ArticlePDF Available

Regulation of tumour necrosis factor-alpha release from human adipose tissue in vitro

Authors:

Abstract and Figures

Tumour necrosis factor-alpha (TNF-alpha), secreted by cells of the macrophage-monocyte lineage, has a well established role in inflammation and host-defence. The more recent discovery that adipocytes also secrete TNF-alpha has led to a substantial body of research implicating this molecule in the insulin resistance of obesity. However, little is known about the normal regulation of TNF-alpha release from human adipose tissue. In particular, it is not known whether adipocyte production of TNF-alpha is responsive to similar or different molecular regulators than those relevant to macrophages. TNF-alpha release from cultured human adipose tissue and isolated adipocytes was examined using an ELISA. Insulin, cortisol or the thiazolidinedione, BRL 49653, did not have a significant effect on TNF-alpha release from adipose tissue or isolated adipocytes. In contrast, lipopolysaccharide (LPS), a major stimulus of TNF-alpha protein production in monocytes and macrophages, resulted in a fivefold stimulation of TNF-alpha release from human adipose tissue. Significant stimulation of TNF-alpha release was also seen from isolated adipocytes, indicating that the increase in TNF-alpha release from adipose tissue in the presence of LPS is unlikely to be entirely attributable to contaminating monocytes or macrophages. Consistent with this observation was the finding that mRNA for CD14, a known cellular receptor for LPS, is expressed in human adipocytes. The increase in TNF-alpha protein release in response to LPS was blocked by an inhibitor of the matrix metalloproteinase responsible for the cleavage of the membrane-bound proform of TNF-alpha, indicating that this release represented regulated secretion and was not due to cell lysis. In conclusion, the regulation of TNF-alpha protein release from human adipose tissue and isolated adipocytes appears to be similar to its regulation in cell types more traditionally implicated in host defence. The production by the adipocyte of a range of molecules involved in host defence-TNF-alpha, factors D, B and C3, interleukin-6, and macrophage colony-stimulating factor--suggest that this cell type may make a significant contribution to innate immunity.
Content may be subject to copyright.
Regulation of tumour necrosis factor-alpha release from human
adipose tissue in vitro
C P Sewter, J E Digby, F Blows, J Prins and S O’Rahilly
Departments of Medicine and Clinical Biochemistry, University of Cambridge, Addenbrooke’s Hospital, Hills Road, Cambridge CB2 2QQ, UK
(Requests for offprints should be addressed to C P Sewter)
Abstract
Tumour necrosis factor-alpha (TNF-á), secreted by cells
of the macrophage-monocyte lineage, has a well estab-
lished role in inflammation and host-defence. The more
recent discovery that adipocytes also secrete TNF-á has
led to a substantial body of research implicating this
molecule in the insulin resistance of obesity. However,
little is known about the normal regulation of TNF-á
release from human adipose tissue. In particular, it is not
known whether adipocyte production of TNF-á is
responsive to similar or dierent molecular regulators than
those relevant to macrophages. TNF-á release from cul-
tured human adipose tissue and isolated adipocytes was
examined using an ELISA. Insulin, cortisol or the thiazo-
lidinedione, BRL 49653, did not have a significant eect
on TNF-á release from adipose tissue or isolated adi-
pocytes. In contrast, lipopolysaccaride (LPS), a major
stimulus of TNF-á protein production in monocytes and
macrophages, resulted in a fivefold stimulation of TNF-á
release from human adipose tissue. Significant stimulation
of TNF-á release was also seen from isolated adipocytes,
indicating that the increase in TNF-á release from adipose
tissue in the presence of LPS is unlikely to be entirely
attributable to contaminating monocytes or macrophages.
Consistent with this observation was the finding that
mRNA for CD14, a known cellular receptor for LPS, is
expressed in human adipocytes. The increase in TNF-á
protein release in response to LPS was blocked by an
inhibitor of the matrix metalloproteinase responsible for
the cleavage of the membrane-bound proform of TNF-á,
indicating that this release represented regulated secretion
and was not due to cell lysis.
In conclusion, the regulation of TNF-á protein release
from human adipose tissue and isolated adipocytes appears
to be similar to its regulation in cell types more tradition-
ally implicated in host defence. The production by the
adipocyte of a range of molecules involved in host defence
TNF-á, factors D, B and C3, interleukin-6, and
macrophage colony-stimulating factor suggest that this
cell type may make a significant contribution to innate
immunity.
Journal of Endocrinology (1999) 163, 33–38
Introduction
Tumour necrosis factor-alpha (TNF-á) is a proinflam-
matory cytokine secreted by several cell types such as
monocytes (Waage & Bakke 1988), macrophages
(Mijatovic et al. 1997) and adipocytes (Hotamisligil et al.
1995). It is produced as a 26 kDa active membrane-bound
precursor that is proteolytically cleaved by a matrix
metalloproteinase (TNF-á converting enzyme (TACE))
to release a 17 kDa soluble form (Gearing et al. 1994). Its
expression and production in macrophages have been
shown to be increased on exposure to endotoxins such as
lipopolysaccharide (LPS) (Mijatovic et al. 1997), a compo-
nent of the cell wall of Gram-negative bacteria. TNF-á
has an important role as a mediator of the acute phase
response and, in addition to an immunological function, it
has been shown to have a variety of eects on lipid
metabolism and adipocyte function. These include the
stimulation of lipolysis through increases in hormone-
sensitive lipase expression (Sumida et al. 1997) and inhi-
bition of lipoprotein lipase (Fried & Zechner 1989),
induction of preadipocyte dedierentiation (decreasing
CCAAT/enhancer binding protein (Stephens & Pekala
1991) and peroxisome proliferator-activated receptor
gamma (PPARã) (Zhang et al. 1996)), promotion of
adipocyte apoptosis (Prins et al. 1997) and induction of
insulin resistance (decreasing GLUT4 (Hauner et al. 1995)
and insulin-stimulated autophosphorylation of insulin
receptor and phosphorylation of insulin receptor
substrate-1 (Feinstein et al. 1993)). Expression of TNF-á
in adipose tissue has been shown to be increased in rodent
and human obesity and to correlate with insulin resistance
(Hotamisligil et al. 1995, Kern et al. 1995). As a result of
these observations, the paracrine eects of TNF-á released
by adipocytes have been proposed to underlie the link
between obesity and insulin resistance (Hotamisiligil et al.
1993). Despite the potential pathogenic importance of
adipocyte TNF-á in obesity remarkably little is known
33
Journal of Endocrinology (1999) 163, 33–38
0022–0795/99/0163–0033 1999 Society for Endocrinology Printed in Great Britain
Online version via http://www.endocrinology.org
about the normal role of TNF-á in human adipose tissue.
We reasoned that a better definition of the molecular
regulators of TNF-á in human adipose tissue might
provide clues to the function of this cytokine in normal
human fat cell biology.
Materials and Methods
Acquisition of human tissue
Omental and subcutaneous adipose tissue biopsies
(approximately 3 g) were obtained from patients under-
going elective open surgery. None of the patients had
diabetes or severe systemic illness. Cambridge Local
Research Ethics Committee approval was obtained, and all
patients gave their informed consent. The tissue donor
group consisted of 11 women (ages 49·215·2 years;
body mass index (BMI) 25·94·3 kg/m
2
) and five men
(ages 71·28·8 years; BMI 264·5 kg/m
2
).
Adipose tissue culture
Whole adipose tissue was separated from surrounding
blood vessels, diced finely and washed twice in Hanks’
balanced salt solution (HBSS) containing 0·2% BSA. Small
pieces of tissue were then placed in sterile tissue culture
plates containing pre-equilibrated medium 199 containing
10% fetal bovine serum, 2 mM -glutamine, 100 U/ml
penicillin, 0·1 mg/ml streptomycin, 33 µM biotin,
17 µM pantothenic acid. Dierent test compounds
were also added to the medium: human insulin (7 µM),
cortisol (1 mg/ml), LPS (50 µg/ml), a thiazolidinedione
BRL 49653 (10
-7
M) (from SmithKline Beecham
Pharmaceuticals, Harlow, Essex, UK), a metalloproteinase
inhibitor BB3103 (10 µM) (from British Biotechnology,
Oxford, UK) and dimethyl sulphoxide. The plates were
then incubated under standard conditions (37 C/5%
CO
2
). After 20 h, medium was collected from each well
and frozen at 80 C before measurement of TNF-á
protein levels by ELISA. The tissue samples were collected
from each well, blotted onto filter paper to remove excess
medium, and weighed. TNF-á levels were expressed as
pg TNFá per mg adipose tissue per hour and fold increase
or decrease relative to the control (medium only). Unless
specifically indicated, all reagents were obtained from
Sigma Chemical Company, Poole, Dorset, UK.
Adipocyte isolation and culture
Adipose tissue biopsies were taken under sterile conditions
at the time of surgery and placed in normal saline. The
transport time to the laboratory was less than 1 h. The
tissue was diced finely into 1–2 mm pieces. All instru-
ments were sterile disposable or autoclaved. The diced
tissue was digested in a collagenase solution containing
HBSS, 3 mg/ml collagenase (type II) and 1·5% BSA. At
least three volumes more digest solution than the volume
of tissue was used. The digestion was carried out in a
shaking waterbath at 37 C for approximately 1 h. After
filtration through a 260 µm steel mesh, the adipocytes
were allowed to float to the surface and collected using a
pasteur pipette. After two washes in HBSS, aliquots of
adipocytes were dispensed into tissue culture plates con-
taining pre-equilibrated 199 medium and test compounds
(see adipose tissue culture). After 20 h, infranatants were
harvested from beneath the adipocyte layer and frozen at
80 C before measurement of TNF-á by ELISA. Re-
sults are expressed as pg TNFá per µl adipocyte suspension
per hour or percentage increase or decrease relative to the
control (medium alone).
TNF-á protein measurement
TNF-á levels were determined in culture medium using
a commercially available ELISA kit (Genzyme Diag-
nostics, Cambridge, MA, USA). The manufacturer’s
procedure for enhanced sensitivity was followed. The
assay range was 4–512 pg/ml, with a limit of sensitivity
of 500 fg/ml. The intra-assay coecient of variation was
9·67%, and the inter-assay coecent of variation was
12·47%.
Detection of CD14 mRNA by reverse
transcription-polymerase chain reaction
RT-PCR was carried out on RNA extracted from human
adipocytes, preadipocytes and monocyte cells. Adipocyte
and preadipocyte cells were isolated by collagense
digestion of whole adipose tissue as described above.
Monocyte cells were isolated from whole blood using
HISTOPAQUE-1077 from Sigma. The manufacturer’s
procedure was followed. RNA was extracted using a
guanidium thiocyanate–phenol technique (Tri Reagent,
Sigma). RNA samples were treated with RNase inhibitor
and DNase and quantified by spectrophotometry. The
integrity of RNA samples was assessed by agarose gel
electrophoresis and ethidium bromide staining. The
reverse transcription reaction was performed in a 20 µl
volume containing 5 mM MgCl
2
,1 reverse tran-
scriptase buer (Promega, Cambridge, UK), 1 mM
dNTPs (Pharmacia Biotech, St Albans, Herts, UK), 10 ng
random hexamers (Promega). The reactions were incu-
bated at 65 C for 5 min to remove any residual DNAase
A activity and allowed to cool to room temperature. One
microlitre Moloney murine leukemia virus reverse tran-
scriptase (Promega) was added to the reactions (except the
control) and incubated at 37 C for 1 h. The control
reaction, containing no reverse transcriptase, allowed for
the detection of any possible genomic DNA contami-
nation in the RNA samples after PCR. A further control
containing all reagents except RNA was also included to
C P SEWTER and others · Regulation of human adipose TNF release34
Journal of Endocrinology (1999) 163, 33–38
allow the detection of any possible DNA/RNA con-
tamination of reagents. PCR was then performed on
200 ng first-strand cDNA in a 50 µl volume containing
1 mM MgCl
2
, 0·2 µM each of CD14 sense (5-
GGAAAGAAGCTAAAGCACTT-3) and CD14 anti-
sense (5-TTTAGAAACGGCTCTAGGTT-3) primers,
0·15 mM dNTPs, 1 NH
4
reaction buer, 2·5 units
Advantage 2 Taq polymerase (Clontech, Palo Alto, CA,
USA). Cycling parameters were: 94 C for 1 min, 50 C
for 1 min, 68 C for 1 min (30 cycles). PCR products
were confirmed as CD14 by nucleotide sequencing.
All sequencing was carried out using Dideoxy termi-
nator chemistry (Perkin-Elmer, Foster City, CA, USA)
and electrophoresed on an ABI 377 automated DNA
sequencer.
Statistical analysis
A paired Wilcoxon non-parametric test was used for
statistical analysis. Results are expressed as means...
Results
Mean release of TNF-á from whole adipose tissue
and isolated adipocytes in culture medium was
0·0680·02 pg/mg adipose tissue per hour and
0·0170·01 pg/µl adipocyte suspension per hour respect-
ively. Stability of TNF-á protein in the medium was
confirmed by spiking medium containing whole adipose
tissue with known amounts of TNF-á and quantitating
TNF-á protein concentrations after a 20-h incubation
relative to an unspiked control (data not shown). There
was no correlation between TNF-á protein release and
BMI or age and no consistent gender dierences
were seen.
Exposure to insulin (7 µM), cortisol (1 mg/ml) or BRL
49653 (10
-7
M) had no significant eect on TNF-á release
from either whole adipose tissue (Fig. 1) or isolated
adipocytes (Fig. 2). In contrast, exposure to LPS resulted
in a mean fivefold (1·85 ...) increase in TNF-á
release from adipose tissue after 20 h (relative to controls)
(P=0·0134). Figure 3 shows that BB3103 (10 µM), a
matrix metalloproteinase inhibitor, was able to inhibit
LPS-stimulated TNF-á release from adipose tissue over
the 20-h period by 903·8% (P=0·002). BB3103
(10 µM) inhibited basal (unstimulated) TNF-á release
from adipose tissue by 3017·4% (P=NS). The inhibi-
tory eect of BB3103 suggests that the increase in release
of TNF-á from adipose tissue exposed to LPS is due to a
bona fide eect of LPS and not due to cell lysis. LPS also
had a stimulatory eect on TNF-á release from isolated
adipocytes (Fig. 2), although to a lesser extent compared
with whole adipose tissue (1·50·15-fold relative to
control; P=0·0469). In the presence of 10 µM BB3103,
this increase was inhibited by 7013·75% (P=0·03)
(Fig. 4). Under identical conditions, exposure of human
monocyte-macrophages to LPS resulted in a 50-fold
increase in TNF-á release relative to unstimulated controls
(P=<0·0001 data not shown).
Figure 1 TNF-á release into the incubation medium from human
adipose tissue after a 20-h incubation period in medium alone
(Control) or medium containing 50 µg/ml LPS (n=14), 7 µM insulin
(n=11), 1 mg/ml cortisol (n=11),or10
7
M BRL 49653 (n=11). In
each experiment, samples were incubated in duplicate. Data are
expressed as means
S.E.M. *P=0·0314 compared with Control.
Figure 2 TNF-á release into the incubation medium from isolated
human adipocytes after a 20-h incubation period in medium alone
(Control) or medium containing 50 µg/ml LPS (n=7), 7 µM insulin
(n=3), 1 mg/ml cortisol (n=3),or10
7
M BRL 49653 (n=4). In
each experiment, samples were incubated in duplicate. Data are
expressed as means
S.E.M. *P=0·0469 compared with Control.
Regulation of human adipose TNF release · C P SEWTER and others 35
Journal of Endocrinology (1999) 163, 33–38
Previous reports on the study of LPS induction of
TNF-á protein production in monocytes and macrophages
have shown that this is mediated by CD14, a cell-surface
receptor for LPS. CD14 mRNA was detected by RT-
PCR in isolated adipocytes, preadipocytes and monocytes
(Fig. 5).
Discussion
Although a large body of research on TNF-á action in
metabolically important tissue such as muscle and fat has
recently emerged, little information has been forthcoming
on the regulation of TNF-á release from adipose tissue,
and even less on TNF-á release from human adipose
tissue. In this study, we have demonstrated that TNF-á
release from human adipose tissue is responsive to LPS but
not to insulin, cortisol or a thiazolidinedione. LPS also
stimulated TNF-á release from isolated adipocytes, albeit
less robustly than in whole adipose tissue. The LPS-
stimulated release of TNF-á was prevented by an inhibitor
of the metalloproteinase responsible for the cleavage of the
membrane-bound form of TNF. Finally, adipocytes were
found to express mRNA encoding CD14, a known
receptor for LPS.
As in unstimulated monocytes and macrophages
(Crawford et al. 1997), TNF-á release from human
adipose tissue and isolated adipocytes under control con-
ditions occurred at low levels. This is consistent with our
previously published data concerning TNF-á mRNA
expression which indicated that this gene is expressed at
very low abundance in human adipocytes (Montague et al.
1998). Interestingly, we were unable to demonstrate any
correlation between adipose tissue TNF-á release and the
BMI of the subjects studied. This is in contrast to previous
studies which demonstrated a 2·5-fold increase in fat tissue
TNF-á expression in obese (BMI=39·91·4 kg/m
2
)
compared with lean (BMI=21·40·3 kg/m
2
) individuals
(Hotamisiligil et al. 1995). The narrow range of BMIs in
the present study (20–30 kg/m
2
) would not have allowed
us to detect an eect of marked obesity. This is consistent
with our previously published data concerning TNF-á
mRNA expression in adipocytes from a study population
with a similar range of BMI values (Montague et al. 1998).
Circulating TNF-á protein concentrations have been
reported to correlate with hyperinsulinaemia in humans
Figure 3 TNF-á release from human adipose tissue after a 20-h
incubation in medium alone (Control), medium plus BB3103
(10 µM), medium plus LPS (50 µg/ml) plus BB3103 vehicle, or
medium containing LPS (50 µg/ml) plus 10 µM BB3103 (n=10). In
each experiment, samples were incubated in duplicate. Data are
expressed as means
S.E.M. *P=0·0078, LPS+BB3103 vehicle
compared with Control; **P=0·002, LPS+BB3103 vehicle
compared with LPS+BB3103.
Figure 4 TNF-á release from isolated human adipocytes after a
20-h incubation in medium alone (Control), medium plus LPS
(50 µg/ml) plus BB3103 vehicle, or medium containing LPS
(50 µg/ml) plus 10 µM BB3103 (n=2). In each experiment,
samples were incubated in duplicate. Data are expressed as
means
S.E.M. *P=0·0497, LPS+BB3103 vehicle compared with
Control; **P=0·03, LPS+BB3103 vehicle compared with
LPS+BB3103.
C P SEWTER and others · Regulation of human adipose TNF release36
Journal of Endocrinology (1999) 163, 33–38
(Hotamisligil et al. 1995). In addition, insulin has been
reported to have a stimulatory eect on TNF-á release
from monocyte and macrophages (Gepner-Atlan et al.
1996, Hahn & Filkins 1993). In these studies, however,
insulin did not have any significant eect on release of
TNF-á protein from either adipose tissue or isolated
adipocytes. Cortisol has previously been shown to
markedly suppress the endotoxin-induced increase in
TNF-á protein production in human macrophages
(Beutler et al. 1986). No significant suppression of adipo-
cyte TNF-á by cortisol was seen in this study, although
there was a trend towards a reduction. The low basal levels
of TNF-á release may have made an inhibitory eect
dicult to detect, and future studies should consider the
eects of cortisol on LPS-induced secretion. Thiazolidin-
edione compounds are agonists at the PPARã receptor
(Berger et al. 1996), a nuclear hormone receptor highly
expressed in fat (Chawla et al. 1994). They act as insulin
sensitisers (Saltiel & Olefsky 1996) and, in obese rodents
treated in vivo have been demonstrated to reduce adi-
pocyte TNF-á expression (Peraldi et al. 1997). In contrast,
no eects of the thiazolidinedione, BRL 49653, on
TNF-á release from fat tissue were seen in the present
study. This apparent discrepancy may relate to species
dierences, to in vivo and in vitro dierences, or to the fact
that only fat from non-obese humans was studied.
LPS is a cell-wall component of Gram-negative bacteria
that powerfully stimulates TNF-á protein production in
monocytes and macrophages (Beutler et al. 1986). In this
study, we have demonstrated that LPS also stimulates
TNF-á release from human adipose tissue and from
isolated human adipocytes. The greater stimulation of
TNF-á from whole adipose tissue than from isolated
adipocytes may suggest that cells other than adipocytes are
responsible for most of the LPS response. However,
precise quantitative comparisons are dicult, as the iso-
lated adipocytes have inevitably undergone an isolation
procedure that may impair their subsequent responsiveness
in vitro. In this regard, Hotamisligil et al. (1993) have
reported that the majority of TNF-á expression in fat
tissue is in adipocytes, with a minor contribution from the
stromovascular fraction.
We have shown that the stimulatory eect of LPS on
TNF-á release can be blocked by BB3103, a matrix
metalloproteinase inhibitor, and that this eect is seen in
both whole adipose tissue and isolated adipocytes. These
findings suggest that the increase in TNF-á levels seen
on exposure to LPS occurs via normal physiological
mechanisms and is unlikely to represent a non-specific
lytic eect of LPS. Our results also demonstrate, for the
first time, that the release of TNF-á from adipocytes
occurs through the same metalloproteinase-mediated
mechanism as that seen in more classical TNF-á secreting
cells.
The finding that LPS stimulates TNF-á production
from human adipocytes suggests a role for adipocyte TNF
in host defence. This adds to a growing body of evidence
pointing towards a contribution of adipose tissue to innate
immunity. Thus adipocytes are a major source of several
components of the complement system including factors D
(adipsin), B and C3 (Peake et al. 1997). Levine et al. (1998)
have recently shown that adipocytes produce macrophage
colony-stimulating factor, a proliferative stimulus for mac-
rophages, and that this is upregulated in growing adipose
tissue, as seen in ‘creeping fat’ that surrounds inflammatory
intestinal lesions such as in Crohn’s disease. Adipose tissue
is also known to be a source of interleukin-6 (Fried et al.
1998), a pleiotropic cytokine that has a systemic role in
infections and inflammatory disorders as part of the acute
phase response. Finally, leptin, a major secretory product of
adipocytes, has important eects on lymphocyte function
(Lord et al. 1998). In addition, Gainsford and co-workers
(1996) found that addition of leptin to culture medium
enhanced cytokine production and phagocytosis of
Leishmania parasites by murine peritoneal macrophages.
Thus our finding that LPS is a significant secretagogue
for TNF-á release from human adipose tissue and adi-
pocytes adds to this growing body of evidence for a role of
the adipocyte in innate immunity. These findings do not,
however, exclude additional roles for adipocyte TNF-á in
the control of other aspects of fat cell biology, such as
dierentiation, apoptosis and lipid metabolism.
Acknowledgements
This study was supported by the British Diabetic Associ-
ation and the Wellcome Trust. J P is a Wellcome Trust
International Travelling Fellow. The authors would like to
thank Professor Ken Siddle for helpful discussions. The
authors would also like to acknowledge the support of
Dominic Corkill from British Biotechnology Limited. The
assistance of members of the departments of Surgery and
Figure 5 Detection of CD14 mRNA by RT-PCR in isolated adipocytes (A), monocytes (M) and
preadipocytes (PA). C, RT-PCR reactions in which reverse transcriptase enzyme was omitted; L,
1 kb ladder; RB, RT-PCR amplification mixture without template as a negative control.
Regulation of human adipose TNF release · C P SEWTER and others 37
Journal of Endocrinology (1999) 163, 33–38
Obstetrics and Gynaecology of Addenbrooke’s Hospital
and the University of Cambridge, in particular Peter
Friend, Neville Jamieson, John Williamson and Andrew
Prentice, is greatly appreciated.
References
Berger J, Bailey P, Biswas C, Cullinan CA, Doebber TW, Hayes NS,
Saperstein R, Smith RG & Leibowitz MD 1996 Thiazolidinediones
produce a conformational change in peroxisomal proliferator-
activated receptor-ã: binding and activation correlate with
antidiabetic actions in db/db mice. Endocrinology 137 4189–4195.
Beutler B, Krochin N, Milsark I, LuedkeC&Cerami A 1986
Control of cachectin (tumor necrosis factor) synthesis: mechanisms
of endotoxin resistance. Science 232 977–979.
Chawla A, Schwarz E, DimaculanganD&Lazar M 1994 Peroxisome
proliferator-activated receptor (PPAR) gamma: adipose-
predominant expression and induction early in adipocyte
dierentiation. Endocrinology 135 798–800.
Crawford E, Ensor J, KalvakolanuI&Hadsay J 1997 The role of 3
poly (A) tail metabolism in tumor necrosis factor-alpha regulation.
Journal of Biological Chemistry 272 21120–21127.
Feinstein R, Kanety H, Papa M, LunenfeldB&Karasik A 1993
Tumor necrosis factor-alpha suppresses insulin-induced tyrosine
phosphorylation of insulin-receptor and its substrates. Journal of
Biological Chemistry 268 26055–26058.
FriedS&ZechnerR1989 Cachectin tumor necrosis factor decreases
human adipose-tissue lipoprotein-lipase messenger-RNA levels,
synthesis, and activity. Journal of Lipid Research 30 1917–1923.
Fried S, BunkinD&Greenberg A 1998 Omental and subcutaneous
adipose tissues of obese subjects release interleukin-6: depot
dierence and regulation by glucocorticoid. Journal of Clinical
Endocrinology and Metabolism 83 847–850.
Gainsford T, Willson T, Metcali D, Handman E, McFarlane C, Ng
A, Nicola NA, Alexander WS & Hilton DJ 1996 Leptin can induce
proliferation, dierentiation, and functional activation of hemo-
poietic cells. Proceedings of the National Academy of Sciences of the
USA 93 14564–14568.
Gearing A, Beckett P, Christodoulou M, Churchill M, Clements J,
Davidson AH, Drummond AH, Galloway WA, Gilbert R, Gordon
JL, Leber TM, Mangan M, Miller K, Nayee P, Owen K, Patel S,
Thomas W, Wells G, Wood LM & Woolley K 1994 Processing of
tumor necrosis factor-á precursor by metalloproteinases. Nature 370
555–557.
Gepner-Atlan C, Bongrand P, Farnarier C, Xerri L, Choux R,
Gauthier J-F, Brue T, Vague P, Grob J-J & Vialettes B 1996
Insulin-induced lipoatrophy in type I diabetes. Diabetes Care 19
1283–1285.
HahnE&Filkins J 1993 Insulin restores plasma tumor-necrosis-factor
(TNF) in the dexamethasone protected endotoxic rat. FASEB
Journal 7 A420.
Hauner H, Petruschke T, Russ M, RohrigK&Eckel J 1995 Eects
of tumor necrosis factor-alpha (TNF-alpha) on glucose transport and
lipid metabolism in newly-dierentiated fat cells in culture.
Diabetologia 38 764–771.
Hotamisligil G, Shargill N & Speigelman B 1993 Adipose expression
of tumor necrosis factor-á: direct role in obesity-linked insulin
resistance. Science 259 87–91.
Hotamisligil G, Arner P, Caro J, AtkinsonR&Spiegelman B 1995
Increased adipose tissue expression of tumor necrosis factor-á in
human obesity and insulin resistance. Journal of Clinical Investigation
95 2409–2415.
Kern P, Saghizadeh M, Ong J, Bosch R, DeemR&SimsoloR1995
The expression of tumor necrosis factor in human adipose tissue.
Journal of Clinical Investigation 95 2111–2119.
Levine J, Jensen M, EberhardtN&OBrien T 1998 Adipocyte
macrophage colony-stimulating factor is a mediator of adipose tissue
growth. Journal of Clinical Investigation 101 1557–1564.
Lord G, Matarese G, Howard J, Baker R, BloomR&LechlerR
1998 Leptin modulates the T-cell immune response and reverses
starvation-induced immunosuppression. Nature 394 897–901.
Mijatovic T, Kruys V, Caput D, DefranceP&HuezG1997
Interleukin-4 and -13 inhibit tumor necrosis factor alpha mRNA
translational activation in lipopolysaccaride-induced mouse
macrophages. Journal of Biological Chemistry 22 14394–14398.
Montague C, Prins J, Sanders L, Zhang JL, Sewter CP, Digby J,
Byrne CD & O’Rahilly S 1998 Depot-related gene expression in
human subcutaneous and omental adipocytes. Diabetes 47
1384–1391.
Peake P, O’Grady S, PussellB&Charlesworth J 1997 Detection and
quantification of the control proteins of the alternative pathway of
the complement in 3T3-L1 adipocytes. European Journal of Clinical
Investigation 27 922–927.
Peraldi P, Xu M & Spiegelman B 1997 Thiazolidinediones block
tumor necrosis factor-á-induced inhibition of insulin signaling.
Journal of Clinical Investigation 100 1863–1869.
Prins J, Niesler C, Winterford C, Bright NA, Siddle K, O’Rahilly S,
Walker NI & Cameron DP 1997 Tumor necrosis factor-á induces
apoptosis of human adipose cells. Diabetes 46 1939–1944.
SaltielA&Olefsky J 1996 Thiazolidinediones in the treatment of
insulin resistance and type II diabetes. Diabetes 45 1661–1669.
StephensJ&Pekala P 1991 Transcriptional repression of the GLUT4
and C/EBP genes in 3T3-L1 adipocytes by tumor necrosis
factor-alpha. Journal of Biological Chemistry 266 21839–21845.
Sumida M, Shiosaka T, Nagar A, Isshikimasuda M, Okuda H &
Hamada M 1997 Suppresive eect of tumor necrosis factor-alpha
on adipogenic cell dierentiation and on gene expression of
hormone-sensitive lipase. Journal of Clinical Biochemistry and Nutrition
22 1–11.
WaageA&Bakke O 1988 Glucocorticoids suppress the production of
tumor necrosis factor by lipopolysaccaride-stimulated human
monocytes. Immunology 63 299–302.
Zhang B, Berger J, Hu E, Szalkowski D, White-Carrington S,
Spiegelman BM & Moller DE 1996 Negative regulation of
peroxisome proliferator-activated receptor-ã gene expression
contributes to the anti-adipogenic eects of tumor necrosis factor-á.
Molecular Endrocrinology 10 1457–1466.
Received 29 December 1998
Revised manuscript received 28 April 1999
Accepted 5 May 1999
C P SEWTER and others · Regulation of human adipose TNF release38
Journal of Endocrinology (1999) 163, 33–38
... -In patients with CKD, higher levels of visfatin are associated with lower GFR, and increased triglyceride and low-density lipoprotein; it is also proatherogenic in nature (89) TNF -TNF is a pro-inflammatory, proatherogenic compound, released by macrophage cells; it correlates positively with CKD severity and associates with higher cystatin C and urinary albumin/creatinine ratio (UACR) (90)(91)(92)(93). ...
... -The effects of TNF involve endothelial cell adhesion, ROS activation, increased albumin permeability, cytotoxicity, apoptosis and necrosis (93). ...
Article
Full-text available
Background: Obesity poses significant challenges to healthcare globally, particularly through its bi-directional relationship with co-morbid metabolic conditions such as type 2 diabetes and hypertension. There is also emerging evidence of an association between obesity and chronic kidney disease (CKD) which is less well characterized. Methods: A literature search of electronic libraries was conducted to identify and present a narrative review of the interplay between obesity and CKD. Findings: Obesity may predispose to CKD directly as it is linked to the histopathological finding of obesity-related glomerulopathy and indirectly through its widely recognized complications such as atherosclerosis, hypertension, and type 2 diabetes. The biochemical and endocrine products of adipose tissue contribute to pathophysiological processes such as inflammation, oxidative stress, endothelial dysfunction, and proteinuria. The prevention and management of obesity may prove critical in counteracting both the development and advancement of CKD. Moreover, measures of abdominal adiposity such as waist circumference, are generally associated with worse morbidity and mortality in individuals receiving maintenance hemodialysis. Conclusion: Obesity is a risk factor for the onset and progression of CKD and should be recognized as a potential target for a preventative public health approach to reduce CKD rates within the general population. Future research should focus on the use of glucagon-like peptide-1 receptor agonists and sodium-glucose cotransporter 2 inhibitors in patients with CKD and obesity due to their multi-faceted actions on major outcomes.
... Moreover, our data indicated that the absence of mGluR5 is associated to a higher IL-10/TNF ratio in the adipose tissue, which may indicate an anti-inflammatory tendency. Importantly, adipocytes produce several cytokines, but do not express mGluR5 53,54 . However, some studies demonstrated that activation of mGluR5 is important for the polarization and differentiation of macrophages and T lymphocytes 17,18 . ...
Article
Full-text available
Obesity represents a global health problem and is characterized by metabolic dysfunctions and a low-grade chronic inflammatory state, which can increase the risk of comorbidities, such as atherosclerosis, diabetes and insulin resistance. Here we tested the hypothesis that the genetic deletion of metabotropic glutamate receptor 5 (mGluR5) may rescue metabolic and inflammatory features present in BACHD mice, a mouse model of Huntington's disease (HD) with an obese phenotype. For that, we crossed BACHD and mGluR5 knockout mice (mGluR5 −/−) in order to obtain the following groups: Wild type (WT), mGluR5 −/− , BACHD and BACHD/mGluR5 −/− (double mutant mice). Our results showed that the double mutant mice present decreased body weight as compared to BACHD mice in all tested ages and reduced visceral adiposity as compared to BACHD at 6 months of age. Additionally, 12-month-old double mutant mice present increased adipose tissue levels of adiponectin, decreased leptin levels, and increased IL-10/TNF ratio as compared to BACHD mice. Taken together, our preliminary data propose that the absence of mGluR5 reduce weight gain and visceral adiposity in BACHD mice, along with a decrease in the inflammatory state in the visceral adipose tissue (VAT), which may indicate that mGluR5 may play a role in adiposity modulation.
... Despite not being dependent on insulin equal to glucose, fructose-induced DNL may decrease insulin sensitivity in virtue of increased insulin secretion, as the accumulation of TG in the liver leads to the raised secretion of inflammatory molecules such as CRP, interleukin-6, TNF-α, and decreased adiponectin. Herein, we analyzed some of these inflammatory biomarkers, providing evidence that a low free sugar diet can reduce the serum concentrations of TNF-α and hs-CRP, as well as NF-kb, thus inferring a clinical impact using the main molecules released by the liver and adipose tissue that orchestrate the low-grade inflammation [38][39][40]. Along these lines, we provide a declined hepatic contribution to systemic inflammation in patients with NAFLD, whose findings can be regarded as a favorable prognosis to avoid related cardiovascular disease in this population [41]. ...
Article
Full-text available
Background Although the role of a diet rich in fructose and saccharose in development of nonalcoholic fatty liver disease (NAFLD) is well known, the effects of a low free sugar diet in the management of the disease have not yet been investigated in adult patients with NAFLD. We aimed, therefore, to analyze the effects of a low-free sugar diet on NAFLD main features. Methods Participants with FibroScan-proven NAFLD were randomized to a 12-week dietary intervention (low free sugar diet or usual diet). The primary outcome was change in hepatic steatosis measurement between baseline and 12 weeks. The secondary outcomes included changes in anthropometric measurements, lipid profile, glycemic indices, liver enzymes, and inflammatory factors. Results Forty-three subjects completed the 12-week intervention. Low free sugar diet compared with the usual diet significantly decreased the concentrations of ALT (43.00 ± 27.54 to 27.95 ± 20.77 U/L), TG (172.86 ± 83.04 to 144.19 ± 65.55), TC (155.54 ± 37.55 to 139.86 ± 33.63 mg/dL), FBS (103.95 ± 15.42 to 91.00 ± 14.36 mg/dL), insulin (14.37 ± 5.79 to 8.92 ± 5.43 mU/L), HOMA-IR (3.81 ± 1.80 to 2.06 ± 1.29), hs-CRP (3.80 ± 1.09 to 2.88 ± 0.52 mg/L), TNF-α (4.60 ± 1.54 to 3.41 ± 0.69 pg/mL), NF-kb (3.89 ± 1.34 to 3.35 ± 1.33), as well as resulted in reduced fibrosis score and steatosis score, with increased QUICKI (P < 0.05). The differences in AST, GGT, HDL-C and LDL-C were not significant (P > 0.05). Conclusion Low free sugar diet in overweight/obese NAFLD patients may reduce hepatic steatosis and fibrosis while improving glycemic indices, decreasing the concentrations of biomarkers of inflammation, TG, and TC levels.
... An important limitation of our study is that we did not show the predominant cell population responsible for producing the cytokines modulated by the absence of mGluR5 in the adipose tissue of the double mutant mice. Adipocytes produce several cytokines, but do not express mGluR5 51,52 . However, some studies demonstrated that activation of mGluR5 is important for the polarization and differentiation of macrophages and T lymphocytes 9,10 . ...
Preprint
Full-text available
Obesity represents a serious global public health problem and is characterized by a low-grade chronic inflammatory state, which can increase the risk of comorbidities, such as: atherosclerosis, diabetes and insulin resistance. In this study, we investigated the effects of the genetic deletion of the metabotropic glutamate receptor 5 (mGluR5) on the modulation of obesity features in BACHD mice, a mouse model of Huntington´s disease. For that, we crossed BACHD and mGluR5 knockout mice (mGluR5 −/− ) in order to obtain the following groups: Wild type (WT), mGluR5 −/− , BACHD and BACHD/mGluR5 −/− (double mutant mice). Our results showed that the double mutant mice present decreased body weight as compared to BACHD mice at all tested ages and reduced visceral adiposity as compared to BACHD mice at 6 and 12 months of age. Additionally, 12-month-old double mutant mice present increased adipose tissue levels of adiponectin, decreased leptin levels, and increased IL-10/TNF ratio as compared to BACHD mice. Taken together, our data propose that the absence of mGluR5 reduce weight gain and visceral adiposity in BACHD mice, along with a decrease in the inflammatory state in the visceral adipose tissue (VAT), which may indicate that mGluR5 may play a role in adiposity modulation.
... (7,8) TNFα, a member of the tumor necrosis factor superfamily, is produced by different cell types such as monocytes, neutrophils, and adipocytes. (9,10) It plays important roles in cell proliferation, wound healing, and cancer progression. (8,11) TNFα is also found predominantly expressed in activated macrophages in inflammatory diseases such as rheumatoid arthritis. ...
Article
Full-text available
In mammals, osteoclasts differentiate from macrophages in the monocyte lineage. Although many factors driving osteoclast formation are known, the detailed processes underlying precursor recruitment, differentiation and interaction of macrophages with other cell types involved in bone remodeling are poorly understood. Using live imaging in a transgenic medaka osteoporosis model where ectopic osteoclasts are induced by Receptor activator of nuclear factor‐kappa Β ligand (Rankl) expression, we show that a subset of macrophages is recruited to bone matrix to physically interact with bone forming osteoblast progenitors. These macrophages subsequently differentiate into cathepsin K (ctsk)‐positive osteoclasts. One day later, other macrophages are recruited to clear dying osteoclasts from resorbed bone by phagocytosis. To better understand the molecular changes underlying these dynamic processes, we performed transcriptome profiling of activated macrophages upon Rankl induction. This revealed an upregulation of several bone‐related transcripts. Besides osteoclast markers, we unexpectedly also found expression of osteoblast‐promoting signals in activated macrophages, suggesting a possible non‐cell autonomous role in osteogenesis. Finally, we show that macrophage differentiation into osteoclasts is dependent on inflammatory signals. Medaka deficient for Tumor necrosis factor alpha (Tnfa) or treated with the Tnfa inhibitor pentoxifylline exhibited impaired macrophage recruitment and osteoclast differentiation. These results demonstrate the involvement of inflammatory signals and the dynamics of a distinct subset of macrophages during osteoclast formation. This article is protected by copyright. All rights reserved.
... In contrast to myeloid cells 5 , regulation of adipocyte inflammatory potential and its contribution to obesity-associated inflammation is poorly understood. Like myeloid cells, adipocytes produce proinflammatory cytokines 6 , express innate immune receptors (e.g. Toll-like receptor (TLR)) 7 and major histocompatibility complex (MHC) class I and class II molecules and present antigens 8,9 . ...
Article
Full-text available
White adipose tissue inflammation, in part via myeloid cell contribution, is central to obesity pathogenesis. Mechanisms regulating adipocyte inflammatory potential and consequent impact of such inflammation in disease pathogenesis remain poorly defined. We show that activation of the type I interferon (IFN)/IFNα receptor (IFNAR) axis amplifies adipocyte inflammatory vigor and uncovers dormant gene expression patterns resembling inflammatory myeloid cells. IFNβ-sensing promotes adipocyte glycolysis, while glycolysis inhibition impeded IFNβ-driven intra-adipocyte inflammation. Obesity-driven induction of the type I IFN axis and activation of adipocyte IFNAR signaling contributes to obesity-associated pathogenesis in mice. Notably, IFNβ effects are conserved in human adipocytes and detection of the type I IFN/IFNAR axis-associated signatures positively correlates with obesity-driven metabolic derangements in humans. Collectively, our findings reveal a capacity for the type I IFN/IFNAR axis to regulate unifying inflammatory features in both myeloid cells and adipocytes and hint at an underappreciated contribution of adipocyte inflammation in disease pathogenesis.
Article
Full-text available
Canine leishmaniosis is caused by infection with parasite Leishmania infantum, which are transmitted by sandflies Phlebotomus. Canine leishmaniosis is an endemic disease in the Mediterranean region. The immune response could vary between hosts and determines the severity of the disease and clinical features. The aim of this study was to analyze the serum levels of cytokines TNF-α, IFN-γ, IL-2, IL-6, and IL-8, which are related to the activation of Th1 or Th2 immune responses in dogs living in the L. infantum endemic region. Moreover, we intend to relate and correlate these levels with different factors, such as sex, age, diet, lifestyle, and breed. Epidemiological data and serum were recovered for seventy-eight dogs, and serum levels of cytokines described previously were analyzed by using the ELISA method. The results showed differences in serum levels of IFN-γ, IL-2, and IL-8 between breeds. The lifestyle also affected serum levels of IL-2. The main conclusion of this study is that Ibizan hounds and crossbred dogs have a serological profile of cytokines that seems to indicate certain protections against infection by L. infantum compared to boxer and purebred breeds.
Chapter
Recently, evidence derived from different studies has shown that metabolic routes determine the fate of immune cells and impact the immune response, giving rise to an enormous potentially opportunity to treat autoimmunity, cancer, infections or inflammatory diseases. This chapter explores the general metabolic signatures and metabolic drivers of T and B cells from ontogeny, following to activation, functional differentiation, effector functions and finally exhaustion to depicts how the metabolic switching is related with development and functionality of the adaptive immune cells.
Article
Full-text available
Obesity is associated with poorer executive functioning and reward sensitivity. Yet, we know very little about whether weight loss through diet and/or increased exercise engagement improves cognitive function. This study evaluated whether weight loss following a dietary and exercise intervention was associated with improved cognitive performance. We enrolled 125 middle-aged adults with overweight and obesity (98 female) into a 12-month behavioral weight loss intervention. Participants were assigned to one of three groups: energy-restricted diet alone, an energy-restricted diet plus 150 min of moderate intensity exercise per week or an energy restricted diet plus 250 min of exercise per week. All participants completed tests measuring executive functioning and/or reward sensitivity, including the Iowa Gambling Task (IGT). Following the intervention, weight significantly decreased in all groups. A MANCOVA controlling for age, sex and race revealed a significant multivariate effect of group on cognitive changes. Post-hoc ANCOVAs revealed a Group × Time interaction only on IGT reward sensitivity, such that the high exercise group improved their performance relative to the other two intervention groups. Post-hoc ANCOVAs also revealed a main effect of Time, independent of intervention group, on IGT net payoff score. Changes in weight were not associated with other changes in cognitive performance. Engaging in a high amount of exercise improved reward sensitivity above and beyond weight loss alone. This suggests that there is additional benefit to adding exercise into behavioral weight loss regimens on executive functioning, even without additional benefit to weight loss.
Article
Full-text available
Obesity is associated with significantly higher mortality rates, and excess adipose tissue is involved in respective pathologies. Here we established a human adipose tissue slice cultures (HATSC) model ex vivo. HATSC match the in vivo cell composition of human adipose tissue with, among others, mature adipocytes, mesenchymal stem cells as well as stroma tissue and immune cells. This is a new method, optimized for live imaging, to study adipose tissue and cell-based mechanisms of obesity in particular. HATSC survival was tested by means of conventional and immunofluorescence histological techniques, functional analyses and live imaging. Surgery-derived tissue was cut with a tissue chopper in 500 μm sections and transferred onto membranes building an air-liquid interface. HATSC were cultured in six-well plates filled with Dulbecco’s Modified Eagle’s Medium (DMEM), insulin, transferrin, and selenium, both with and without serum. After 0, 1, 7 and 14 days in vitro, slices were fixated and analyzed by morphology and Perilipin A for tissue viability. Immunofluorescent staining against IBA1, CD68 and Ki67 was performed to determine macrophage survival and proliferation. These experiments showed preservation of adipose tissue as well as survival and proliferation of monocytes and stroma tissue for at least 14 days in vitro even in the absence of serum. The physiological capabilities of adipocytes were functionally tested by insulin stimulation and measurement of Phospho-Akt on day 7 and 14 in vitro. Viability was further confirmed by live imaging using Calcein-AM (viable cells) and propidium iodide (apoptosis/necrosis). In conclusion, HATSC have been successfully established by preserving the monovacuolar form of adipocytes and surrounding macrophages and connective tissue. This model allows further analysis of mature human adipose tissue biology ex vivo.
Article
Full-text available
Tumor necrosis factor-alpha (TNF) has recently been shown to induce insulin resistance. We have examined the possible effect of TNF on the early events in insulin transmembrane signaling. Incubation of the insulin-sensitive rat hepatoma Fao cells with 5 nM TNF for 1 h led to a 65% decrease in insulin-induced tyrosine phosphorylation of both the insulin receptor beta-subunit and IPS-1, its major cytosolic substrate. TNF-induced impairment of tyrosine phosphorylation was maximal at 0.5 nM and was not accompanied by any reduction in insulin binding. Sixteen hours of TNF incubation led to further impairment in insulin-induced tyrosine phosphorylation of these proteins. Our findings suggest that TNF may exert its anti-insulin effect by interrupting the early insulin-stimulated tyrosine phosphorylation events, which are crucial to insulin transmembrane signature.
Article
Full-text available
Many cytokines exert their biological effect through members of the hemopoietin receptor family. Using degenerate oligonucleotides to the common WSXWS motif, we have cloned from human hemopoietic cell cDNA libraries various forms of the receptor that was recently shown to bind the obesity hormone, leptin. mRNAs encoding long and short forms of the human leptin receptor were found to be coexpressed in a range of human and murine hemopoietic organs, and a subset of cells from these tissues bound leptin at the cell surface. Ectopic expression in murine Ba/F3 and M1 cell lines revealed that the long, but not the short, form of the leptin receptor can signal proliferation and differentiation, respectively. In cultures of murine or human marrow cells, human leptin exhibited no capacity to stimulate cell survival or proliferation, but it enhanced cytokine production and phagocytosis of Leishmania parasites by murine peritoneal macrophages. Our data provide evidence that, in addition to its role in fat regulation, leptin may also be able to regulate aspects of hemopoiesis and macrophage function.
Article
Full-text available
Adipose tissue growth results from de novo adipocyte recruitment (hyperplasia) and increased size of preexisting adipocytes. Adipocyte hyperplasia accounts for the severalfold increase in adipose tissue mass that occurs throughout life, yet the mechanism of adipocyte hyperplasia is unknown. We studied the potential of macrophage colony-stimulating factor (MCSF) to mediate adipocyte hyperplasia because of the profound effects MCSF exerts on pluripotent cell recruitment and differentiation in other tissues. We found that MCSF mRNA and protein were expressed by human adipocytes and that adipocyte MCSF expression was upregulated in rapidly growing adipose tissue that encircled acutely inflamed bowel and in adipose tissue from humans gaining weight (4-7 kg) with overfeeding. Localized overexpression of adipocyte MCSF was then induced in rabbit subcutaneous adipose tissue in vivo using adenoviral-mediated gene transfer. Successful overexpression of MCSF was associated with 16-fold increases in adipose tissue growth compared with a control adenovirus expressing beta-galactosidase. This occurred in the absence of increased cell size and in the presence of increased nuclear staining for MIB-1, a marker of proliferation. We conclude that MCSF participates in adipocyte hyperplasia and the physiological regulation of adipose tissue growth.
Article
Full-text available
We have previously demonstrated the ability of tumor necrosis factor-alpha (TNF) to down-regulate the expression of GLUT4 (insulin-responsive glucose transporter) and C/EBP-alpha (CCAAT/enhancer-binding protein) (Stephens J. M., and Pekala, P. H. (1991) J. Biol. Chem. 266, 21839-21845). As C/EBP-alpha has been suggested to control GLUT4 expression, we have examined the time course for attenuation of transcription of these genes. Run-on transcription assays indicate a coordinate transcriptional repression of both GLUT4 and C/EBP-alpha genes (as well as the 422/aP2 gene, the adipocyte lipid-binding protein, whose expression has also been proposed to be controlled by C/EBP-alpha). Inhibition of transcription was observed within 1 h of TNF addition, with maximal suppression observed after 4 h. The inhibition was not blocked by cycloheximide. Okadaic acid treatment (1 h, 0.5 microM) also resulted in the coordinate transcriptional repression of the C/EBP-alpha, GLUT4, and 422/aP2 genes, consistent with involvement of a kinase-phosphatase system in the regulation of these genes. The decrease in C/EBP-alpha protein content was detectable 4 h after TNF addition and declined to 25% of controls within 24 h. A minor decrease in the protein content of GLUT4 was observed during the first 24 h of exposure to TNF; however, after 72 h of exposure GLUT4 protein was not detectable. The rapid coordinate transcriptional regulation of C/EBP-alpha, GLUT4, and 422/aP2 by TNF in the presence of cycloheximide suggests that the TNF-induced loss of GLUT4 protein may be mediated by a post-translational modification of an existing transcription factor. However, the rapid loss of C/EBP-alpha protein may be a contributing factor to further transcriptional suppression of the GLUT4 gene at the later time points. In addition to the transcriptional effect, we report that TNF-induced destabilization of these mRNAs contributes to decreased expression of all three genes.
Article
The thiazolidinediones are novel insulin sensitizers that serve as orally active antidiabetic agents, in rodents, nonhuman primates, and man. We have examined the effects of 4-week oral administration of three thiazolidinediones (AD-5075, BRL 49653, and CS-045) on plasma glucose and triglyceride concentrations in obese hyperglycemic db/db mice. All three agents lower plasma glucose and triglyceride concentrations. Normal levels of glucose are achieved after treatment with AD-5075 (> 1.7 mg/kg) or BRL 49653 (> or = 30 mg/kg), whereas CS-045 (100 or 300 mg/kg) produces only modest reductions in either parameter. Although the thiazolidinediones have demonstrated insulin-sensitizing activities both in vivo and in vitro, their primary molecular target has been unclear. We have compared the in vivo antidiabetic actions described above with the in vitro activities on peroxisomal proliferator-activated receptor-gamma (PPAR gamma). Hamster PPAR gamma 1 was transiently expressed in COS-1 cells to study the binding of [3H]AD-5075. The concentrations of compounds needed to displace radiolabeled AD-5075 from PPAR gamma correlate with their in vivo potency; the Ki values for displacement by cold AD-5075, BRL 49653, and CS-045 are 22, 68, and 1600 nM, respectively. To examine activation of the receptor, it was transiently cotransfected into COS-1 cells with a reporter plasmid containing two copies of a peroxisome proliferator response element. The EC50 values for activation are 2, 6, and 140 nM for AD-5075, BRL 49653, and CS-045, respectively. We have also analyzed limited proteolytic digests of in vitro translated hamster PPAR gamma. The thiazolidinediones produce a conformational change in PPAR gamma analogous to those produced by agonists of other nuclear hormone receptors. In the presence of saturating concentrations of either AD-5075 or BRL 49653, a receptor fragment of 27 kDa is protected from proteolysis by trypsin. These data support the conclusion that the antidiabetic actions of the thiazolidinediones are directly mediated through binding to PPAR gamma and the resulting active conformation of the receptor. Therefore, binding and transactivation assays using PPAR gamma should serve to identify other novel therapeutic agents with potential antidiabetic activities.
Article
Activators of peroxisome proliferator activated receptor (PPAR) regulate fatty acid metabolism and can induce adipocyte differentiation. We show here that the gamma subtype of PPAR is expressed at high levels in adipose tissue in contrast to a variety of other tissues, where little gene expression was noted. In addition, PPAR gamma is present at low levels in 3T3-L1 preadipocytes and is induced dramatically during adipocyte conversion using either normal differentiating conditions (fetal calf serum, dexamethasone, isobutyl-methylxanthine, and insulin) or the PPAR activator, WY-14,643. Thus PPAR gamma may be important for adipose cell development and function.
Article
Insulin resistance, characterized by reduced responsiveness to normal circulating concentrations of insulin, is a common feature of almost all patients with type II diabetes. The presumed central roles of both peripheral and hepatic insulin resistance suggest that the enhancement of insulin action might be an effective pharmacological approach to diabetes. Thiazolidinediones are a new class of orally active drugs that are designed to enhance the actions of insulin. These agents reduce insulin resistance by increasing insulin-dependent glucose disposal and reducing hepatic glucose output. Clinical studies in patients with type II diabetes, as well as other syndromes characterized by insulin resistance, have demonstrated that thiazolidinediones may represent a safe and effective new treatment. Although the precise mechanism of action of these drugs remains unknown, transcriptional changes are observed in tissue culture cells that produce enhanced insulin action. This regulation of gene expression appears to be mediated by the interactions of thiazolidinediones with a family of nuclear receptors known as the peroxisome proliferator-activated receptors (PPARs). The further elucidation of the molecular actions of these drugs may reveal much about the underlying mechanisms of insulin resistance.
Article
Addition of human recombinant tumor necrosis factor-α (hrTNF) to confluent 3T3-L1 preadipocytes in the presence of differentiation inducers depressed both cellular differentiation into adipocytes and gene expression of hormone-sensitive lipase (HSL) without diminishing cellular viability. When hrTNF was added to 3T3-L1 cells that had differentiated into adipocytes, the cellular level of HSL mRNA was dose dependently reduced over the period of 3-12h with a concomitant reduction in HSL activity. The reducing effect of hrTNF on the HSL mRNA level was reversible. Cycloheximide (0.1mg/ml) suppressed the reducing effect of hrTNF, indicating that de novo protein synthesis is required for the hrTNF action. Actinomycin D (1μg/ml) gradually reduced the HSL mRNA level in the adipocytes over 12h without affecting the ratio of HSL mRNA to β-actin mRNA. The HSL mRNA level in primary cultures of Leydig cells was not affected by 1nM hrTNF. In addition, interleukin-1β(100nM) did not inhibit HSL gene expression when it was added to 3T3-L1 cells, regardless of their differentiation stage.
Article
The purpose of this study was to determine whether human adipocytes from different depots of obese subjects produce interleukin-6 (IL-6) and whether IL-6 release is regulated by glucocorticoids. Fragments of omental and abdominal sc adipose tissue released immunodetectable IL-6 into the medium during acute incubations. Omental adipose tissue released 2-3 times more IL-6 than did sc adipose tissue. Isolated adipocytes prepared from these tissues also released IL-6 (omental > sc), but this accounted for only 10% of the total tissue release. Culture of adipose tissue fragments for 7 days with the glucocorticoid dexamethasone markedly suppressed IL-6 production. These data show for the first time that substantial quantities of IL-6 (up to 75 ng/mL) accumulate in the medium during incubations of both adipocytes and adipose tissue. Although little is known about the effects of IL-6 on adipose tissue, one action is a down-regulation of adipose tissue lipoprotein lipase. The regulated production of this multifunctional cytokine may modulate regional adipose tissue metabolism and may contribute to the recently reported correlation between serum IL-6 and the level of obesity.