, Krennmayer T, Koutsari C, Zhou L, Ali AH, Jensen MD.
A novel ELISA for measuring CD36 protein in human adipose tissue. J
Lipid Res 52: 408–415, 2011.
2. Argani H, Mahdavi R, Ghorbani-haghjo A, Razzaghi R, Nikniaz L,
Gaemmaghami SJ. Effects of zinc supplementation on serum zinc and
leptin levels, BMI and body composition in hemodialysis patients. J Trace
Elem Med Biol 28: 35–38, 2014.
3. Aydemir TB, Chang SM, Guthrie GJ, Maki AB, Ryu MS, Karabiyik
A, Cousins RJ. Zinc transporter ZIP14 functions in hepatic zinc, iron and
glucose homeostasis during the innate immune response (endotoxemia).
PLoS One 7: e48679, 2012.
4. Aydemir TB, Liuzzi JP, McClellan S, Cousins RJ. Zinc transporter
ZIP8 (SLC39A8) and zinc influence IFN-␥ expression in activated human
T cells. J Leukoc Biol 86: 337–348, 2009.
5. Aydemir TB, Sitren HS, Cousins RJ. The zinc transporter Zip14 Influ-
ences c-Met phosphorylation and hepatocyte proliferation during liver
regeneration in mice. Gastroenterology 142: 1536–1546, 2012.
E266 ZINC TRANSPORTER Zip14 AND ADIPOSE TISSUE REGULATION
AJP-Endocrinol Metab • doi:10.1152/ajpendo.00421.2015 • www.ajpendo.org
6. Baker RG, Hayden MS, Ghosh S. NF-␬B, inflammation, and metabolic
disease. Cell Metab 13: 11–22, 2011.
7. Beattie JH, Wood AM, Newman AM, Bremner I, Choo KH, Michal-
ska AE, Trayhurn P. Obesity and hyperleptinemia in metallothionein (-I
and -II) null mice. Proc Natl Acad Sci USA 95: 358–363, 1998.
8. Bing C, Bao Y, Jenkins J, Sanders P, Manieri M, Cinti S, Tisdale MJ,
Trayhurn P. Zinc-alpha2-glycoprotein, a lipid mobilizing factor is ex-
pressed in adipocytes and is up-regulated in mice with cancer cachexia.
Proc Natl Acad Sci USA 101: 2500–2505, 2004.
9. Blagosklonny MV. TOR-centric view on insulin resistance and diabetic
complications: perspective for endocrinologists and gerontologists. Cell
Death Dis 4: e964, 2013.
10. Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D,
Neyrinck AM, Fava F, Tuohy KM, Chabo C, Waget A, Delmée E,
Cousin B, Sulpice T, Cani PD, Bibiloni R, Knauf C, Waget A,
Neyrinck AM, Delzenne NM, Burcelin R. Changes in gut microbiota
control metabolic endotoxemia-induced inflammation in high-fat diet-
induced obesity and diabetes in mice. Diabetes 57: 1470–1481, 2008.
11. Catrysse L, Vereecke L, Beyaert R, van Loo G. A20 in inflammation
and autoimmunity. Trends Immunol 35: 23–31, 2013.
12. Chamontin B, Ferrières J, Tanti JF, Gibson GR, Casteilla L, Delzenne
NM, Alessi MC, Burcelin R. Metabolic endotoxemia initiates obesity and
insulin resistance. Diabetes 56: 1761–1772, 2007.
13. Charrière G, Cousin B, Arnaud E, André M, Bacou F, Pénicaud L,
Casteilla L. Preadipocyte conversion to macrophage. Evidence of plas-
ticity. J Biol Chem 278: 9850–9855, 2003.
14. Chung S, Lapoint K, Martinez K, Kennedy A, Boysen Sandberg M,
McIntosh MK. Preadipocytes mediate lipopolysaccharide-induced in-
flammation and insulin resistance in primary cultures of newly differen-
tiated human adipocytes. Endocrinology 147: 5340–5351, 2006.
15. Croswell A, Amir E, Teggatz P, Barman M, Salzman NH. Prolonged
impact of antibiotics on intestinal microbial ecology and susceptibility to
enteric Salmonella infection. Infect Immun 77: 2741–2753, 2009.
16. Coulston L, Dandona P. Insulin-like effect of zinc on adipocytes.
Diabetes 29: 665–667, 1980.
17. Cui W, Taub DD, Gardner K. qPrimerDepot: a primer database for
quantitative real time PCR. Nucleic Acids Res 35: D805–D809, 2007.
18. de Luis DA, Pacheco D, Izaola O, Terroba MC, Cuellar L, Martin T.
Zinc and copper serum levels of morbidly obese patients before and after
biliopancreatic diversion: 4 years of follow-up. J Gastrointest Surg 15:
2178–2181, 2011.
19. de Luis DA, Pacheco D, Izaola O, Terroba MC, Cuellar L, Cabezas G.
Micronutrient status in morbidly obese women before bariatric surgery.
Surg Obes Relat Dis 9: 323–327, 2013.
20. Drolet R, Richard C, Sniderman AD, Mailloux J, Fortier M, Huot C,
Rhéaume C, Tchernof A. Hypertrophy and hyperplasia of abdominal
adipose tissues in women. Int J Obes (Lond) 32: 283–291, 2008.
21. Ferrante AW Jr. The immune cells in adipose tissue. Diabetes Obes
Metab 15, Suppl 3: 34–38, 2013.
22. Galarraga M, Campión J, Muñoz-Barrutia A, Boqué N, Moreno H,
Martínez JA, Milagro F, Ortiz-de-Solórzano C. Adiposoft: automated
software for the analysis of white adipose tissue cellularity in histological
sections. J Lipid Res 53: 2791–2796, 2012.
23. Grisouard J, Bouillet E, Timper K, Radimerski T, Dembinski K, Frey
DM, Peterli R, Zulewski H, Keller U, Müller B, Christ-Crain M. Both
inflammatory and classical lipolytic pathways are involved in lipopoly-
saccharide-induced lipolysis in human adipocytes. Innate Immun 18:
25–34, 2012.
24. Gustafson B, Gogg S, Hedjazifar S, Jenndahl L, Hammarstedt A,
Smith U. Inflammation and impaired adipogenesis in hypertrophic obesity
in man. Am J Physiol Endocrinol Metab 297: E999–E1003, 2009.
25. Guthrie GJ, Aydemir TB, Troche C, Martin AB, Chang SM, Cousins
RJ. Influence of ZIP14 (slc39A14) on intestinal zinc processing and
barrier function. Am J Physiol Gastrointest Liver Physiol 308: G171–
G178, 2015.
26. Henninger AM, Eliasson B, Jenndahl LE, Hammarstedt A. Adipocyte
hypertrophy, inflammation and fibrosis characterize subcutaneous adipose
tissue of healthy, non-obese subjects predisposed to type 2 diabetes. PLoS
One 9: e105262, 2014.
27. Jeong J, Eide DJ. The SLC39 family of zinc transporters. Mol Aspects
Med 34: 612–619, 2013.
28. Kershaw E, Flier J. Adipose tissue as an endocrine organ. J Clin
Endocrinol Metab 89: 2548–2556, 2004.
29. Kim YJ, Min TS, Seo KS, Kim SH. Expression of pref-1/dlk-1 is
regulated by microRNA-143 in 3T3-L1 cells. Mol Biol Rep 42: 617–624,
2015.
30. Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Microbial ecology: human
gut microbes associated with obesity. Nature 444: 1022–1023, 2006.
31. Lichten LA, Cousins RJ. Mammalian zinc transporters: nutritional and
physiologic regulation. Annu Rev Nutr 29: 153–176, 2009.
32. Liu MJ, Bao S, Gálvez-Peralta M, Pyle CJ, Rudawsky AC, Pavlovicz
RE, Killilea DW, Li C, Nebert DW, Wewers MD, Knoell DL. ZIP8
regulates host defense through zinc-mediated inhibition of NF-␬B. Cell
Rep 3: 386–400, 2013.
33. Liuzzi JP, Aydemir F, Nam H, Knutson MD, Cousins RJ. Zip14
(Slc39a14) mediates non-transferrin-bound iron uptake into cells. Proc
Natl Acad Sci USA 103: 13612–13617, 2006.
34. Liuzzi JP, Lichten LA, Rivera S, Blanchard RK, Aydemir TB, Knut-
son MD, Ganz T, Cousins RJ. Interleukin-6 regulates the zinc transporter
Zip14 in liver and contributes to the hypozincemia of the acute-phase
response. Proc Natl Acad Sci USA 102: 6843–6848, 2005.
35. Luche E, Cousin B, Garidou L, Serino M, Waget A, Barreau C, André
M, Valet P, Courtney M, Casteilla L, Burcelin R. Metabolic endotox-
emia directly increases the proliferation of adipocyte precursors at the
onset of metabolic diseases through a CD14-dependent mechanism. Mol
Metab 2: 281–291, 2013.
36. May J, Contoreggi C. The mechanism of the insulin-like effects of ionic
zinc. J Biol Chem 257: 4362–4368, 1982.
37. Makki K, Froguel P, Wolowczuk I. Adipose tissue in obesity-related
inflammation and insulin resistance: cells, cytokines, and chemokines.
ISRN Inflamm 2013: 139239, 2013.
38. Meerarani P, Reiterer G, Toborek M, Hennig B. Zinc modulates
PPARgamma signaling and activation of porcine endothelial cells. J Nutr
133: 3058–3064, 2003.
39. Mracek T, Gao D, Tzanavari T, Bao Y, Xiao X, Stocker C, Trayhurn
P, Bing C. Downregulation of zinc-{alpha} 2-glycoprotein in adipose
tissue and liver of obese ob/ob mice and by tumour necrosis factor-alpha
in adipocytes. J Endocrinol 204: 165–172, 2010.
40. O’Hara A, Lim F, Mazzatti DJ, Trayhurn P. Microarray analysis
identifies matrix metalloproteinases (MMPs) as key genes whose expres-
sion is up-regulated in human adipocytes by macrophage-conditioned
medium. Pflugers Arch 458: 1103–1114, 2009.
41. Ott ES, Shay NF. Zinc deficiency reduces leptin gene expression and
leptin secretion in rat adipocytes. Exp Biol Med (Maywood) 226: 841–846,
2001.
42. Rim J, Mynatt R, Gawronska-Kozak B. Mesenchymal stem cells from
the outer ear: a novel adult stem cell model system for the study of
adipogenesis. FASEB J 19: 1205–1207, 2005.
43. Rink L, Haase H. Zinc homeostasis and immunity. Trends Immunol 28:
1–4, 2007.
44. Sato M, Kawakami T, Kondoh M, Takiguchi M, Kadota Y, Himeno S,
Suzuki S. Development of high-fat-diet-induced obesity in female metal-
lothionein-null mice. FASEB J 24: 2375–2384, 2010.
45. Shi H, Kokoeva MV, Inouye K, Tzameli I, Yin H, Flier JS. TLR4 links
innate immunity and fatty acid-induced insulin resistance. J Clin Invest
116: 3015–3025, 2006.
46. Siebler J, Galle PR, Weber MM. The gut-liver-axis: endotoxemia,
inflammation, insulin resistance and NASH. J Hepatol 48: 1032–1034,
2008.
47. Smidt K, Pedersen SB, Brock B, Schmitz O, Fisker S, Bendix J,
Wogensen L, Rungby J. Zinc-transporter genes in human visceral and
subcutaneous adipocytes: lean versus obese. Mol Cell Endocrinol 264:
68–73, 2007.
48. Song MJ, Kim KH, Yoon JM, Kim JB. Activation of Toll-like receptor
4 is associated with insulin resistance in adipocytes. Biochem Biophys Res
Commun 346: 739–745, 2006.
49. Stefan N, Kantartzis K, Machann J, Schick F, Thamer C, Rittig K,
Balletshofer B, Machicao F, Fritsche A, Haring HU. Identification and
characterization of metabolically benign obesity in humans. Arch Intern
Med 168: 1609–1616, 2008.
50. Suliburska J, Bogdanski P, Pupek-Musialik D, Krejpcio Z. Dietary
intake and serum and hair concentrations of mineral and their relationship
with serum lipids and glucose levels in hypertensive and obese patients
with insulin resistance. Biol Trace Elem Res 139: 137–150, 2011.
51. Tallman DL, Taylor CG. Effects of dietary fat and zinc on adiposity,
serum leptin and adipose fatty acid composition in C57BL/6J mice. J Nutr
Biochem 14: 17–23, 2003.
E267ZINC TRANSPORTER Zip14 AND ADIPOSE TISSUE REGULATION
AJP-Endocrinol Metab • doi:10.1152/ajpendo.00421.2015 • www.ajpendo.org
52. Tam CS, Heilbronn LK, Henegar C, Wong M, Cowell CT, Cowley
MJ, Kaplan W, Clément K, Baur LA. An early inflammatory gene
profile in visceral adipose tissue in children. Int J Pediatr Obes 6:
e360–e363, 2011.
53. Tominaga K, Kagata T, Johmura Y, Hishida T, Nishizuka M,
Imagawa M. SLC39A14, a LZT protein, is induced in adipogenesis and
transports zinc. FEBS J 272: 1590–1599, 2005.
54. Trayhurn P, Duncan JS, Wood AM, Beattie JH. Metallothionein gene
expression and secretion in white adipose tissue. Am J Physiol Regul
Integr Comp Physiol 279: R2329–R2335, 2000.
55. Trujello ME, Sullivan S, Harten I, Schneider SH, Greenberg AS, Fried SK.
Interleukin-6 regulates human adipose tissue lipid metabolism and leptin produc-
tion in vitro. J Clin Endocrinol Metab 89: 5577–5582, 2004.
56. Tornatore L, Thotakura AK, Bennett J, Moretti M, Franzoso G. The
nuclear factor kappa B signaling pathway: integrating metabolism with
inflammation. Trends Cell Biol 22: 557–566, 2012.
57. Wang LH, Yang XY, Zhang X, Huang J, Hou J, Li J, Xiong H,
Mihalic K, Zhu H, Xiao W, Farrar WL. Transcriptional inactivation of
STAT3 by PPARgamma suppresses IL6-responsive multiple myeloma
cells. Immunity 20: 205–218, 2004.
58. Wang Y, Kim KA, Kim JH, Sul HS. Pref-1, a preadipocyte secreted
factor that inhibits adipogenesis. J Nutr 136: 2953–2956, 2006.
59. Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Fer-
rante AW Jr. Obesity is associated with macrophage accumulation in
adipose tissue. J Clin Invest 112: 1796–1808, 2003.
60. Wernstedt Asterholm I, Tao C, Morley TS, Wang QA, Delgado-Lopez F,
Wang ZV, Scherer PE. Adipocyte inflammation is essential for healthy adipose
tissue expansion and remodeling. Cell Metab 20: 103–118, 2014.
61. Xu H, Barnes GT, Yang Q, Tan G, Yang D, Chou CJ, Sole J, Nichols
A, Ross JS, Tartaglia LA, Chen H. Chronic inflammation in fat plays a
crucial role in the development of obesity-related insulin resistance. J Clin
Invest 112: 1821–1830, 2003.
62. Zhanga Y, Goldmana S, Baergaa R, Zhaoa Y, Komatsub M, Jin S.
Adipose-specific deletion of autophagy-related gene 7 (atg7) in mice
reveals a role in adipogenesis. Proc Natl Acad Sci USA 106: 19860–
19865, 2009.
63. Zhou Z, Wang L, Song Z, Saari JT, McClain CJ, Kang J. Abrogation
of nuclear factor-␬B activation is involved in zinc inhibition of lipopoly-
saccharide-induced tumor necrosis factor-␣ production and liver injury.
Am J Pathol 164: 1547–1556, 2004.
64. Zu L, He J, Jiang H, Xu C, Pu S, Xu G. Bacterial endotoxin stimulates
adipose lipolysis via toll-like receptor 4 and extracellular signal-regulated
kinase pathway. J Biol Chem 284: 5915–5926, 2009.
E268 ZINC TRANSPORTER Zip14 AND ADIPOSE TISSUE REGULATION
AJP-Endocrinol Metab • doi:10.1152/ajpendo.00421.2015 • www.ajpendo.org

More Related Content

What's hot

Autophagy the housekeeper in every cell
Autophagy the housekeeper in every cellAutophagy the housekeeper in every cell
Autophagy the housekeeper in every cellfathi neana
 
‘From Molecular to Systems Nutrition. Lessons from mouse to man’ NUGO Dublin...
‘From Molecular to Systems Nutrition. Lessons from mouse to man’ NUGO Dublin...‘From Molecular to Systems Nutrition. Lessons from mouse to man’ NUGO Dublin...
‘From Molecular to Systems Nutrition. Lessons from mouse to man’ NUGO Dublin...Norwich Research Park
 
Loss of the RNA polymerase III repressor Maf1 confers obesity resistance
Loss of the RNA polymerase III repressor Maf1 confers obesity resistanceLoss of the RNA polymerase III repressor Maf1 confers obesity resistance
Loss of the RNA polymerase III repressor Maf1 confers obesity resistanceAsh Byrnes
 
Moving into the Post-MetagenomicEra of Gut Microbiome Research
Moving into the Post-MetagenomicEra of Gut Microbiome ResearchMoving into the Post-MetagenomicEra of Gut Microbiome Research
Moving into the Post-MetagenomicEra of Gut Microbiome ResearchJonathan Clarke
 
Serum lipid profile and liver enzymes of Rats fed Lageneria sphaerica (Wild b...
Serum lipid profile and liver enzymes of Rats fed Lageneria sphaerica (Wild b...Serum lipid profile and liver enzymes of Rats fed Lageneria sphaerica (Wild b...
Serum lipid profile and liver enzymes of Rats fed Lageneria sphaerica (Wild b...Premier Publishers
 
Systems Nutrition of the Gut-Liver Axis and the Role of the Microbiome
Systems Nutrition of the Gut-Liver Axis and the Role of the MicrobiomeSystems Nutrition of the Gut-Liver Axis and the Role of the Microbiome
Systems Nutrition of the Gut-Liver Axis and the Role of the MicrobiomeNorwich Research Park
 
Nanjing 2 2013 Lecture "Nutrigenomics part 2" From healthy to too much: The r...
Nanjing 2 2013 Lecture "Nutrigenomics part 2" From healthy to too much: The r...Nanjing 2 2013 Lecture "Nutrigenomics part 2" From healthy to too much: The r...
Nanjing 2 2013 Lecture "Nutrigenomics part 2" From healthy to too much: The r...Norwich Research Park
 
Effects of pre transport fasting on the
Effects of pre transport fasting on theEffects of pre transport fasting on the
Effects of pre transport fasting on theambiusanton
 
Appetence maturation and control in Anopheles gambiae
Appetence maturation and control in Anopheles gambiaeAppetence maturation and control in Anopheles gambiae
Appetence maturation and control in Anopheles gambiaedarsic
 
art%3A10.1007%2Fs00125-013-3030-x
art%3A10.1007%2Fs00125-013-3030-xart%3A10.1007%2Fs00125-013-3030-x
art%3A10.1007%2Fs00125-013-3030-xJoana Oliveira
 
KUMC Presentation
KUMC PresentationKUMC Presentation
KUMC PresentationMichael Epp
 
Ermak styela clava kinetics stomach j exp zool 1976
Ermak styela clava kinetics stomach j exp zool 1976Ermak styela clava kinetics stomach j exp zool 1976
Ermak styela clava kinetics stomach j exp zool 1976Thomas Ermak
 
Inaugural lecture Norwich UEA 14 oct 2014
Inaugural lecture Norwich UEA 14 oct 2014Inaugural lecture Norwich UEA 14 oct 2014
Inaugural lecture Norwich UEA 14 oct 2014Norwich Research Park
 
We are what we eat - The role of diets in the gut-microbiota-health interaction
We are what we eat - The role of diets in the gut-microbiota-health interactionWe are what we eat - The role of diets in the gut-microbiota-health interaction
We are what we eat - The role of diets in the gut-microbiota-health interactionNorwich Research Park
 
2013 List_The role of GH in adipose tissue-LiGHRKO mice
2013 List_The role of GH in adipose tissue-LiGHRKO mice2013 List_The role of GH in adipose tissue-LiGHRKO mice
2013 List_The role of GH in adipose tissue-LiGHRKO miceEdward List
 

What's hot (20)

Autophagy the housekeeper in every cell
Autophagy the housekeeper in every cellAutophagy the housekeeper in every cell
Autophagy the housekeeper in every cell
 
‘From Molecular to Systems Nutrition. Lessons from mouse to man’ NUGO Dublin...
‘From Molecular to Systems Nutrition. Lessons from mouse to man’ NUGO Dublin...‘From Molecular to Systems Nutrition. Lessons from mouse to man’ NUGO Dublin...
‘From Molecular to Systems Nutrition. Lessons from mouse to man’ NUGO Dublin...
 
Loss of the RNA polymerase III repressor Maf1 confers obesity resistance
Loss of the RNA polymerase III repressor Maf1 confers obesity resistanceLoss of the RNA polymerase III repressor Maf1 confers obesity resistance
Loss of the RNA polymerase III repressor Maf1 confers obesity resistance
 
Moving into the Post-MetagenomicEra of Gut Microbiome Research
Moving into the Post-MetagenomicEra of Gut Microbiome ResearchMoving into the Post-MetagenomicEra of Gut Microbiome Research
Moving into the Post-MetagenomicEra of Gut Microbiome Research
 
Serum lipid profile and liver enzymes of Rats fed Lageneria sphaerica (Wild b...
Serum lipid profile and liver enzymes of Rats fed Lageneria sphaerica (Wild b...Serum lipid profile and liver enzymes of Rats fed Lageneria sphaerica (Wild b...
Serum lipid profile and liver enzymes of Rats fed Lageneria sphaerica (Wild b...
 
Systems Nutrition of the Gut-Liver Axis and the Role of the Microbiome
Systems Nutrition of the Gut-Liver Axis and the Role of the MicrobiomeSystems Nutrition of the Gut-Liver Axis and the Role of the Microbiome
Systems Nutrition of the Gut-Liver Axis and the Role of the Microbiome
 
Nanjing 2 2013 Lecture "Nutrigenomics part 2" From healthy to too much: The r...
Nanjing 2 2013 Lecture "Nutrigenomics part 2" From healthy to too much: The r...Nanjing 2 2013 Lecture "Nutrigenomics part 2" From healthy to too much: The r...
Nanjing 2 2013 Lecture "Nutrigenomics part 2" From healthy to too much: The r...
 
Matsunami et al., 2010 4th
Matsunami et al., 2010 4thMatsunami et al., 2010 4th
Matsunami et al., 2010 4th
 
EB 2016 - 4th Yogurt Summit 2016 - Li Wen
EB 2016 - 4th Yogurt Summit 2016 - Li Wen EB 2016 - 4th Yogurt Summit 2016 - Li Wen
EB 2016 - 4th Yogurt Summit 2016 - Li Wen
 
Effects of pre transport fasting on the
Effects of pre transport fasting on theEffects of pre transport fasting on the
Effects of pre transport fasting on the
 
Nutrigenomics of FAT
Nutrigenomics of FAT Nutrigenomics of FAT
Nutrigenomics of FAT
 
Appetence maturation and control in Anopheles gambiae
Appetence maturation and control in Anopheles gambiaeAppetence maturation and control in Anopheles gambiae
Appetence maturation and control in Anopheles gambiae
 
art%3A10.1007%2Fs00125-013-3030-x
art%3A10.1007%2Fs00125-013-3030-xart%3A10.1007%2Fs00125-013-3030-x
art%3A10.1007%2Fs00125-013-3030-x
 
KUMC Presentation
KUMC PresentationKUMC Presentation
KUMC Presentation
 
Ermak styela clava kinetics stomach j exp zool 1976
Ermak styela clava kinetics stomach j exp zool 1976Ermak styela clava kinetics stomach j exp zool 1976
Ermak styela clava kinetics stomach j exp zool 1976
 
Inaugural lecture Norwich UEA 14 oct 2014
Inaugural lecture Norwich UEA 14 oct 2014Inaugural lecture Norwich UEA 14 oct 2014
Inaugural lecture Norwich UEA 14 oct 2014
 
191 195
191 195191 195
191 195
 
We are what we eat - The role of diets in the gut-microbiota-health interaction
We are what we eat - The role of diets in the gut-microbiota-health interactionWe are what we eat - The role of diets in the gut-microbiota-health interaction
We are what we eat - The role of diets in the gut-microbiota-health interaction
 
Zz c0610-1003 diabetes-np_final
Zz c0610-1003 diabetes-np_finalZz c0610-1003 diabetes-np_final
Zz c0610-1003 diabetes-np_final
 
2013 List_The role of GH in adipose tissue-LiGHRKO mice
2013 List_The role of GH in adipose tissue-LiGHRKO mice2013 List_The role of GH in adipose tissue-LiGHRKO mice
2013 List_The role of GH in adipose tissue-LiGHRKO mice
 

Viewers also liked

Viewers also liked (8)

LEÇON 275 – La Voix guérissante de Dieu protège toutes choses aujourd’hui.
LEÇON 275 – La Voix guérissante de Dieu protège toutes choses aujourd’hui.LEÇON 275 – La Voix guérissante de Dieu protège toutes choses aujourd’hui.
LEÇON 275 – La Voix guérissante de Dieu protège toutes choses aujourd’hui.
 
Testimonial-Sony
Testimonial-SonyTestimonial-Sony
Testimonial-Sony
 
3ero j
3ero j3ero j
3ero j
 
Ttlv nguyen tuan khanh
Ttlv nguyen tuan khanhTtlv nguyen tuan khanh
Ttlv nguyen tuan khanh
 
On set, 2016
On set, 2016 On set, 2016
On set, 2016
 
mm
mmmm
mm
 
Rathi_Mrinalini
Rathi_MrinaliniRathi_Mrinalini
Rathi_Mrinalini
 
A biblia em esb0ços
A biblia em esb0çosA biblia em esb0ços
A biblia em esb0ços
 

Similar to E258.full

Hyperlipidemia & Diabetes & Gut Microbiota
Hyperlipidemia & Diabetes & Gut Microbiota Hyperlipidemia & Diabetes & Gut Microbiota
Hyperlipidemia & Diabetes & Gut Microbiota Seyed Davar Siadat
 
Dysbiosis and NAFLD.pptx
Dysbiosis and NAFLD.pptxDysbiosis and NAFLD.pptx
Dysbiosis and NAFLD.pptxMohamed Wifi
 
Austin publishing group - Oral kefir grains supplementation improves metaboli...
Austin publishing group - Oral kefir grains supplementation improves metaboli...Austin publishing group - Oral kefir grains supplementation improves metaboli...
Austin publishing group - Oral kefir grains supplementation improves metaboli...Austin Publishing Group
 
Adipose tissue innate immunity & inflammation - a nutrigenomics perspective o...
Adipose tissue innate immunity & inflammation - a nutrigenomics perspective o...Adipose tissue innate immunity & inflammation - a nutrigenomics perspective o...
Adipose tissue innate immunity & inflammation - a nutrigenomics perspective o...Norwich Research Park
 
2007 List SKIN PROTEOMICS
2007 List SKIN PROTEOMICS2007 List SKIN PROTEOMICS
2007 List SKIN PROTEOMICSEdward List
 
Alterations of Mitochondrial Functions and DNA in Diabetic Cardiomyopathy of ...
Alterations of Mitochondrial Functions and DNA in Diabetic Cardiomyopathy of ...Alterations of Mitochondrial Functions and DNA in Diabetic Cardiomyopathy of ...
Alterations of Mitochondrial Functions and DNA in Diabetic Cardiomyopathy of ...CrimsonPublishersIOD
 
Low beneficial effects of short term antidiabetic diet treatment in streptozo...
Low beneficial effects of short term antidiabetic diet treatment in streptozo...Low beneficial effects of short term antidiabetic diet treatment in streptozo...
Low beneficial effects of short term antidiabetic diet treatment in streptozo...iosrphr_editor
 
FinalPrattPresentationPoster-1
FinalPrattPresentationPoster-1FinalPrattPresentationPoster-1
FinalPrattPresentationPoster-1Alexa Turner
 
Effect of Nutritional Factors on Regulation of Gene Expression
 Effect of Nutritional Factors on Regulation of Gene Expression Effect of Nutritional Factors on Regulation of Gene Expression
Effect of Nutritional Factors on Regulation of Gene Expressionsohinisc14
 
The role of curcumin in streptozotocin induced hepatic damage and the trans-d...
The role of curcumin in streptozotocin induced hepatic damage and the trans-d...The role of curcumin in streptozotocin induced hepatic damage and the trans-d...
The role of curcumin in streptozotocin induced hepatic damage and the trans-d...Prof. Hesham N. Mustafa
 
Infect. Immun.-2011-Fabich-2430-9
Infect. Immun.-2011-Fabich-2430-9Infect. Immun.-2011-Fabich-2430-9
Infect. Immun.-2011-Fabich-2430-9Andrew Fabich
 
Gut flora in diabetes
Gut flora in diabetesGut flora in diabetes
Gut flora in diabetesNadia Shams
 
Hepatoprotective screening models
Hepatoprotective screening modelsHepatoprotective screening models
Hepatoprotective screening modelsPavan Shukla
 
Screening of hepatoprotective drugs
Screening of hepatoprotective drugsScreening of hepatoprotective drugs
Screening of hepatoprotective drugsDipanjaliKamthe
 
Chan & Roth 2008
Chan & Roth 2008Chan & Roth 2008
Chan & Roth 2008Kin Chan
 
Personalised nutrition for the gut microbiome
Personalised nutrition for the gut microbiomePersonalised nutrition for the gut microbiome
Personalised nutrition for the gut microbiomeEFSA EU
 
The association between gut microbiota and body weight
The association between gut microbiota and body weightThe association between gut microbiota and body weight
The association between gut microbiota and body weightAshwath Venkatasubramanian
 

Similar to E258.full (20)

Hyperlipidemia & Diabetes & Gut Microbiota
Hyperlipidemia & Diabetes & Gut Microbiota Hyperlipidemia & Diabetes & Gut Microbiota
Hyperlipidemia & Diabetes & Gut Microbiota
 
Dysbiosis and NAFLD.pptx
Dysbiosis and NAFLD.pptxDysbiosis and NAFLD.pptx
Dysbiosis and NAFLD.pptx
 
Austin publishing group - Oral kefir grains supplementation improves metaboli...
Austin publishing group - Oral kefir grains supplementation improves metaboli...Austin publishing group - Oral kefir grains supplementation improves metaboli...
Austin publishing group - Oral kefir grains supplementation improves metaboli...
 
Adipose tissue innate immunity & inflammation - a nutrigenomics perspective o...
Adipose tissue innate immunity & inflammation - a nutrigenomics perspective o...Adipose tissue innate immunity & inflammation - a nutrigenomics perspective o...
Adipose tissue innate immunity & inflammation - a nutrigenomics perspective o...
 
2007 List SKIN PROTEOMICS
2007 List SKIN PROTEOMICS2007 List SKIN PROTEOMICS
2007 List SKIN PROTEOMICS
 
Alterations of Mitochondrial Functions and DNA in Diabetic Cardiomyopathy of ...
Alterations of Mitochondrial Functions and DNA in Diabetic Cardiomyopathy of ...Alterations of Mitochondrial Functions and DNA in Diabetic Cardiomyopathy of ...
Alterations of Mitochondrial Functions and DNA in Diabetic Cardiomyopathy of ...
 
Low beneficial effects of short term antidiabetic diet treatment in streptozo...
Low beneficial effects of short term antidiabetic diet treatment in streptozo...Low beneficial effects of short term antidiabetic diet treatment in streptozo...
Low beneficial effects of short term antidiabetic diet treatment in streptozo...
 
FinalPrattPresentationPoster-1
FinalPrattPresentationPoster-1FinalPrattPresentationPoster-1
FinalPrattPresentationPoster-1
 
Effect of Nutritional Factors on Regulation of Gene Expression
 Effect of Nutritional Factors on Regulation of Gene Expression Effect of Nutritional Factors on Regulation of Gene Expression
Effect of Nutritional Factors on Regulation of Gene Expression
 
The role of curcumin in streptozotocin induced hepatic damage and the trans-d...
The role of curcumin in streptozotocin induced hepatic damage and the trans-d...The role of curcumin in streptozotocin induced hepatic damage and the trans-d...
The role of curcumin in streptozotocin induced hepatic damage and the trans-d...
 
Infect. Immun.-2011-Fabich-2430-9
Infect. Immun.-2011-Fabich-2430-9Infect. Immun.-2011-Fabich-2430-9
Infect. Immun.-2011-Fabich-2430-9
 
Gut flora in diabetes
Gut flora in diabetesGut flora in diabetes
Gut flora in diabetes
 
Hepatoprotective screening models
Hepatoprotective screening modelsHepatoprotective screening models
Hepatoprotective screening models
 
Food Chem-2008
Food Chem-2008Food Chem-2008
Food Chem-2008
 
Screening of hepatoprotective drugs
Screening of hepatoprotective drugsScreening of hepatoprotective drugs
Screening of hepatoprotective drugs
 
Chan & Roth 2008
Chan & Roth 2008Chan & Roth 2008
Chan & Roth 2008
 
Personalised nutrition for the gut microbiome
Personalised nutrition for the gut microbiomePersonalised nutrition for the gut microbiome
Personalised nutrition for the gut microbiome
 
published journals
published journalspublished journals
published journals
 
Obesity ppt 2
Obesity ppt 2Obesity ppt 2
Obesity ppt 2
 
The association between gut microbiota and body weight
The association between gut microbiota and body weightThe association between gut microbiota and body weight
The association between gut microbiota and body weight
 

E258.full

  • 1. CALL FOR PAPERS Novel Aspects of Adipocyte Biology Zinc transporter Slc39a14 regulates inflammatory signaling associated with hypertrophic adiposity Catalina Troche, Tolunay Beker Aydemir, and Robert J. Cousins Food Science and Human Nutrition Department and Center for Nutritional Sciences, College of Agricultural and Life Sciences, University of Florida, Gainesville, Florida Submitted 23 September 2015; accepted in final form 2 December 2015 Troche C, Beker Aydemir T, Cousins RJ. Zinc transporter Slc39a14 regulates inflammatory signaling associated with hypertrophic adiposity. Am J Physiol Endocrinol Metab 310: E258–E268, 2016. First published December 8, 2015; doi:10.1152/ajpendo.00421.2015.—Zinc is a signal- ing molecule in numerous metabolic pathways, the coordination of which occurs through activity of zinc transporters. The expression of zinc transporter Zip14 (Slc39a14), a zinc importer of the solute carrier 39 family, is stimulated under proinflammatory conditions. Adipose tissue upregulates Zip14 during lipopolysaccharide-induced endotoxemia. A null mutation of Zip14 (KO) revealed that phenotypic changes in adipose include increased cytokine production, increased plasma lep- tin, hypertrophied adipocytes, and dampened insulin signaling. Adi- pose tissue from KO mice had increased levels of preadipocyte markers, lower expression of the differentiation marker (PPAR␥), and activation of NF-␬B and STAT3 pathways. Our overall hypothesis was that ZIP14 would play a role in adipocyte differentiation and inflammatory obesity. Global Zip14 KO causes systemic endotox- emia. The observed metabolic changes in adipose metabolism were reversed when oral antibiotics were administrated, indicating that circulating levels of endotoxin were in part responsible for the adipose phenotype. To evaluate a mechanism, 3T3-L1 cells were differenti- ated into adipocytes and treated with siRNA to knock down Zip14. These cells had an impaired ability to mobilize zinc, which caused dysregulation of inflammatory pathways (JAK2/STAT3 and NF-␬B). The Zip14 deletion may limit the availability of intracellular zinc, yielding the unique phenotype of inflammation coupled with hyper- trophy. Taken together, these results suggest that aberrant zinc distri- bution observed with Zip14 ablation impacts adipose cytokine pro- duction and metabolism, ultimately increasing fat deposition when exposed to endotoxin. To our knowledge, this is the first investigation into the mechanistic role of ZIP14 in adipose tissue regulation and metabolism. adipose; endotoxemia; zinc signaling; hypertrophy; inflammation WHITE ADIPOSE TISSUE (WAT) contains a wide array of cell types that are characterized as connective tissue, nervous tissue, stromovascular cells, and immune cells. Together, these cells produce unique substances, i.e., leptin, adiponectin, and resis- tin, which contribute to the para- and autoendocrine regulation of lipid metabolism (21, 28). Limited epidemiological studies have linked aberrant trace mineral metabolism with adipose pathology; e.g., obesity is clinically correlated to deficiencies in iron, calcium, and zinc (18, 19, 49, 50). Zinc was first thought to be involved in metabolic activity of adipocytes through insulin-like effects (36) and through its inherent anti- oxidant properties (16). Human adipose tissue expresses many zinc transporters that may respond to the metabolic status of the patients, e.g., lean vs. obese (47). Despite the apparent correlation between zinc and adipose metabolism, a mecha- nism of action has yet to be defined. Zinc partitioning within mammalian cells is due to ZIP and ZnT transporter activity, which control influx and efflux, re- spectively, of cytosolic zinc (27, 31). ZIP14 is a zinc influx transporter that is known to be upregulated during inflamma- tion (3–5, 34, 40). We have focused on ZIP14 since it was found in our initial experiments to be the most responsive zinc transporter in mouse liver post-lipopolysaccharide administra- tion (34). Knockout (KO) of ZIP14 results in a variety of unique phenotypes. We have reported previously that mice lacking ZIP14 have impaired liver zinc uptake (3) and elevated levels of serum endotoxin (25). Previously, Aydemir et al. (3) reported an increase in fat/lean ratios in Zip14-KO mice, a finding that led to our further investigation of ZIP14 in adipose function. The elevated level of serum endotoxin is particularly relevant to adipose in that chronic exposure to endotoxin can initiate obesity and insulin resistance (10). Inflammatory cyto- kines have a proliferative effect on adipocytes, leading to expansion of cell mass through both hypertrophy and hyper- plasia (14, 20). Collectively, the marked induction of ZIP14 in WAT during inflammation along with the KO phenotype of increased adiposity, and metabolic endotoxemia suggests that this transporter alters zinc trafficking in adipocytes with func- tional outcomes. Based on these previous findings, we hypothesized that ZIP14 would be critical to the inflammatory response and ultimately metabolic activity of WAT. Here, we report that WAT from KO mice appears to be insulin insensitive with hypertrophied adipocytes and dampened insulin signaling. Insulin resistance was linked to chronic inflammation within KO adipose tissue through upregulated cytokine expression and the Toll-like receptor 4 (TLR4) accessory protein my- eloid differentiation primary response gene 88 (MyD88). Finally, we show that aberrant zinc signaling within the KO adipocyte is linked to enhanced JAK/STAT and NF-␬B signaling, leading to impaired differentiation. Both findings tie directly to dyslipidemia and hypertrophy. The involve- ment of ZIP14 in major inflammatory pathways impacts adipocyte development and makes it a potential therapeutic target for inflammatory disorders in adipose, e.g., obesity and insulin resistance. Address for reprint requests and other correspondence: R. J. Cousins, Univ. of Florida, 572 Newell Dr., P. O. Box 110370, Gainesville, FL 32611-0370 (e-mail: cousins@ufl.edu). Am J Physiol Endocrinol Metab 310: E258–E268, 2016. First published December 8, 2015; doi:10.1152/ajpendo.00421.2015. 0193-1849/16 Copyright © 2016 the American Physiological Society http://www.ajpendo.orgE258
  • 2. MATERIALS AND METHODS Animals and diets. Design and genomic validation of the Zip14-KO (Zip14Ϫ/Ϫ ) mice have been described previously (3, 5, 33). Briefly, Zip14 heterozygous mice were used to establish a breeding colony to generate wild-type (WT; Zip14ϩ/ϩ ) and KO mice. For the experi- ments described herein, female KO and WT mice were used between 8 and 16 wk of age. Mice were provided with ad libitum access to a commercial chow rodent diet (7912, with 60 mg zinc/kg provided by ZnO; Harlan-Teklad, Indianapolis, IN) and tap water. Animal treatments. In experiments modeling acute inflammation, lipopolysaccharide (LPS; E. coli serotype 055:B5; Sigma, St. Louis, MO) in phosphate-buffered saline (PBS) was administered at 2.0 mg/kg via intraperitoneal (ip) injection. Mice received LPS injections up to 18 h prior to euthanasia. Control animals received ip injections of PBS. In other experiments, fasted mice were gavaged with 65 Zn (2 ␮Ci/mouse in 250 ␮l of saline) and euthanized 3 h postgavage to determine the tissue distribution. Specific activity of the 65 Zn (Perkin- Elmer, Waltham, MA) when used was 4.4 mCi/mg. Tissue accumu- lation of 65 Zn was measured via ␥-scintillation spectrometry. For the antibiotic experiment, ultrapure (MiliQ, Billerica, MA) drinking water was supplemented with neomycin (0.5 mg/ml) and ampicillin (1 mg/ml) for 4 wk prior to tissue collection. Mice were anesthetized using isoflurane (Baxter, Deerfield, IL) prior to injection or gavage. Euthanasia was conducted via cardiac puncture. Blood from cardiac puncture was collected into a clot activator microgel barrier collection tube (Capiject; Terumo Medical, Somerset, NJ). Serum was separated via centrifugation and stored at Ϫ80°C prior to further analysis. All harvested tissues (intestine, muscle, liver, and adipose) were snap- frozen in liquid nitrogen and stored at Ϫ80°C prior to further pro- cessing. All studies described herein used intra-abdominal white fat pads (parameterial fat pads) that lay along the uterine horn. Unless otherwise specified, tissues were homogenized in assay-specific buf- fers using a Bullet blender with zirconium oxide beads (Next Ad- vance, Averill Park, NY). All animal protocols were approved by the University of Florida Institutional Animal Care and Use Committee. Cell culture. A well-established embryonic mouse fibroblast cell line (3T3-L1; ATCC, Manassas, VA) was used to model adipocyte growth and differentiation. Cells were cultured in growth medium (DMEM;4.5 g/l glucose, 15% FBS, and 1% pencillin-streptomycin; all from Corning, Manassas, VA). After two passages, subconfluent primary cultures were trypsinized and plated into 12-well plates for experimentation. Transient knockdown of ZIP14 was carried out with HiPerfect (Qiagen, Valencia, CA). 3T3-L1 cells were grown to 90% confluence, and cells were treated with 25 nM siRNA (Darmacon, Pittsburgh, PA) for 48 h prior to incubation with differentiation medium with or without LPS (10 ng/ml). Thereafter, cells were exposed to differentiation medium (DMEM; 4.5 g/l glucose, 10% FBS, and 1% pencillin-streptomycin supplemented with 1.7 ␮M insulin, 1 ␮M dexamethasone, and 0.5 mM isobutylmethylxanthine). Cells were exposed to differentiation medium for three days prior to postdifferentiation medium (DMEM; 4.5 g/l glucose, 10% FBS, and 1% pencillin-streptomycin). The entire experimental period from differentiation to collection was 8 days. For experiments which involved LPS, cells were plated into their experimental dishes (pas- sage 3), and 10 ng/ml LPS was supplemented to the medium the next day. Thereafter, cells were exposed to LPS throughout the entire experimental course through differentiation; medium was changed every 2 days. To confirm our findings in 3T3L1 cells, ear mesenchymal stem cells (EMSC) were collected from WT and KO mice, as described previously by Rim et al. (42). Briefly, ears were minced and digested in medium containing collagenase type I (Worthington, Lakewood, NJ). Prior to the cell suspension being filtered (100 ␮m; Fisher Scientific, Suwanee, GA) minced tissue and collagenase medium were placed in a 37°C shaking water bath for 1 h. Filtered cells were pelleted through centrifugation (360 g, 5 min, room temperature), and red blood cells were lysed. Isolated cells were plated in 100-mm petri dishes in the previously described growth medium. Cells were al- lowed to expand for 3–5 days, and then these subconfluent primary cultures were trypsinized and plated into 24-well plates for experi- mentation. At visually confirmed confluence the EMSC were stimu- lated into adipocytes with previously described differentiation me- dium. After 3 days in differentiation medium, cells were maintained in postdifferentiation media. The entire experimental period from differ- entiation to collection was 10 days. Analytical procedures. Serum was diluted 1:3 using ultrapure water, and zinc levels were determined using flame atomic absorption spectrophotometry (AAS). Adipose tissue was weighed prior to HNO3 digestion (90°C for 3 h) and diluted 1:1 with ultrapure water prior to AAS analysis. In experiments where total zinc was determined in cells, 3T3-L1 cells were first cultured in 150-mm plates (Corning). Cells were collected in ice-cold PBS, and an aliquot was obtained for total protein determination (Pierce BCA assay; Thermo Fisher Scien- tific, Waltham, MA). The remaining cell pellet was digested in HNO3 (80°C for 3 h) and diluted 1:1 with ultrapure water. Total zinc was measured by AAS, and values were normalized to total protein, as described above. Serum endotoxin was measured as reported previ- ously (25) using the LAL Chromogenic Endotoxin Quantitation Kit (Thermo Fisher Scientific), with absorbance read at 407 nm. Serum and tissue leptin were measured using a mouse/rat-specific ELISA kit (Alpco, Salem, NH). Tissue was first homogenized in cell extraction buffer (20 mM Tris·HCl, 1 mM EDTA, and 254 mM sucrose, pH 7.4), and absorbance was measured at 450 nm (1). Leptin values were normalized to total protein, as described above. Adipose glucose content was determined enzymatically with a total glucose assay kit (Sigma-Aldridge, St. Louis, MO). Prior to analysis, tissues were homogenized in ultrapure water and diluted 1:2. Absorbance was measured at 540 nm. RNA quantification. For RNA isolation, a portion of frozen fat pad was placed into TRIzol reagent (Life Technologies, Thermo Fisher Scientific). Early experiments (Fig. 1, A–C) utilized a Polytron blender to lyse tissue. Subsequent tissues were homogenized as described previously using the Bullet Blender. cDNA was generated using the iScript reagents (Bio-Rad, Hercules, CA). Quantitative real-time PCR was performed using the Real-time PCR Fast SYBR Green Master Mix and a StepOnePlus Fast Thermocycler (Applied Biosystems, Thermo Fisher Scientific). Primers to detect specific mRNAs were designed to span intron/exon boundaries: peroxisome proliferator-activated receptor-␥ (Ppar␥), 5=-GGAAGACCACTC- GCATTCCTT-3= and 5=-GTAATCAGCAACCATTGGGTCA-3=; plasminogen activator inhibitor-1 (Pai-1), 5=-AGGGCTTCATGC- CCCACTTCTTCA-3= and 5=-GTAGAGGGCATTCACCAGCA- CCA-3=. All other primer sequences were selected using the qPrim- erDepot database, http://mouseprimerdepot.nci.nih.gov/, as described in Cui et al. (17). TBP mRNA was the normalizer for relative expression, as described previously (3, 5). Western blotting. Western analysis was performed using polyclonal rabbit antibodies against ZIP14 developed and affinity purified (Life Technologies, Thermo Fisher Scientific) as described previously (3, 5). Akt, phosphorylated Akt (p-Ser473 ), hormone-sensitive lipase (HSL), phosphorylated HSL (Ser660 ), I␬B, phosphorylated I␬B (p- Ser32/36 ), IR1␤, phosphorylated IR1␤ (p-Y1150/1151 ), mammalian tar- get of rapamycin (mTOR), phosphorylated mTOR (Ser2448 ), MyD88, NF-␬B, phosphorylated NF-␬B (p65), PPAR␥, preadipocyte factor-1 (PREF-1/DLK-1), SOCS3, STAT3, and phosphorylated STAT3 (p- Y705 ) antibodies were purchased from Cell Signaling Technology (Boston, MA). F4/80 and zinc ␣2-glycoprotein (ZAG) antibody were purchased from Santa Cruz Biotechnology (Dallas, TX). Frozen tissues were processed over liquid nitrogen to prevent freeze thaw and protein degradation. Tissues were homogenized in RIPA lysis buffer (Santa Cruz Biotechnology) with 100ϫ protease and phosphatase inhibitors (Thermo Fisher Scientific), along with phenylmethanesul- fonyl fluoride (Sigma-Aldrich). Proteins were separated using a 10% E259ZINC TRANSPORTER Zip14 AND ADIPOSE TISSUE REGULATION AJP-Endocrinol Metab • doi:10.1152/ajpendo.00421.2015 • www.ajpendo.org
  • 3. acrylamide gel for SDS-PAGE and transferred to nitrocellulose mem- branes. Transfer was verified through Ponceau Red staining, and proteins were visualized through chemiluminescence (SuperSignal West Pico, Thermo Fisher Scientific) and digital imaging (Protein Simple, San Jose, CA). Tubulin (Abcam, Cambridge, MA) abundance was used as the loading control. Adipose histology. Collected fat pads (parametrial fat pads) were fixed in 10% neutral buffered formalin for 24 h prior to paraffin embedding and sectioning. Slides were hematoxylin and eosin stained prior to microscopy using a Zeiss Axiovert 100 microscope at ϫ10 magnification. Adipocyte area was measured using ImageJ software (National Institute of Mental Health, Bethesda, MD). Cell areas were determined on cells with contiguous borders using the Adiposoft open-source ImageJ plugin http://fiji.sc/Adiposoft (22). Between 600 and 1,500 cells/image were used. Ten full images per genotype (5 mice, 2 images/mouse) were used to generate cell area images. In cell experiments where Oil Red O was used, cells were first fixed in 3.3% formaldehyde and stained in 3 ␮g/ml Oil Red O. Images were collected using the previously described Zeiss Axiovert 100 at ϫ10 magnification. To quantify staining, Oil Red O was extracted with isopropanol containing 4% NP-40, and absorbance was measured at 520 nm (62). Confocal laser-scanning microscopy. Imaging was done with a Leica TCS SP5 laser-scanning confocal microscope with LAS-AF imaging software, using a ϫ40 oil objective. For detection of labile zinc, cells were incubated with FluoZin-3 AM (Invitrogen, Waltham, MA), as described previously (4). Briefly, cells were incubated in 5 ␮M FluoZin-3 for 30 min, followed by a 30-min incubation in Locke’s buffer (154 mM NaCl, 5.6 mM KCl, 3.6 mM NaHCO3, 1.2 mM MgSO4, 5.6 mM glucose, 2.5 mM CaCl2, and 10 mM HEPES). Cells were then stimulated with 40 ␮M ZnCl2, and fluorescence was measured at 516 nm with excitation at 494 nm. A similar method was used to quantify FluoZin-3 AM in 12-well plates, with fluorescence being determined as above. Immediately after fluorescence was read, the cells were trypsanized and diluted 1:1 with trypan blue to deter- mine cell number. FluoZin-3 AM relative fluorescent units were Fig. 2. LPS-induced Zip14 expression is correlated with cytokine expression. A: mice received LPS (2 mg/kg ip) or the same volume (250 ␮l) of saline [control (CTRL)] 1–18 h before being euthanized. Relative transcript levels of Zip14 and specific cytokines; n ϭ 3 mice/treatment Ϯ SE. B: serum hypozincemia confirms the acute-phase response. Zinc concentrations in serum and WAT were measured by atomic absorption spectrophotometry (AAS). Adipose zinc values were normalized to wet tissue weight; n ϭ 3 mice/treatment Ϯ SE. C: Western analysis of ZIP14, lipolytic marker hormone-sensitive lipase (HSL), and differentiation marker PPAR␥ (peroxisome proliferator-activated receptor-␥) during the acute-phase response in WAT. Each lane is a pooled lysate of 3 mice/treatment. *P Ͻ 0.05; **P Ͻ 0.01; ***P Ͻ 0.0001. P-IR, phosphorylated insulin receptor. Fig. 1. Expression of Zip14 in white adipose tissue (WAT) is induced by lipopolysaccharide (LPS). A: tissue expression profile of Zip14 transcripts. Transcript abundance was determined by quantitative PCR, starting with an equal amount of total RNA from each tissue. B: fold change in Zip14 tissue expression 18 h post-intraperitoneal (ip) injection of LPS (2 mg/kg). C: of the WAT zinc transporters significantly (P Յ 0.05) impacted by LPS, Zip14 had the highest induction. For all graphs, n ϭ 5 mice/genotype Ϯ SE. DUO, duodenum. E260 ZINC TRANSPORTER Zip14 AND ADIPOSE TISSUE REGULATION AJP-Endocrinol Metab • doi:10.1152/ajpendo.00421.2015 • www.ajpendo.org
  • 4. normalized to cell number. To generate confocal images, 3T3-L1 cells were passaged into two-well chambered coverglass (Thermo Fisher Scientific) prior to siRNA treatment and a subsequent 8-day differ- entiation period. Imaging was performed at the University of Florida Cell & Tissue Analysis Core with a Leica TCS SP5 laser-scanning confocal microscope, using a ϫ40 oil objective. LAS-AF imaging software was used for image analysis. Statistics. Statistical analyses was performed using SAS verison 9.2 (SAS Institute, Cary, NC). The effects of WT vs. KO genotype were compared using Student’s t-tests. The independent effects of genotype and antibiotic were analyzed using the Proc Mixed procedure (SAS) with mouse within treatment as the random effect. Multiple compar- ison significance was analyzed using the Tukey adjustment. Reported values represent means Ϯ SE. RESULTS Zip14 is highly expressed in WAT during acute inflammation. Relative Zip14 mRNA abundance in WAT of WT mice is low compared with that found in duodenum or liver but greater than that found in muscle (Fig. 1A). Aydemir et al. (3) demonstrated that the highest induction of liver Zip14 mRNA occurred 18 h post-ip injection of LPS. Therefore, the 18-h time point was used to examine the comparative expression of Zip14 across tissues during LPS challenge (Fig. 1B). Expression of Zip14 mRNA increased two- and 32-fold in liver and WAT, respec- tively, with LPS administration. A transcript screening of 24 zinc transporters was performed. The transporter mRNAs that were significantly (P Յ 0.05) altered by LPS are shown (Fig. 1C). With the exception of ZnT3 and ZnT8, all zinc transporter genes were expressed in WAT. Zip14 had the greatest LPS induction of the zinc transporters. LPS-induced Zip14 expression is correlated with upregu- lated cytokine expression. To compare WAT with our previous liver findings (3), we next sought to evaluate the induction of WAT ZIP14 during acute LPS challenge. The endotoxin- induced acute phase response was shown through increased expression of Il-6, Tnf␣, and Il-1␤ mRNA (Fig. 2A). The induction of Zip14 expression peaked at 6 h post-ip injection and preceded the 9-h peaks of cytokine transcripts (Fig. 2A). Fig. 3. Zip14-knockout (KO) mice have enhanced levels of circulating endotoxin, predicating an inflammatory state characterized by adipose hypertrophy and dampened insulin signaling. A: colorimetrically determined that serum endotoxin was higher in Zip14-KO mice; n ϭ 3/genotype Ϯ SE (left). Proinflammatory signaling pathways in wild-type (WT) and KO mice are shown in WAT (right). Pooled lysates are depicted; lanes are triplicate repeats. B: expression of cytokines Il-6, Tnf␣, Il-1␤, and adiponectin (Adpn) in WT and KO mice. Preadipocyte marker [plasminogen activator inhibitor-1 (Pai-1) and preadipocyte factor-1 (Pref-1)] and differentiation marker (Ppar␥) are also depicted; n ϭ 28 mice/genotype Ϯ SE. C: serum and WAT leptin concentrations in WT and KO mice; n ϭ 3 mice/genotype Ϯ SE. ELISA values from WAT were normalized to total protein. D: representative hematoxylin and eosin (H & E) images of WAT from WT and KO adipose. Cell areas are from n ϭ 10 images/genotype Ϯ SE. Bars, 150 ␮m. E: transcripts for key adipogenic enzymes (n ϭ 28 mice/genotype Ϯ SE) and Western analysis of lipolytic markers (n ϭ 3 mice/genotype). F: Western analysis of insulin signaling (n ϭ 3 mice/genotype). MyD88, myeloid differentiation primary response gene; mTOR, mammalian target of rapamycin; ZAG, zinc ␣2-glycoprotein. E261ZINC TRANSPORTER Zip14 AND ADIPOSE TISSUE REGULATION AJP-Endocrinol Metab • doi:10.1152/ajpendo.00421.2015 • www.ajpendo.org
  • 5. Serum hypozincemia occurred as expected (3, 5); however, adipose levels of zinc fluctuated over the course of the LPS challenge (Fig. 2B). This finding suggests that a redistribution of zinc occurs within adipose during inflammation. The up- regulation of cytokine mRNA transcripts coincided with in- creased ZIP14 protein levels, as shown in the Western blots from adipose lysates (Fig. 2C). In adipose, the acute-phase response is associated with increased markers of lipolysis and decreased markers of differentiation (14, 23, 64). Our data confirmed findings of others in that the phosphorylation of HSL (HSL660) was upregulated during endotoxemia. In con- trast, phosphorylated insulin receptor and PPAR␥ were down- regulated, which suggested a loss of adipocyte differentiation. Zip14 KO mice have hypertrophy of adipocytes and greater leptin production. It is apparent that ZIP14 is highly induced by endotoxin in WAT. Previously, we established that global Zip14-KO impaired intestinal barrier function, which precipi- tates systemic endotoxemia (25). Therefore, we hypothesized that systemic endotoxemia would enhance the inflammatory status of adipose tissue. In confirmation of our previous find- ings, plasma endotoxin levels were greater in KO mice (Fig. 3A). We first sought to characterize the phenotypic profile of KO adipose. Enhanced expression of MyD88 (an adaptor protein that facilitates LPS induction of IL-6), NF-␬B, and STAT3 is indicative of WAT inflammation in the KO mice (Fig. 3A, right). In agreement, the steady-state levels of Il-6, Tnf␣, and Il-1␤ mRNAs were greater in WAT from the KO mice (Fig. 3B). Similarly, upregulation of Pref-1 (29, 58) and Pai-1 (35) mRNAs combined with downregulation of Ppar␥ in the WAT suggests a proinflammatory state, a characteristic of preadipocytic cells (Fig. 3B). Adiponectin (Adpn) expression, a hormone positively correlated with insulin sensitivity, was reduced with KO. Leptin protein levels were significantly (P Ͻ 0.05) greater in both KO serum and WAT (Fig. 3C). Moreover, cell areas of KO adipocytes were on average 40% larger than WT adipocytes (Fig. 3D). KO adipose had increased expres- sion of key adipogenic enzyme transcripts and altered lipid homeostasis (Fig. 3E). Compared with WT mice, steady-state lipolysis markers, p-HSL (HSL660), and ZAG (39) were re- duced in WAT from KO mice. The observed dyslipidemia was accompanied by reduced PPAR␥ levels (Fig. 3E). Overall, the KO mutation appeared to enhance adiposity and depress adi- pocyte differentiation. An evaluation of steady-state insulin signaling revealed that KO WAT had decreased phosphoryla- Fig. 4. Zip14 KO enhances LPS-induced inflammation. The effects of the KO are reversed with oral antibiotics (AB). A: WT and KO mice were administered LPS (2 mg/kg ip) for 6 h. Zip14, Tnf␣, and Il-1␤ mRNAs were measured by quantitative PCR. Serum and WAT IL-6 concentrations were measured by ELISA; n ϭ 3 mice/treatment Ϯ SE. In the absence of LPS, baseline IL-6 levels in either genotype were not detected (ND). B: WT and KO mice were provided AB in drinking water (0.5 mg/ml neomycin and 1 mg/ml ampicillin) for 4 wk. WAT expression of Il-6, Tnf␣, and Il-1␤ mRNAs in WT and KO mice with AB treatment; n ϭ 5 mice/treatment Ϯ SE. C: H & E images of WAT were used to measure cell areas; n ϭ 10 images/treatment Ϯ SE. Bars, 150 ␮m. D: Western analysis of WAT insulin signaling in WT and KO mice with AB treatment; n ϭ 3 mice/treatment. E262 ZINC TRANSPORTER Zip14 AND ADIPOSE TISSUE REGULATION AJP-Endocrinol Metab • doi:10.1152/ajpendo.00421.2015 • www.ajpendo.org
  • 6. tion of the insulin receptor (IR), protein kinase B (Akt), and mTOR (Fig. 3F) (9). These data show that KO adipose dem- onstrates a phenotype of dampened insulin signaling. Further- more, data in Fig. 3F also show that the macrophage marker (F4/80) is not different between genotypes, suggesting minimal influence of macrophage infiltration (59) on our observed KO phenotype. The Zip14 phenotype is enhanced with LPS challenge and minimized with oral antibiotics. Given the systemic endotox- emia in the KO, we hypothesized that the phenotype would be exacerbated by acute LPS and improved with antibiotics (AB). The highest induction of Zip14 mRNA during our LPS time course occurred 6 h post-ip injection (Fig. 1C). Therefore, we used the 6-h LPS response as the point of comparison between genotypes. The loss of Zip14 caused higher induction of WAT cytokine transcripts (Fig. 4A, left) along with serum IL-6 (Fig. 4A, right). These findings suggest that ZIP14 may serve as a negative regulator of cytokine induction. In contrast to the exacerbating effects of LPS, the impact of endotoxin-induced inflammation was reduced with AB. Expression of Il-6 and Tnf␣ mRNAs in KO WAT was reduced with AB (Fig. 4B). Furthermore, AB prevented the adipocyte hypertrophy seen in KO WAT (Fig. 4C). The AB treatment also normalized insulin signaling between the genotypes (Fig. 4D). Upregulated cytokine pathways due to the KO mutation lead to impaired capacity to differentiate. In an effort to eliminate the effect of circulating endotoxin on KO adipose, mesenchy- mal stem cells were cultured in the absence of LPS. Stem cells were derived from WT and KO ears and differentiated into adipocytes. Zip14-KO was confirmed with quantitative PCR (Fig. 5A). We hypothesized that KO cells would exhibit en- hanced steady-state cytokine signaling in culture. Cytokine mRNA expression (Il-6 and Il-1␤) was higher in fully differ- entiated KO cells. Mt1 expression was not significantly differ- ent between genotype but tended to be lower in KO cells. Additionally, KO preadipocytic markers Pref-1 (29, 58) and Pai-1 (35) were upregulated (Fig. 5A). Western analysis of both PPAR␥ and PREF-1 (Fig. 5B) confirmed our KO tissue findings (Figs. 3, B and E, and 4D). Although the same antibody was used for cells and tissue, persistent bands appear in the Western blots of primary mesenchymal stem cells (Fig. 5B). However, this same persistent banding was again noted in siRNA-treated 3T3-L1 cells, which may serve as our negative control for cellular ZIP14 expression (Fig. 6F). Oil Red O staining also demonstrated that cultured KO cells are more preadipocytic and have a lower capacity to differentiate and accumulate lipids (Fig. 5C, top). It was not until cells were cultured in LPS that KO cells began to overproduce lipids (Fig. Fig. 5. Stem cells from Zip14-KO mice are inflammatory and preadipocytic in culture. Primary stem cells from WT and KO mice were cultured in 3T3-L1 growth media. Upon confluence, cells were placed in differentiation medium and cultured for 10 days. A: transcript levels of Zip14, cytokine (Il-6, Tnf␣, and Il-1␤), Mt1, and preadipocytic markers (Pref-1 and Pai-1) were measured; n ϭ 3 pooled wells Ϯ SE. B: Western analysis of the preadipocytic markers in WT and KO cells; n ϭ 3 pooled wells Ϯ SE. C: accumulation of lipids in WT- and KO-derived cells, as measured by Oil Red O (Ϯ 10 ng/ml LPS for 10 days). D: Oil Red O was eluted from cell cultures and measured colorimetrically and normalized to protein; n ϭ 3 wells/treatment Ϯ SE. E263ZINC TRANSPORTER Zip14 AND ADIPOSE TISSUE REGULATION AJP-Endocrinol Metab • doi:10.1152/ajpendo.00421.2015 • www.ajpendo.org
  • 7. 5C, bottom). Quantification of Oil Red O confirmed our visual observations (Fig. 5D). ZIP14-KO alters intracellular zinc, which enhances key inflammatory pathways. Since the focus of the molecular site responsible for the KO phenotype is likely related to metal transport, it was necessary to place those findings within a function of dyshomeostasis of cellular zinc metabolism. 65 Zn was administered orally to measure zinc uptake/retention. KO mice accumulated more zinc (65 Zn) in WAT. Figure 6A shows that Mt1 mRNA expression, a surrogate measure of cytosolic zinc, is reduced. Adipogenic 3T3-L1 cells transfected with siRNA had a pattern of zinc distribution similar to KO tissue. Specifically Zip14 knockdown increased total zinc in cells, whereas Mt1 mRNA was decreased (Fig. 6B). Intracellular labile zinc was visualized with a zinc probe, fluozin-3 AM. Fluorescence was detected using laser-scanning confocal mi- croscopy (Fig. 6C). The silencing of Zip14 with siRNA clearly resulted in increased (P Ͻ 0.0001) vesicular zinc. Transfection with siRNA increased expression of ZIP8, a transporter that is a homologue of ZIP14 (Fig. 6D) (31). These in vitro data are compatible with the hypothesis that less functional cellular zinc is available in adipocytes of KO mice. This finding is indica- tive of the “zinc trap” hypothesis associated with Zip14 abla- tion that we advanced earlier (25) in mouse intestine. Zip14 siRNA clearly resulted in increased expression of Il-6 and Tnf␣ (P Ͻ 0.05) and to a lesser extent Il-1␤ (Fig. 6E). Zinc’s ability to inhibit key cytokine pathways is one way in which zinc may regulate cytokine expression. We hypothe- sized that zinc trapped within vesicles, through either Zip14 ablation in mice or with siRNA transfection in cells, would limit the inhibition of key proinflammatory signaling cascades by zinc. With the exception of A20 (11), Zip14 knockdown enhanced activation of both the JAK2/STAT3 and NF-␬B pathways in 3T3-L1 cells (Fig. 6F). Upregulation of these pathways depressed the ability of cells to differentiate (upregu- lation of PREF-1 expression) and increase cytokine expression. As shown in the model presented in Fig. 7, zinc transported by ZIP14 appears to impact the TLR4 pathway as early as MyD88 and also influences signaling further downstream, e.g., phos- phorylated I␬B␣ (p-I␬B␣) and NF-␬B. Zip14 knockdown Fig. 6. Zip14-KO adipose and 3T3-L1 adipocytes treated with Zip14 siRNA display altered zinc homeostasis and enhanced proinflammatory signaling though the NF-␬B and JAK2/STAT3 pathways. A: WT and KO mice received 65 Zn by gavage, and WAT zinc accumulation was calculated from 65 Zn-specific activity. Mt1 mRNA was measured as an index of available intracellular zinc; n ϭ 10 mice/genotype Ϯ SE. B: 3T3L1 cells were transfected with Zip14 siRNA. Total zinc (AAS) and Mt1 mRNA; n ϭ 3 plates/group replicated 3 times. C: representative laser-scanning confocal images used to detect labile zinc pools using FluoZin-3 AM fluorescence. FluoZin-3 AM was quantified and normalized to cell count; n ϭ 24 wells/group replicated twice. D: representative Western blots show the effect of Zip14 siRNA on ZIP14 and ZIP8 (n ϭ 2 replicated 3 times). E: relative mRNA expression of Zip14, Il-6, Tnf␣, and Il-1␤ in Zip14 siRNA-transfected 3T3-L1 cells Ϯ 10 ng/ml LPS (n ϭ 3 wells/group Ϯ SE). F: representative Western blots show the effect of Zip14 siRNA on proinflammatory signaling pathways (n ϭ 4 replicated 3 times). SCR, scrambled; RFU, relative fluorescence units. E264 ZINC TRANSPORTER Zip14 AND ADIPOSE TISSUE REGULATION AJP-Endocrinol Metab • doi:10.1152/ajpendo.00421.2015 • www.ajpendo.org
  • 8. limits cytosolic availability of zinc, and inhibition of NF-␬B is lifted, allowing enhanced induction of proinflammatory genes Il-6, Tnf␣, and Il-1␤ (Fig. 6E). Similarly, Zip14 deficiency enhanced JAK2 and STAT3 activity (Fig. 3A, right) with a concomitant stimulation of the Il-6 transcript (Fig. 3B) that could in turn execute an autocrine response leading to in- creased leptin production and secretion (Fig. 3C). DISCUSSION Here we show for the first time that deletion of Zip14 impacts adipose function. Although Zip14 was first cloned from adipogenic 3T3-L1 cells (53), little has been reported regarding its expression/function in adipose metabolism. In this report, it is shown that KO alters zinc signaling pathways, intracellular zinc trafficking, and ultimately adipocyte metab- olism. Inflammation, expansion of tissue mass, and recruitment of inflammatory cells are normal functions of healthy adipose (49, 60). However, a prolonged inflammatory response will even- tually lead to metabolic alterations within WAT. Acute inflam- mation impacts ZIP14 expression in a tissue-specific manner (3, 25). From the experiments described to this point, it has been established that adipose ZIP14 is highly responsive to LPS. Zip14 transcript abundance was upregulated 30-fold by 18 h after LPS injection, suggesting a critical role for ZIP14 in adipose inflammatory response. These data suggest that up- regulation of Zip14 expression in WAT is kinetically more rapid than liver expression (3). Furthermore, Zip14 ablation drastically influences cytokine and leptin production along with signaling pathway activity in WAT. Upregulated Il-6 production with KO may be responsible for the induction of leptin secretion (55). We interpret these responses as being indicative of a greater requirement for ZIP14 and/or zinc during inflammatory challenge (43). The reversal of the KO phenotype with antibiotics supports this hypothesis. The findings with antibiotics are noteworthy in that the treatment was a cocktail of neomycin and ampicillin. Neomy- cin in particular, is poorly absorbed through the gastrointestinal tract (15). Therefore, these antibiotics target intestinal micro- flora populations directly, a finding that suggests that intestinal microflora (along with any treatment that alters the microfloral load, dietary or otherwise) impacts adipose development and function. In fact, the tie between microflora and adipose has been established previously by Ley et al. (30). The impact of KO on the intestinal microfloral load is certainly relevant considering the proposed links between the microbiome and metabolic diseases, a finding that we plan to evaluate in further studies. Relevant to this report is that the in vivo phenotype was successfully modeled in vitro using LPS alone, confirming that KO predisposes cells to an inflammatory phenotype that is exacerbated with LPS. Increased intestinal permeability increased serum endotoxin in KO mice (25). This finding was of particular interest, as chronic exposure to endotoxin leads to metabolic dysfunction. Metabolic endotoxemia is characterized by atypically high levels of circulating endotoxin, which produces low-grade, systemic inflammation (10, 34, 46). This inflammation has far-reaching physiological implications, including increased weight gain, hepatic insulin resistance (12, 26, 37), and mac- rophage recruitment (52, 59), all of which precede the devel- opment of type 2 diabetes upon the consumption of a high-fat diet. Luche et al. (35) found that a 28-day LPS pretreatment caused mice consuming high-fat diets to gain more weight than cohorts who had not received the LPS conditioning. The reported increase in body weight was coupled with an in- creased fat/lean ratio, an influx of small inflammatory preadi- Fig. 7. Proposed model for increased IL-6 and leptin production caused by global Zip14 deletion. Increased cytosolic zinc due to ZIP14 inhibits cytokine signaling. Sys- temic endotoxemia and/or LPS administration leads to Toll-like receptor 4 (TLR4) activation and induction of the NF-␬B pathway. Without surface expression of ZIP14, zinc is trapped in intracellular compartments, where it is unable to inhibit activities of the NF-␬B and JAK2/ STAT3 pathways, thereby increasing IL-6 and leptin production. E265ZINC TRANSPORTER Zip14 AND ADIPOSE TISSUE REGULATION AJP-Endocrinol Metab • doi:10.1152/ajpendo.00421.2015 • www.ajpendo.org
  • 9. pocytes, and an overall higher glycemic index (35). Unique to our KO model, dampened insulin signaling and low-grade inflammation were coupled with hypertrophy. Enhanced mark- ers of lipogenesis and decreased markers of lipolysis (8, 38) were also noted with KO. These markers are suggestive of a chronic (vs. acute; see Ref. 61) inflammatory state in KO WAT. Furthermore, these physiological differences occur in the absence of a dietary intervention such as the feeding of a high-fat diet. Inflammatory cytokines are well-known mediators of altered adipose function and development (12, 48). Furthermore, prea- dipocytes themselves are able to acquire phagocytic activity and express macrophage-specific antigens under acute inflam- matory conditions (13). Using cultures at tiered stages of differentiation (0, 50, and 90%), Chung et al. (14) elegantly demonstrated that preadipocytes were the primary producers of the endogenous cytokines responsible for endotoxin-induced suppression of insulin-stimulated glucose uptake. It has been shown previously that Zip14 mRNA is induced during the early stages of adipocyte differentiation (53). Here, we show that KO increased preadipocytic markers and enhanced cytokine ex- pression. The differentiation marker PPAR␥ was downregu- lated with KO. This finding is relevant, as PPAR␥ is zinc responsive and has the ability to inactivate STAT3 in myeloma cells (38, 56, 57). Zip14 KO limits the ability of mesenchymal stem cells to differentiate and accumulate lipids (Fig. 4). Although this may initially seem counterintuitive, limiting a preadipocyte’s ability to differentiate precipitates hypertrophic obesity in humans (24, 49). Hypertrophic obesity is the likely cause of our previous finding where KO mice had enhanced liver lipids (3). Aberrant zinc signaling with Zip14 KO impacts the growth and development of adipocytes. Here, we report a phenotype characterized by high tissue zinc but low Mt1 expression. Trayhurn et al. (54) found that adipose expres- sion of Mt1 was not influenced by subcutaneous zinc injec- tion. However, experiments conducted with metallothion- ein-KO (7) revealed that MtϪ/Ϫ mice had increased visceral WAT weight coupled with adipocyte hypertrophy and higher levels of circulating leptin (7). Furthermore, this phenotype was exacerbated by high-fat feeding (44). Previ- ous literature has reported conflicting relationships between serum leptin and fat pad zinc content (2, 41, 51). In humans, obesity has long been associated with hypozincemia. In terms of zinc transporters, obesity is correlated with down- regulated ZnT (Ϫ2, Ϫ3, Ϫ6, and Ϫ8) and ZIP (Ϫ1, Ϫ2, Ϫ3, Ϫ4, Ϫ5, Ϫ6, and Ϫ7) expression in subcutaneous adipose (47). Our data shows that Zip14 deletion causes adipose to act as if it were experiencing a zinc deficiency. Evidence for that includes the lower Mt1 mRNA levels in WAT of the KO mice (Fig. 6A). This zinc-deficient status apparently cannot be overcome by the concomitant upregulation of zinc trans- porters, most notably ZIP8 (Fig. 6D). The ability of adi- pocytes to regulate zinc trafficking may determine how adipose is remodeled during tissue expansion. Furthermore, ZIP14 appears to lie at the plastic interface of adipose metabolism and inflammation. As shown in Fig. 7, our data show that ZIP14 influences adipocyte cytokine expression through two well-established signaling pathways, NF-␬B and JAK2/STAT3 (6, 34, 55, 63). The adipose mass expresses TLR4 (45) and is a major secre- tory organ of both leptin and IL-6 (28). Hence, the demonstra- tion here that both are overproduced with Zip14 deletion points to ZIP14 acting to provide control over the production of both mediators. Zip14 deletion enhances phosphorylation of these signaling pathways and is a likely cause for the upregulation of cytokines and leptin observed with KO. It should be noted that the ZIP14 homolog ZIP8 has been shown to inhibit the TLR4 pathway though inhibition of I␬B kinase (IKK) (32). In this report, ZIP8 levels are greater with Zip14 ablation, demonstrat- ing that ZIP8 is unable to compensate for the upregulation of the NF-␬B pathway. Therefore, ZIP8-mediated inhibition of IKK may be of minor significance in our Zip14-KO adipose model. In summary, adipocytes with a Zip14 deletion appear to have an impaired ability to mobilize intracellular zinc. In our model, limited availability of intracellular zinc disinhibits key cyto- kine pathways. Adipocytes have enhanced cytokine signaling and are more preadipocytic with KO. ZIP14 knockdown cells did not produce excess lipids until they were cultured in LPS media. These results in part model hypertrophic WAT ob- served with KO in vivo. Furthermore, the KO phenotype of hypertrophy was diminished with oral antibiotics, indicating that low-grade inflammation is necessary to induce enhanced adiposity. Our results demonstrate that ZIP14 is critical to cytokine production in adipose tissue and that targeted zinc transport is critical to adipocyte development. ACKNOWLEDGMENTS Scanning confocal microscopy was performed at the University of Florida Core. We thank Dr. Joseph L. Purswell, US Department of Agriculture/ Agricultural Research Service at Mississippi State University, for assistance with the ImageJ analysis. GRANTS This project was supported by National Institute of Diabetes and Digestive and Kidney Diseases Grant R01-DK-094244 and the Boston Family Endow- ment Funds of the University of Florida to R. J. Cousins. DISCLOSURES The authors report no conflicts of interest, financial or otherwise. AUTHOR CONTRIBUTIONS C.T., T.B.A., and R.J.C. conception and design of research; C.T. and T.B.A. performed experiments; C.T. analyzed data; C.T., T.B.A., and R.J.C. inter- preted results of experiments; C.T. prepared figures; C.T., T.B.A., and R.J.C. drafted manuscript; C.T., T.B.A., and R.J.C. edited and revised manuscript; R.J.C. approved final version of manuscript. REFERENCES 1. Allred CC, Krennmayer T, Koutsari C, Zhou L, Ali AH, Jensen MD. A novel ELISA for measuring CD36 protein in human adipose tissue. J Lipid Res 52: 408–415, 2011. 2. Argani H, Mahdavi R, Ghorbani-haghjo A, Razzaghi R, Nikniaz L, Gaemmaghami SJ. Effects of zinc supplementation on serum zinc and leptin levels, BMI and body composition in hemodialysis patients. J Trace Elem Med Biol 28: 35–38, 2014. 3. Aydemir TB, Chang SM, Guthrie GJ, Maki AB, Ryu MS, Karabiyik A, Cousins RJ. Zinc transporter ZIP14 functions in hepatic zinc, iron and glucose homeostasis during the innate immune response (endotoxemia). PLoS One 7: e48679, 2012. 4. Aydemir TB, Liuzzi JP, McClellan S, Cousins RJ. Zinc transporter ZIP8 (SLC39A8) and zinc influence IFN-␥ expression in activated human T cells. J Leukoc Biol 86: 337–348, 2009. 5. Aydemir TB, Sitren HS, Cousins RJ. The zinc transporter Zip14 Influ- ences c-Met phosphorylation and hepatocyte proliferation during liver regeneration in mice. Gastroenterology 142: 1536–1546, 2012. E266 ZINC TRANSPORTER Zip14 AND ADIPOSE TISSUE REGULATION AJP-Endocrinol Metab • doi:10.1152/ajpendo.00421.2015 • www.ajpendo.org
  • 10. 6. Baker RG, Hayden MS, Ghosh S. NF-␬B, inflammation, and metabolic disease. Cell Metab 13: 11–22, 2011. 7. Beattie JH, Wood AM, Newman AM, Bremner I, Choo KH, Michal- ska AE, Trayhurn P. Obesity and hyperleptinemia in metallothionein (-I and -II) null mice. Proc Natl Acad Sci USA 95: 358–363, 1998. 8. Bing C, Bao Y, Jenkins J, Sanders P, Manieri M, Cinti S, Tisdale MJ, Trayhurn P. Zinc-alpha2-glycoprotein, a lipid mobilizing factor is ex- pressed in adipocytes and is up-regulated in mice with cancer cachexia. Proc Natl Acad Sci USA 101: 2500–2505, 2004. 9. Blagosklonny MV. TOR-centric view on insulin resistance and diabetic complications: perspective for endocrinologists and gerontologists. Cell Death Dis 4: e964, 2013. 10. Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, Neyrinck AM, Fava F, Tuohy KM, Chabo C, Waget A, Delmée E, Cousin B, Sulpice T, Cani PD, Bibiloni R, Knauf C, Waget A, Neyrinck AM, Delzenne NM, Burcelin R. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet- induced obesity and diabetes in mice. Diabetes 57: 1470–1481, 2008. 11. Catrysse L, Vereecke L, Beyaert R, van Loo G. A20 in inflammation and autoimmunity. Trends Immunol 35: 23–31, 2013. 12. Chamontin B, Ferrières J, Tanti JF, Gibson GR, Casteilla L, Delzenne NM, Alessi MC, Burcelin R. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 56: 1761–1772, 2007. 13. Charrière G, Cousin B, Arnaud E, André M, Bacou F, Pénicaud L, Casteilla L. Preadipocyte conversion to macrophage. Evidence of plas- ticity. J Biol Chem 278: 9850–9855, 2003. 14. Chung S, Lapoint K, Martinez K, Kennedy A, Boysen Sandberg M, McIntosh MK. Preadipocytes mediate lipopolysaccharide-induced in- flammation and insulin resistance in primary cultures of newly differen- tiated human adipocytes. Endocrinology 147: 5340–5351, 2006. 15. Croswell A, Amir E, Teggatz P, Barman M, Salzman NH. Prolonged impact of antibiotics on intestinal microbial ecology and susceptibility to enteric Salmonella infection. Infect Immun 77: 2741–2753, 2009. 16. Coulston L, Dandona P. Insulin-like effect of zinc on adipocytes. Diabetes 29: 665–667, 1980. 17. Cui W, Taub DD, Gardner K. qPrimerDepot: a primer database for quantitative real time PCR. Nucleic Acids Res 35: D805–D809, 2007. 18. de Luis DA, Pacheco D, Izaola O, Terroba MC, Cuellar L, Martin T. Zinc and copper serum levels of morbidly obese patients before and after biliopancreatic diversion: 4 years of follow-up. J Gastrointest Surg 15: 2178–2181, 2011. 19. de Luis DA, Pacheco D, Izaola O, Terroba MC, Cuellar L, Cabezas G. Micronutrient status in morbidly obese women before bariatric surgery. Surg Obes Relat Dis 9: 323–327, 2013. 20. Drolet R, Richard C, Sniderman AD, Mailloux J, Fortier M, Huot C, Rhéaume C, Tchernof A. Hypertrophy and hyperplasia of abdominal adipose tissues in women. Int J Obes (Lond) 32: 283–291, 2008. 21. Ferrante AW Jr. The immune cells in adipose tissue. Diabetes Obes Metab 15, Suppl 3: 34–38, 2013. 22. Galarraga M, Campión J, Muñoz-Barrutia A, Boqué N, Moreno H, Martínez JA, Milagro F, Ortiz-de-Solórzano C. Adiposoft: automated software for the analysis of white adipose tissue cellularity in histological sections. J Lipid Res 53: 2791–2796, 2012. 23. Grisouard J, Bouillet E, Timper K, Radimerski T, Dembinski K, Frey DM, Peterli R, Zulewski H, Keller U, Müller B, Christ-Crain M. Both inflammatory and classical lipolytic pathways are involved in lipopoly- saccharide-induced lipolysis in human adipocytes. Innate Immun 18: 25–34, 2012. 24. Gustafson B, Gogg S, Hedjazifar S, Jenndahl L, Hammarstedt A, Smith U. Inflammation and impaired adipogenesis in hypertrophic obesity in man. Am J Physiol Endocrinol Metab 297: E999–E1003, 2009. 25. Guthrie GJ, Aydemir TB, Troche C, Martin AB, Chang SM, Cousins RJ. Influence of ZIP14 (slc39A14) on intestinal zinc processing and barrier function. Am J Physiol Gastrointest Liver Physiol 308: G171– G178, 2015. 26. Henninger AM, Eliasson B, Jenndahl LE, Hammarstedt A. Adipocyte hypertrophy, inflammation and fibrosis characterize subcutaneous adipose tissue of healthy, non-obese subjects predisposed to type 2 diabetes. PLoS One 9: e105262, 2014. 27. Jeong J, Eide DJ. The SLC39 family of zinc transporters. Mol Aspects Med 34: 612–619, 2013. 28. Kershaw E, Flier J. Adipose tissue as an endocrine organ. J Clin Endocrinol Metab 89: 2548–2556, 2004. 29. Kim YJ, Min TS, Seo KS, Kim SH. Expression of pref-1/dlk-1 is regulated by microRNA-143 in 3T3-L1 cells. Mol Biol Rep 42: 617–624, 2015. 30. Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Microbial ecology: human gut microbes associated with obesity. Nature 444: 1022–1023, 2006. 31. Lichten LA, Cousins RJ. Mammalian zinc transporters: nutritional and physiologic regulation. Annu Rev Nutr 29: 153–176, 2009. 32. Liu MJ, Bao S, Gálvez-Peralta M, Pyle CJ, Rudawsky AC, Pavlovicz RE, Killilea DW, Li C, Nebert DW, Wewers MD, Knoell DL. ZIP8 regulates host defense through zinc-mediated inhibition of NF-␬B. Cell Rep 3: 386–400, 2013. 33. Liuzzi JP, Aydemir F, Nam H, Knutson MD, Cousins RJ. Zip14 (Slc39a14) mediates non-transferrin-bound iron uptake into cells. Proc Natl Acad Sci USA 103: 13612–13617, 2006. 34. Liuzzi JP, Lichten LA, Rivera S, Blanchard RK, Aydemir TB, Knut- son MD, Ganz T, Cousins RJ. Interleukin-6 regulates the zinc transporter Zip14 in liver and contributes to the hypozincemia of the acute-phase response. Proc Natl Acad Sci USA 102: 6843–6848, 2005. 35. Luche E, Cousin B, Garidou L, Serino M, Waget A, Barreau C, André M, Valet P, Courtney M, Casteilla L, Burcelin R. Metabolic endotox- emia directly increases the proliferation of adipocyte precursors at the onset of metabolic diseases through a CD14-dependent mechanism. Mol Metab 2: 281–291, 2013. 36. May J, Contoreggi C. The mechanism of the insulin-like effects of ionic zinc. J Biol Chem 257: 4362–4368, 1982. 37. Makki K, Froguel P, Wolowczuk I. Adipose tissue in obesity-related inflammation and insulin resistance: cells, cytokines, and chemokines. ISRN Inflamm 2013: 139239, 2013. 38. Meerarani P, Reiterer G, Toborek M, Hennig B. Zinc modulates PPARgamma signaling and activation of porcine endothelial cells. J Nutr 133: 3058–3064, 2003. 39. Mracek T, Gao D, Tzanavari T, Bao Y, Xiao X, Stocker C, Trayhurn P, Bing C. Downregulation of zinc-{alpha} 2-glycoprotein in adipose tissue and liver of obese ob/ob mice and by tumour necrosis factor-alpha in adipocytes. J Endocrinol 204: 165–172, 2010. 40. O’Hara A, Lim F, Mazzatti DJ, Trayhurn P. Microarray analysis identifies matrix metalloproteinases (MMPs) as key genes whose expres- sion is up-regulated in human adipocytes by macrophage-conditioned medium. Pflugers Arch 458: 1103–1114, 2009. 41. Ott ES, Shay NF. Zinc deficiency reduces leptin gene expression and leptin secretion in rat adipocytes. Exp Biol Med (Maywood) 226: 841–846, 2001. 42. Rim J, Mynatt R, Gawronska-Kozak B. Mesenchymal stem cells from the outer ear: a novel adult stem cell model system for the study of adipogenesis. FASEB J 19: 1205–1207, 2005. 43. Rink L, Haase H. Zinc homeostasis and immunity. Trends Immunol 28: 1–4, 2007. 44. Sato M, Kawakami T, Kondoh M, Takiguchi M, Kadota Y, Himeno S, Suzuki S. Development of high-fat-diet-induced obesity in female metal- lothionein-null mice. FASEB J 24: 2375–2384, 2010. 45. Shi H, Kokoeva MV, Inouye K, Tzameli I, Yin H, Flier JS. TLR4 links innate immunity and fatty acid-induced insulin resistance. J Clin Invest 116: 3015–3025, 2006. 46. Siebler J, Galle PR, Weber MM. The gut-liver-axis: endotoxemia, inflammation, insulin resistance and NASH. J Hepatol 48: 1032–1034, 2008. 47. Smidt K, Pedersen SB, Brock B, Schmitz O, Fisker S, Bendix J, Wogensen L, Rungby J. Zinc-transporter genes in human visceral and subcutaneous adipocytes: lean versus obese. Mol Cell Endocrinol 264: 68–73, 2007. 48. Song MJ, Kim KH, Yoon JM, Kim JB. Activation of Toll-like receptor 4 is associated with insulin resistance in adipocytes. Biochem Biophys Res Commun 346: 739–745, 2006. 49. Stefan N, Kantartzis K, Machann J, Schick F, Thamer C, Rittig K, Balletshofer B, Machicao F, Fritsche A, Haring HU. Identification and characterization of metabolically benign obesity in humans. Arch Intern Med 168: 1609–1616, 2008. 50. Suliburska J, Bogdanski P, Pupek-Musialik D, Krejpcio Z. Dietary intake and serum and hair concentrations of mineral and their relationship with serum lipids and glucose levels in hypertensive and obese patients with insulin resistance. Biol Trace Elem Res 139: 137–150, 2011. 51. Tallman DL, Taylor CG. Effects of dietary fat and zinc on adiposity, serum leptin and adipose fatty acid composition in C57BL/6J mice. J Nutr Biochem 14: 17–23, 2003. E267ZINC TRANSPORTER Zip14 AND ADIPOSE TISSUE REGULATION AJP-Endocrinol Metab • doi:10.1152/ajpendo.00421.2015 • www.ajpendo.org
  • 11. 52. Tam CS, Heilbronn LK, Henegar C, Wong M, Cowell CT, Cowley MJ, Kaplan W, Clément K, Baur LA. An early inflammatory gene profile in visceral adipose tissue in children. Int J Pediatr Obes 6: e360–e363, 2011. 53. Tominaga K, Kagata T, Johmura Y, Hishida T, Nishizuka M, Imagawa M. SLC39A14, a LZT protein, is induced in adipogenesis and transports zinc. FEBS J 272: 1590–1599, 2005. 54. Trayhurn P, Duncan JS, Wood AM, Beattie JH. Metallothionein gene expression and secretion in white adipose tissue. Am J Physiol Regul Integr Comp Physiol 279: R2329–R2335, 2000. 55. Trujello ME, Sullivan S, Harten I, Schneider SH, Greenberg AS, Fried SK. Interleukin-6 regulates human adipose tissue lipid metabolism and leptin produc- tion in vitro. J Clin Endocrinol Metab 89: 5577–5582, 2004. 56. Tornatore L, Thotakura AK, Bennett J, Moretti M, Franzoso G. The nuclear factor kappa B signaling pathway: integrating metabolism with inflammation. Trends Cell Biol 22: 557–566, 2012. 57. Wang LH, Yang XY, Zhang X, Huang J, Hou J, Li J, Xiong H, Mihalic K, Zhu H, Xiao W, Farrar WL. Transcriptional inactivation of STAT3 by PPARgamma suppresses IL6-responsive multiple myeloma cells. Immunity 20: 205–218, 2004. 58. Wang Y, Kim KA, Kim JH, Sul HS. Pref-1, a preadipocyte secreted factor that inhibits adipogenesis. J Nutr 136: 2953–2956, 2006. 59. Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Fer- rante AW Jr. Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest 112: 1796–1808, 2003. 60. Wernstedt Asterholm I, Tao C, Morley TS, Wang QA, Delgado-Lopez F, Wang ZV, Scherer PE. Adipocyte inflammation is essential for healthy adipose tissue expansion and remodeling. Cell Metab 20: 103–118, 2014. 61. Xu H, Barnes GT, Yang Q, Tan G, Yang D, Chou CJ, Sole J, Nichols A, Ross JS, Tartaglia LA, Chen H. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J Clin Invest 112: 1821–1830, 2003. 62. Zhanga Y, Goldmana S, Baergaa R, Zhaoa Y, Komatsub M, Jin S. Adipose-specific deletion of autophagy-related gene 7 (atg7) in mice reveals a role in adipogenesis. Proc Natl Acad Sci USA 106: 19860– 19865, 2009. 63. Zhou Z, Wang L, Song Z, Saari JT, McClain CJ, Kang J. Abrogation of nuclear factor-␬B activation is involved in zinc inhibition of lipopoly- saccharide-induced tumor necrosis factor-␣ production and liver injury. Am J Pathol 164: 1547–1556, 2004. 64. Zu L, He J, Jiang H, Xu C, Pu S, Xu G. Bacterial endotoxin stimulates adipose lipolysis via toll-like receptor 4 and extracellular signal-regulated kinase pathway. J Biol Chem 284: 5915–5926, 2009. E268 ZINC TRANSPORTER Zip14 AND ADIPOSE TISSUE REGULATION AJP-Endocrinol Metab • doi:10.1152/ajpendo.00421.2015 • www.ajpendo.org