You are on page 1of 14

Clinical Science (2005) 109, 243–256 (Printed in Great Britain) doi:10.

1042/CS20050078 243

R E V I E W

Role of resistin in obesity, insulin


resistance and Type II diabetes

Christine M. KUSMINSKI, Philip G. MCTERNAN and Sudhesh KUMAR


Diabetes and Metabolism Research Laboratory, Clinical Sciences Research Institute, UHCW Trust, Clifford Bridge Road,
Walsgrave, Coventry CV2 2DX, U.K.

A B S T R A C T

Resistin is a member of a class of cysteine-rich proteins collectively termed resistin-like mol-


ecules. Resistin has been implicated in the pathogenesis of obesity-mediated insulin resistance and
T2DM (Type II diabetes mellitus), at least in rodent models. In addition, resistin also appears to
be a pro-inflammatory cytokine. Taken together, resistin, like many other adipocytokines, may
possess a dual role in contributing to disease risk. However, to date there has been considerable
controversy surrounding this 12.5 kDa polypeptide in understanding its physiological relevance in
both human and rodent systems. Furthermore, this has led some to question whether resistin
represents an important pathogenic factor in the aetiology of T2DM and cardiovascular disease.
Although researchers still remain divided as to the role of resistin, this review will place available
data on resistin in the context of our current knowledge of the pathogenesis of obesity-mediated
diabetes, and discuss key controversies and developments.

INTRODUCTION factors leading to diabetes, as such, the expression and


functional properties of adipocytokines and their effects
Adipose tissue is known to produce a vast array of adipo- on metabolism have been the subject of intense research.
cyte-derived factors, known as adipocytokines. Under Indeed, studies on adipocytokines and their potential
‘normal’ physiological conditions adipocytokines may effects in human obesity and T2DM have implicated them
play an influential role in energy homoeostasis, tri- in the pathogenesis of the metabolic syndrome. Such
acylglycerol (triglyceride) storage and mobilization of fat, factors include TNF-α (tumour necrosis factor-α), IL
with increased adiposity, specifically central adiposity. (interleukin)-6, angiotensinogen, leptin, PAI-1 (plas-
These processes can be substantially dysregulated [1]. minogen activator inhibitor-1) and resistin (Table 1).
Furthermore, it seems apparent that the pathogenesis of This review will address our current understanding of
T2DM (Type II diabetes mellitus) is mediated through the pathophysiological role of resistin, and evaluate re-
the concurrent progression of insulin resistance and sub- sistin as a pathogenic factor implicated in metabolic and
clinical inflammation, although the molecular mechan- inflammatory mechanisms leading to diabetes. In ad-
isms for this are less understood. It is, however, apparent dition, we will highlight the continuing complexity of the
that obesity represents one of the foremost contributory biology of resistin and its role in the progression of

Key words: adipocytokine, inflammation, insulin resistance, obesity, resistin, Type II diabetes.
Abbreviations: BMI, body mass index; CAC, coronary artery calcification; CRP, C-reactive protein; CVD, cardiovascular
disease; ET-1, endothelin-1; HMM, high-molecular-mass; ICAM-1, intercellular cell-adhesion molecule-1; IL, interleukin; LMM,
low-molecular-mass; LPS, lipopolysaccharide; NF-κB, nuclear factor κB; PAI-1, plasminogen activator inhibitor-1; PBMC,
peripheral blood mononuclear cell; PPAR-γ , peroxisome-proliferator-activated receptor-γ ; PPRE, PPAR-γ -response element;
PTX3, pentraxin 3; RELM, resistin-like molecule; RSG, rosiglitazone; SNP, single nucleotide polymorphism; SOCS, suppressor of
cytokine signalling; sTNF-R2, soluble TNF-receptor 2; T2DM, Type II diabetes mellitus; TNF-α, tumour necrosis factor-α; TZD,
thiozolidinedione; VCAM-1, vascular cell adhesion molecule-1; WAT, white adipose tissue.
Correspondence: Professor Sudhesh Kumar (email sudhesh.kumar@warwick.ac.uk).


C 2005 The Biochemical Society
244 C. M. Kusminski, P. G. McTernan and S. Kumar

Table 1 Summary of factors implicated in the pathogenesis of obesity-related T2DM


AGT, angiotensinogen; AT, adipose tissue; BAT, brown AT; Om, omental; Sc, subcutaneous; TGF-β, transforming growth factor-β.

Molecule Function/effect Normal distribution Effect of obesity References

Leptin Satiety and appetite; signals to Secreted by WAT, BAT, skeletal ↑In human obesity and correlates [38,112–114]
the brain to regulate energy muscle, stomach and placenta; with BMI, and ↓after fasting or
homoeostasis and body weight. 2.5 times more leptin in Sc AT than weight loss.
Om AT; more leptin in adipocytes
than pre-adipocytes.
TNF-α Reduces insulin secretion and Predominant in adipocytes in Correlates with BMI, ↑ in human [112,115–120]
insulin signalling in AT, pancreatic WAT; 1.67 times more TNF-α obesity; 2-fold increase in obese
cells, liver and muscle. in Sc AT compared with Om AT compared with lean subjects; ↑
in a rodent model of obesity; ↓
adipose differentiation.
IL-6 Affects glucose and lipid 35 % of the basal supply is derived ↑ In morbidly obese patients, [38,121–125]
metabolism; improves insulin from WAT; produced by macrophages, ↓ after weight loss.
sensitivity and glucose tolerance; fibroblasts, endothelial cells and
has anti-inflammatory properties. skeletal muscle cells.
PAI-1 Potent inhibitor of the fibrinolytic Correlation with abdominal adiposity; ↑ In humans leads to ↑ thromboembolic [126]
pathway. pattern of adipose tissue distribution: complications.
AGT Precursor of angiotensin II; mRNA greater in Om AT than Sc AT ↑ In humans. [127,128]
regulator of blood supply and
induces differentiation of
pre-adipocytes.
Adiponectin Regulator of energy Exclusively secreted by adipocytes. ↓ In mouse models of obesity and [76,78,103,129–131]
homoeostasis: enhances mRNA and protein greater in Sc AT insulin resistance (ob/ob and db/db );
insulin sensitivity and glucose than Om AT. ↓ in human obesity and T2DM;
uptake; has anti-inflammatory ↑ after weight loss.
properties.
TGF-β Varied role in proliferation, Multifunctional, produced by ↑ob/ob obese and db/db mice; ↑ in [132]
differentiation, apoptosis and variety of cells; inhibitor of pre-adipocyte cell proliferation,
development. differentiation. as with ↑ in TNF-α.
PPAR-γ Regulates adipose cell Sc adipocytes = Om adipocytes ↑ In Sc WAT 2 times greater than in [133,134]
differentiation. (BMI < 28 kg/m2 ) Om WAT (BMI < 30 kg/m2 )
Ghrelin Implicated in the coordination Secreted by endocrine cells in the ↑ In human obesity and T2DM. [38,135–140]
of energy balance and weight gastrointestinal tract. ↑ Circulating levels after chronic
regulation. weight loss in humans.
Adiponutrin Possible contribution to energy Non-secreted adipocyte protein. ↑ mRNA (50-fold) in obese fa/fa rats; [141–143]
homoeostasis, vesicular targeting Expressed exclusively in inguinal and ↓ fasting mRNA levels; human gene
and protein transport. epididymal WAT and in interscapular expression may be regulated by
BAT. changes in energy balance.

disease and the potential interplay between several mech- regarding resistin, one question remains pertinent to all
anisms associated in the pathogenesis of T2DM. Since others, what is the precise physiological function of res-
the discovery of resistin in 2001, there have been over 260 istin? Although this is not necessarily an easy question to
publications that have informed our present understand- answer and this review may only highlight some aspects
ing of its function. This has paradoxically created a mael- of resistin in disease, it is clear that the physiological role
strom of controversy and ambiguity surrounding the of resistin is more complex than originally envisioned.
functional significance of resistin. Indeed, researchers Despite an incomplete current understanding of the
outside the field of resistin over the last few years have role of resistin, this review will assemble and discuss
been unclear as to whether this molecule may deserve the existing literature on resistin to evaluate the cur-
much attention as an important pathogenic factor in the rent metabolic, inflammatory and cardiovascular effects
progression of obesity-mediated diabetes. Nevertheless, of this protein, with the intent of providing an insight as
although several questions remain unanswered to date to where the role of resistin may lie.


C 2005 The Biochemical Society
Role of resistin in obesity, insulin resistance and Type II diabetes 245

mass) complex, which is unable to form intertrimer


disulphide bonds [6]. This implied that regulated pro-
cessing through disulphide cleavage is required to initiate
bioactivity of the LMM form, and suggested further a
potential target site for receptor interaction [6].

TISSUE DISTRIBUTION OF RESISTIN AND


OTHER RELM (RESISTIN-LIKE MOLECULE)
FAMILY MEMBERS
Resistin belongs to a family of cysteine-rich proteins,
termed RELMs, which include RELM-α/FIZZ 1,
RELM-β/FIZZ 2 and the recently discovered RELM-γ ,
with each member having an unique differential tissue
distribution [2,7,8]. Resistin expression was first de-
scribed in adipose tissue [3], with circulating levels
detected in rodents and humans [9–12]. Our present
knowledge of the expression of resistin and RELM-α, -β
and -γ in human and rodent tissues is shown in Table 2.
Figure 1 Ribbon diagram representations of resistin Although the expression of resistin in mice was
(A) Monomeric structure of resistin, whereas (B) shows its hexameric form com- originally restricted to adipocytes [3], the principle origin
prising of two disulphide-linked trimers. (C) Highly exposed interchain disulphide of human resistin has remained somewhat contentious.
linkages present in the hexameric form of resistin. Reprinted (abstracted/excerpted) Unlike the mouse gene, the human homologue of re-
with permission from Patel, S. D., Rajala, M. W., Rossetti, L., Scherer, P. E. and sistin was sparsely detectable in human adipocytes [13];
Shapiro, L., Disulfide-dependent multimeric assembly of resistin family hormones, this was confounded further by confusion over the pro-
Science 304, 1154–1158.  c (2004) AAAS (www.sciencemag.org). posed sites of resistin production [13–15]. These studies
led to many of the current perceptions that resistin was
THE DISCOVERY OF RESISTIN an inconsequential factor in the progression of obesity-
related T2DM, thereby contrasting rodent data. Con-
Resistin, a putative adipocyte-derived signalling poly- versely, McTernan et al. [16] detected resistin in adipose
peptide, was originally identified by three independent tissue, thus describing resistin as a potential pathogenic
groups using a variety of techniques [2–4]. Initial studies factor increased in central adiposity. The discrepancy
showed that resistin was up-regulated in rodent models between studies may have partially related to method-
of obesity and insulin resistance and down-regulated ology of detection or quantification of resistin. How-
by an insulin-sensitizer, RSG (rosiglitazone) [3]; however, ever, to date, although a difference in resistin mRNA
immunoneutralization of resistin reduced hypergly- levels between adipocytes and macrophages is apparent
caemia and improved insulin sensitivity [3]. These [14,17,18], studies have not yet identified whether this
observations not only brought resistin to much scientific difference is observed at the protein level. Determining
attention, but characterized it as a potential aetiological the relative contribution of the adipocyte in obesity with
link between obesity and diabetes, with a clear functional regards to circulating resistin levels would also prove
role as a pathogenic factor contributing to insulin beneficial.
resistance. Additionally, this revealed possibilities of the
mechanistic action for TZDs (thiozolidinediones) and
their subsequent therapeutic applications.
HOMOLOGY OF HUMAN AND RODENT
RESISTIN

THE STRUCTURE OF RESISTIN Although four genes for the family of RELMs have been
identified in mice, only two homologues have been id-
Studies determined the resistin gene, referred to as Retn, entified in the complete human genome [7,8]. Re-
encoded a 114-amino acid polypeptide [3], secreted as a cently, an alternatively spliced variant of the Retn gene
disulphide-linked homodimer [5]. Recent X-ray crystal- was identified in humans (resistin 2) [19], and
lographic studies of resistin [6] have determined its com- another was identified in rats, termed ‘S-Resistin’ (short
plex hexameric structure (Figure 1). Resistin was shown resistin) [20].
to circulate in two distinct assembly states; the more pre- The human Retn gene is located on chromosome 19
dominant HMM (high-molecular-mass) hexamer and in a region syntenic to the mouse Retn gene on chromo-
the substantially more bioactive LMM (low-molecular- some 8. In relation to this, one group posed the view that


C 2005 The Biochemical Society
246 C. M. Kusminski, P. G. McTernan and S. Kumar

Table 2 Distribution of mRNA and protein expression of resistin and RELM-β in rodents and humans, and RELM-α and
RELM-γ in rodents
R, mRNA expression; P, protein expression.

Resistin RELM-β RELM-α RELM-γ

Rodent Human Rodent Human Rodent Rodent

Tissue R P R P R P R P R P R P References

WAT × × × × × × [3,7,8,16,18,19,23,25,86,144,145]
Pre-adipocytes × × × [14,25]
Adipocytes × × × × [3,15,25,29]
Mononuclear blood cells × × [8,13,15,17,68,86,88]
Hypothalamus × × [19,145,146]
Pituitary gland × [145,146]
Adrenal gland × × [19,72]
Spleen × × × [8,19,147]
Skeletal muscle × × × [19,72,148]
Pancreas × × × [19,149]
Placenta × × [7,19,150]
Gastrointestinal tract × × × × × × × [7,8,19,29,72]
Lung × × × × × [2,7,8,19,29]

explain such diverse expression and regulatory patterns


of this protein, or simply suggest resistin may not be
evolutionary well conserved across species.

THE METABOLIC ROLE FOR RESISTIN:


AN ONGOING DEBATE
Resistin and obesity
Initial observations
Since the initial investigation of resistin in numerous
rodent models of obesity and insulin resistance [3,23,24],
ongoing experimental data has generated further in-
consistency. Human studies have highlighted increased
Figure 2 Amino acid sequence identities of rodent, porcine resistin expression in adipose tissue [15], particularly
and bovine resistin, other RELM family members and the abdominal depots [16,25]; furthermore, positive correl-
recently identified human alternatively spliced variant of ations between serum resistin and body fat content have
resistin, resistin 2 also been reported [26]. On the contrary, several studies
Percentage sequence identities were obtained from the EMBL-EBI (European Bioin- have failed to demonstrate such correlations in rodents,
formatics Institute) server (http://www.ebi.ac.uk). The different colours represent with groups also reporting either reduced [27–30]
the different RELM family members. b, bovine; h, human, p, porcine; r, rodent. or no alteration [31,32] of resistin levels in various
models of obesity. Although it is difficult to address such
orthology existed between human and mouse genes [21], diverse findings using similar, and in some instances the
whereas another questioned the relevance of resistin, due same, rodent models, inconsistencies may depend upon
to the absence of a true homologue for the murine Retn methodological differences [18]. The following sections
gene in humans [22]. Contrasting opinions regarding the will review cases for and against the role for resistin as an
orthology may relate to how genomic, transcribed and important pathogenic factor in obesity, insulin resistance
translated sequence identities are viewed. However, it and T2DM in light of recent studies.
is evident that human and mouse resistin have diverse
genomic organizations [22]. Moreover, at the protein Recent observations supporting a role for resistin in obesity
level, human resistin is only 55 % identical with its Studies by Lee et al. [33] showed that various murine
murine counterpart (Figure 2), and this could, in part, models of obesity had higher circulating resistin levels


C 2005 The Biochemical Society
Role of resistin in obesity, insulin resistance and Type II diabetes 247

compared with their lean counterparts. These obser- ever, it has been suggested recently that resistin mRNA
vations coincided with rodent studies by Rajala and co- expression does not necessarily correlate with protein ex-
workers [34], showing circulating resistin levels were pression [34]. Possible explanations for such diverse
significantly elevated and concordant with increasing observations include differences in post-transcrip-
levels of insulin, glucose and lipids; thus substantiating tional and post-translational modifications, consequently
the initial evidence that addressed the aetiology of resistin affecting secretory rates of resistin. Increased serum levels
with increasing adiposity [3]. Recently, Asensio et al. may enhance transcript degradation rates via negative
[35] determined that high-fat-fed mice had induced feedback mechanisms, or the initiation and recruitment
adipocyte differentiation, denoted by fatty acid binding of inhibitors of translation. The secreted form of resistin
protein (AP-2) gene expression, a surrogate marker of is considered to have paracrine properties, and this
differentiation, which positively correlated with resistin may imply the majority of regulation occurs at the
gene expression. Subsequently, in view of this and previ- protein level. Similarly, Rajala and co-workers [34]
ous studies [4,36], it was suggested that elevated resistin have suggested that binding of serum cofactors to the
expression was a result of adipocyte differentiation [35]. resistin protein may prolong its half-life, thereby reduc-
Moreover, the increase in adipocyte number may have ing clearance; this hypothesis is supported by studies
caused a rise in local resistin production, inhibiting insulin showing protein interactions and tertiary alterations can
action on glucose uptake in adipose tissue and, thus, pre- influence adipocytokines, such as leptin and TNF-α
venting further adipocyte differentiation [35]. Therefore, [45–47]. Indeed, these studies demonstrate the limit-
at least in rodents, a regulatory feedback mechanism ations of attempting to derive all necessary inform-
for resistin in adipogenesis may occur, acting as an adi- ation regarding resistin and obesity from gene expression
pose sensor for nutritional status. In accordance with studies.
these observations, Kim et al. [37] generated transgenic Further recent human studies have shown no cor-
mice overexpressing a dominant inhibitory form of re- relation of serum [48] or plasma levels of resistin with
sistin which functioned to block the inhibition of resistin- any markers of adiposity [49]. Heilbronn et al. [50] re-
mediated adipocyte differentiation. These transgenic mice ported no relationship between resistin serum levels and
developed obesity, possibly owing to enhanced adipocyte percentage body fat, visceral adiposity and BMI. How-
differentiation and adipocyte hypertrophy, as indicated ever, the authors [50] suggested that the lack of correlation
by increased circulating levels of adiponectin and leptin of serum resistin and increased adiposity was partly due
[37]. to the confounding variable of age, as non-obese sub-
Recent investigations of human resistin in relation to jects were significantly younger than obese subjects [50].
obesity have shown higher serum resistin levels in obese Another study showed similar levels of resistin expressed
subjects compared with lean subjects [38–40], which in gluteal femoral and subcutaneous abdominal depots in
positively correlated with the changes in BMI (body mass non-diabetic subjects [51]. However, no indication of the
index) and visceral fat area [11,40–42]. The implication number of ‘gluteal’ subjects compared with ‘abdominal’
that resistin is important in human adipose tissue has subjects was given. Moreover, the study used a high pro-
been corroborated by studies showing increased protein portion of male subjects and, as previous studies had
expression with obesity [40], as well as protein secretion shown resistin to exhibit sexual dimorphism, with women
from isolated adipocytes [43]. These recent observations having approx. 20 % higher levels than men [42,48,49],
are concomitant with initial studies that showed increased this may explain these findings.
serum resistin levels [26] and gene expression levels in Although a high degree of discrepancy has emerged
abdominal depots [16,25] in states of increased adiposity. from publications regarding the association of resistin
A further study has shown a significant reduction in with obesity in the context of mRNA and serum levels,
circulating resistin levels following moderate weight loss it is worth highlighting the importance of developing
[12] and post-gastric bypass [38]. Collectively, these ob- highly accurate methods of determining serum resistin
servations suggest resistin could indirectly be subjected concentrations. Methodological limitations may result in
to nutritional regulation in humans. variations among serum concentrations, mRNA and pro-
tein levels, or may simply indicate that resistin does not
Observations that argue against a major role for resistin play a significant role in the pathophysiology of obesity-
in obesity mediated insulin resistance. However, with reference to
Contrary with the studies suggesting a role for resistin in the use of commercially available ELISAs, both rodent
obesity, Maebuchi et al. [44] have reported resistin was and human ELISAs have potential to cross-react with
undetectable in serum of obese mice, with the same study circulating RELMs. To date, not all studies using resistin
indicating reductions of resistin mRNA and protein ELISAs have assessed RELM cross-reactivity prior to
expression in obesity. Others have reported no associ- analysis; therefore it may be that different ELISAs
ation of resistin expression with increased adiposity, de- used may provide varying serum concentrations [52–55].
spite observing elevated circulating levels [33–35]. How- Similarly, assessment of serum resistin in rodents has


C 2005 The Biochemical Society
248 C. M. Kusminski, P. G. McTernan and S. Kumar

proved contentious [33,34]. Furthermore, due to recent HOMA-IR (homoeostasis model assessment for insulin
advances in the understanding of the tertiary and quatern- resistance), waist circumference, BMI or total cholesterol
ary structure of resistin [6], further studies are required [54,55]. Consequently, these studies suggest resistin is
to establish whether the complex distribution of the unlikely to play a critical endocrine role in insulin re-
individual structural forms of resistin affect the validity sistance or energy homoeostasis in humans. Nevertheless,
of the currently available human and rodent assays. a paracrine or autocrine manner of resistin to moderately
affect metabolism cannot be ruled out.
Resistin, insulin resistance and T2DM
It is currently established that central obesity is a
Effect of resistin on glucose homoeostasis
Recently, it has been reported that transgenic mice over-
contributing factor to the pathogenesis of insulin resist-
expressing resistin exhibited impaired insulin-mediated
ance and consequently to T2DM. Although it is apparent
glucose transport [60]. This altered glucose metabolism
that inconsistencies remain in the data for a role of resistin
appeared to occur without affecting insulin receptor sig-
in obesity, there is a growing body of evidence suggesting
nalling, therefore acting by reducing the intrinsic activity
a role for resistin in the aetiology of insulin resistance and
of cell-surface glucose transporters [60]. Lazar and co-
T2DM.
workers [61] have recently shown resistin induced the
expression of SOCS (suppressor of cytokine signalling)-
Regulation of resistin in models of insulin resistance and 3, a known inhibitor of insulin signalling. Moreover,
glucose intolerance the loss of SOCS function was shown to impair resistin
Early rodent studies determined that reduced serum res- from antagonizing insulin action in adipocytes [61]. This
istin levels in mice were associated with decreased adipos- suggested that the insulin-independent action of resistin
ity and improved insulin sensitivity [56]. Rajala et al. on adipocytes could partly be mediated by SOCS-3,
[34] recently demonstrated that circulating resistin levels which could have an impact on normal glucose homo-
were significantly elevated and positively concordant eostasis [61].
with rising levels of insulin, glucose and lipids in Lepob/ob Rajala et al. [9] have shown that infusion of either
mice. This study also highlighted the potential interplay resistin or RELM-β into rats decreased insulin sensitivity,
between resistin and leptin, with leptin suppressing primarily at the site of the liver. This worsening of glucose
resistin mRNA and protein levels, concomitant with the homoeostasis was shown to be entirely attributable to the
reduction in glucose and insulin [34]. Furthermore, severely impaired insulin-mediated suppression of hep-
Asensio et al. [35] highlighted that leptin administration atic gluconeogenesis, rather than peripheral insulin re-
in ob/ob mice improved insulin sensitivity, which was sistance [9]. The study consequently suggested that fat-
affiliated with a decrease in resistin gene expression. Col- and gut-derived resistin and RELM-β may have clear and
lectively, these studies suggest leptin may exert insulin- rapid effects on stimulating the rate of hepatic glucose
resistance-ameliorating effects via counter-regulatory production, as opposed to increasing glucose uptake or
interactions and potentially suppressive mechanisms influencing peripheral insulin sensitivity [9]. In this
towards resistin. In contrast, Lee and co-workers [33] re- regard, the secretion of RELM-β into the portal venous
ported that neither transcriptional regulation of resistin circulation appears to link the intestinal epithelium to the
nor circulating resistin levels correlated with serum liver, enhancing changes in hepatic intermediate meta-
insulin or glucose levels. Subsequent studies have bolism as a result [9]. Furthermore, this supported the
denoted resistin expression was either suppressed [57] or notion of the existence of a feedback mechanism between
unchanged [31] in rodent models of insulin resistance. adipose tissue and insulin-target organs, such as the liver.
Furthermore, although resistin mRNA levels were These findings have been reinforced by studies show-
increased in insulin-resistant rats, no apparent change in ing that the ablation of the resistin gene in mice lowering
insulin sensitivity was observed [24]. fasting glucose levels through reducing hepatic glucose
In evaluating resistin and its association with insulin production without significantly altering whole-body
sensitivity in humans, several studies have identified posi- glucose disposal [62]. This study showed that improve-
tive correlations between resistin levels and insulin re- ment in glucose homoeostasis was partly mediated via
sistance in vivo [49] and in vitro [51]. Additionally, increased activation of hepatic AMPK (AMP-activated
serum resistin levels were increased by approx. 20 % in protein kinase) with reduced gene expression of the glu-
T2DM subjects [43], such findings have been re-affirmed coneogenic enzymes G6Pase (glucose 6-phosphatase) and
by Fujinami et al. [52]. In contrast, other studies have PEPCK (phosphoenolypyruvate carboxykinase) [62].
reported no associations between serum resistin levels and Conversely, the authors [62] also showed that resistin
markers of insulin resistance in T2DM patients [48,53,58] treatment in these knockout mice enhanced hepatic
or insulin-resistant patients [59]. Moreover, serum and production by increasing glucose levels by approx. 25 %.
plasma resistin levels were either reduced or increased Furthermore, Rangwala et al. [63] documented that mice
in T2DM patients with no significant correlation with with chronic hyper-resistinaemia exhibited higher blood


C 2005 The Biochemical Society
Role of resistin in obesity, insulin resistance and Type II diabetes 249

glucose levels and impaired glucose tolerance; this was dated, although it has been suggested that recruitment of
associated with increased hepatic glucose production, co-repressors of transcription may play a role [69]. RSG
partially due to increased hepatic expression of glu- has also been shown to have anti-inflammatory effects
coneogenic enzymes. Nonetheless, impairment of normal in human macrophages [70] by reducing inflammatory
glucose homoeostasis caused by chronic hyper-resistin- cytokine production, which may consequently affect re-
aemia may require more severe measures to counter- sistin production [71]. Collectively, these human studies
regulate these effects. indicate that suppression of resistin expression may
In the human context, early studies showed contradict- contribute to the insulin-sensitizing and glucose-lower-
ory findings for correlations between resistin and glucose ing actions of the TZDs. Furthermore, the potential anti-
disposal, with recent studies still providing inconsistent inflammatory effects of TZDs on adipocytokine me-
results. Studies in Pima Indians have reported serum re- diation may be of equal importance in the prevention
sistin levels were not associated with fasting glucose and of T2DM.
insulin levels, although they were proportional to the de-
gree of adiposity [41]. Additionally, one study indicated Hormone and cytokine modulators of resistin expression
serum resistin levels were inversely correlated with glu- Studies investigating metabolic hormones and cytokines
cose disposal rates [50], whereas others indicate a modest that are associated with insulin resistance have looked for
effect of resistin on glucose uptake in vitro [43]. Col- their relationship with resistin. Such studies have pro-
lectively, resistin transgenic and gene-deletion studies in duced data suggesting an interplay between hormones,
rodents have provided evidence that resistin may have cytokines and resistin, as shown in Table 3. Although the
a predominant physiological role in the liver by contri- reported regulation of resistin by these factors appears
buting to the regulation of fasting blood glucose levels. intriguing, no underlying mechanistic principles are cur-
Consequently this may have important implications in rently apparent. Therefore the physiological relevance of
humans, but further studies regarding this are clearly most of these factors with respect to resistin remain to be
required. determined.

Gender effects on resistin regulation


Regulation of resistin by insulin sensitizers In WAT, it has been shown that resistin mRNA ex-
Several studies have reported the down-regulation of pression is higher in male than female rats [72,73], in
resistin expression following RSG treatment in WAT contrast with a similar study also undertaken in mice [74].
(white adipose tissue) of db/db mice, diabetic fatty rats In human subjects, Yannakoulia and co-workers [42]
[3,64] and 3T3-L1 adipocytes [36]. Furthermore, resistin reported that resistin concentrations were significantly
expression is down-regulated not only by RSG, but higher in females compared with males, which was also
also by darglitazone [36] and troglitazone [65]. Con- confirmed by two other studies suggesting a significant
versely, subsequent rodent studies report that PPAR-γ gender difference [48,49]. However, a subsequent study
(peroxisome-proliferator-activated receptor-γ ) agonists examining both age and gender in relation to serum
[27], pioglitazone and troglitazone [28], and metformin resistin indicated a lack of association in healthy subjects,
[66] increase resistin gene and protein expression. How- as well as patients with Type I diabetes and T2DM [39]. As
ever, human studies have shown that treatment of mono- a result, the significance of gender on the degree of resistin
cytes with PPAR-γ agonists failed to have any effect expression in rodents and humans remains unclear, per-
on resistin levels [15]. These initial observations suggest haps complicated by the issue of specificity of resistin
that down-regulation of resistin is not crucial for the ELISAs.
antidiabetic effect of TZDs in all model systems. How-
ever, recent human studies have shed favourable light on Adiponectin, insulin resistance and T2DM
the modulation of resistin by TZDs. Pioglitazone treat- Although resistin has been implicated in the pathogenesis
ment suppressed plasma resistin concentrations in T2DM of insulin resistance and T2DM, it is noteworthy to
patients, which positively correlated with decreased mention another novel adipocytokine proposed to have
hepatic fat content and improved insulin sensitivity [67]. a role in the aetiology of human T2DM. Adiponectin
Furthermore, RSG reduced resistin secretion from expression and plasma levels, in contrast with resistin and
human adipocytes [43] and resistin expression in human other adipocytokines whose levels are increased in states
macrophages [17,68]. To understand whether the down- of obesity, are reduced in insulin resistance and obesity
regulation of resistin expression by RSG occurred [75,76]. Additionally, it has been reported that gastric
through a direct PPAR-γ -mediated transcriptional mech- bypass in morbidly obese patients has resulted in elevated
anism, Patel et al. [17] identified five putative PPREs levels of plasma adiponectin [77]. Interestingly, low
(PPAR-γ -response elements) in the resistin gene. One serum levels of adiponectin have been shown to indepen-
such response element, PPRE2, was shown to bind dently predict future risk of developing T2DM [78]. With
PPAR-γ [17]. However, how PPAR-γ exerts its sup- regards to insulin sensitizers, adiponectin synthesis and
pressive actions on resistin expression remains to be eluci- secretion are up-regulated by TZDs in vitro and in vivo


C 2005 The Biochemical Society
250 C. M. Kusminski, P. G. McTernan and S. Kumar

Table 3 Effects of hormones and cytokines on the level of resistin expression and secretion

Hormone/cytokine Effect on resistin References

Pituitary hormones Growth hormone (somatotrophin; 1 mg · kg−1 · day−1 ) ↑ resistin gene expression (720–950 %) in WAT [65,151,152]
of spontaneous dwarf rats and has moderate inhibitory effects on resistin transcript and protein
(30–50 %) levels in 3T3-L1 adipocytes; ↑ gene expression levels in response to hyperprolactinaemia
in mice;↓ mRNA and protein expression (30–50 %) in 3T3-L1 adipocytes.
Steroid hormones Dexamethasone ↑ mRNA and protein levels (2.5- to 3.5–fold) in 3T3-L1 adipocytes and approx. [36,65]
70 % in mouse WAT.
Sex hormones ↑ In mice with elevated androgen levels; ↑ by hyperprolactinaemia and testosterone; administration [73,152–154]
of dehydroepiandrosterone ↑ gene expression in WAT of male Wistar rats; oestrogen ↓ adipose
gene expression in ovariectomized rats and in isolated rat adipocytes in vitro ; testosterone ↑ adipose
tissue mRNA levels in male rats.
Thyroid hormone Severely ↓ expression in hyperthyroid rats; ↓ serum levels in subjects with hyperthyroidism. [73,155]
Adrenaline ↓ Transcript and protein levels (30–50 %) in 3T3-L1 adipocytes. [65]
Neuropeptide Y Intracerebroventricular administration of neuropeptide Y ↑ gene expression in mice WAT. [156]
β 3 -Adrenoreceptors The β 3 -agonist isoproterenol ↓ gene expression levels in vitro by 20 % in 3T3-L1 adipocytes, which [157]
is reversible with the β 3 -antagonist propranolol.
ET-1 ET-1 (100 nM) ↓ basal secretion by 59 % in 3T3-L1 adipocytes. [158]
Insulin ↓ Gene expression (approx. 50 %) in 3T3-L1 adipocytes; ↑ secretion from 3T3-L1 adipocytes; [4,27,34,36,43,65,158]
↑ mRNA synthesis (23-fold) in streptozotocin-diabetic mice or Zucker diabetic fatty rats; gene
expression and protein concentration ↑ in fasted mice; ↑resistin protein secretion in a
concentration-dependent manner in human subcutaneous adipocytes.

[79–81] and suppressed by insulin-resistance-inducing different doses and time points of LPS treatment or the
hormones [82,83]. The precise function of adiponectin animal models used. Assessing the effect of pro-inflam-
as a therapeutic target, improving glucose metabolism by matory cytokines on resistin expression showed that
increasing insulin sensitivity in humans, is currently of treatment with TNF-α down-regulates resistin, suggest-
great interest. ing TNF-α as a negative regulator of resistin [29]. How-
ever, in one case, no apparent effect was observed with
Potential role of resistin in inflammation treatment of IL-6 [87].
Recent studies have highlighted the importance of Although initial rodent studies have produced dis-
inflammation in linking obesity and the occurrence of the crepancies as to whether pro-inflammatory cytokines
metabolic syndrome, suggesting that the metabolic syn- may regulate resistin, several recent human studies have
drome is a low-grade systemic inflammatory condition supported the concept of inflammatory cytokine me-
[84]. Although this emerging concept of inflammation has diation of resistin. One study has shown resistin to be
been recognized in the context of increasing adiposity, its significantly correlated with IL-6 and ICAM-1 (inter-
molecular basis and physiological significance in terms of cellular cell-adhesion molecule-1) in obese patients
the pathogenesis of T2DM is yet to be fully elucidated. with obstructive sleep apnoea syndrome [88], whereas
Thus adipose tissue may act as a site for mediation of another reported LPS, IL-1, TNF-α and IL-6 strongly
this chronic inflammation through the action of known stimulated resistin expression in human [PBMCs (peri-
pro-inflammatory adipocytokines such as leptin, TNF-α, pheral blood mononuclear cells)] [38,43,58]. Correlations
IL-6 and, most recently proposed, resistin [85]. Resistin, between resistin expression and secretion and other in-
although postulated to contribute to insulin resistance, flammatory markers, including IL-6, leptin, sTNF-R2
may also contribute to inflammatory responses. (soluble TNF-receptor 2) and CRP (C-reactive pro-
Early investigations into the role of resistin as an in- tein), have also been reported in patients with severe
flammatory factor demonstrated that LPS (lipopolysac- inflammatory disease, obesity and T2DM [68]. Recently,
charide) up-regulated resistin expression in rat WAT, Lehrke et al. [68] demonstrated that endotoxaemia caused
3T3-L1 adipocytes and human monocytes [86]. However, a dramatic increase in resistin in primary human macro-
contradicting these findings, a down-regulation of resistin phages, and this was attenuated by aspirin and RSG. In
expression in response to LPS in adipose tissue of FVB this situation, RSG was suggested to possess dual insulin-
mice has been identified, with no effect transcriptionally sensitizing and anti-inflammatory properties. Addition-
or translationally in 3T3-L1 adipocytes reported [29]. ally, LPS induced TNF-α, which preceded the increase
Such a discrepancy between studies may have arisen from in resistin, and therefore, in this study, the authors


C 2005 The Biochemical Society
Role of resistin in obesity, insulin resistance and Type II diabetes 251

hypothesized that TNF-α was responsible for the later production in 3T3-L1 adipocytes, and antagonized an
induction of resistin [89]. Thus cytokine induction of endotoxin-induced increase of TNF-α expression [97].
resistin may contribute to insulin resistance in endo- These studies suggest a regulatory role for adiponectin
toxaemia, obesity and inflammation. in inflammation within adipose tissue, possibly acting via
Obesity is associated with adipose tissue infiltration the regulation of NF-κB. Hypoadiponectinaemia may
by macrophages, thus contributing to resistin expression, contribute to the pathogenesis of obesity-related insulin
and this may propagate further recruitment of more resistance, inflammation and T2DM, and therefore fur-
macrophages potentially interacting with adipocytes. ther studies should elucidate mechanisms of action.
Adipose tissue may therefore represent a site of an innate
immune response in humans [90]. Alternatively, it is
hypothesized that macrophage recruitment may instead Relationship of resistin with CVD
arise from the trans-differentiation of pre-adipocytes into (cardiovascular disease)
macrophage-like cells [68,88], highlighting the close inter- Adipocyte-derived hormones not only function promin-
relationship between adipocyte and macrophage lineages. ently in the pathogenesis of T2DM, but may also serve
This would be consistent with the emerging concept as important vasoactive factors directly affecting endo-
that adipocytes and macrophages share the property of thelial function and vascular homoeostasis [98]. Therefore
secreting pro-inflammatory cytokines that regulate meta- these factors may represent an important point of con-
bolic function. To date, resistin gene expression levels vergence in linking insulin resistance to an increased
have been reported to be higher in human PBMCs risk of atherosclerotic vascular disease. Although the
[25]. However, comparative studies have not shown the precise molecular mechanisms responsible for this con-
relative resistin protein expression in macrophages vergence remain nebulous, resistin may have a role in its
compared with adipocytes; this could alter our perception pathogenesis. A recent study by Reilly and co-workers
of the contribution to resistin made by the adipocyte. [98] suggested resistin as a metabolic link between in-
Furthermore, studies have highlighted increased resistin flammation and atherosclerosis. This group reported that,
protein expression in human abdominal fat, irrespective in non-diabetic and diabetic subjects, plasma resistin
of the presence of mononuclear blood cells [91]. At the levels were associated with metabolic and inflammatory
very least, it seems apparent that the findings on the pat- markers, including sTNF-R2, IL-6 and LpPLA2 (lipo-
tern of resistin mRNA expression suggest that adipocytes protein-associated phospholipase A2 ) [98]. Furthermore,
contribute modestly to hyper-resistinaemia. Although, resistin levels correlated with CAC (coronary artery cal-
combined with the significant biomass that adipose tissue cification), a quantitative measure of coronary athero-
can contribute to whole-body weight, particularly in sclerosis [98]. Notably, in metabolic syndrome subjects,
obese individuals, there is little doubt that the systemic resistin levels further predicted CAC, whereas CRP levels
contribution of adipose tissue is significant [92]. It has did not [99]. Such results imply resistin may be an
been suggested that obesity and T2DM are associated independent marker of atherosclerosis in humans.
with chronic inflammation and activation of innate Several studies have examined resistin as a cardiovas-
immune pathways [93,94]. In obesity, where pro-inflam- cular risk factor and a potential contributor to endothelial
matory cytokine levels are known to be elevated, it may dysregulation and atherosclerotic lesion formation [99].
be plausible that resistin could function as a signalling Such studies have shown resistin stimulated factors,
component of the innate immune response to circuitously including ET-1 (endothelin-1), VCAM-1 (vascular cell
induce the production of pro-inflammatory cytokines, adhesion molecule-1) and MCP-1 (monocyte chemoat-
such as TNF-α and IL-6, within macrophages and adipo- tractant protein-1), thus contributing to CVD [100]. Ad-
cytes. In turn, such immune dysregulation could con- ditional confirmation of the potential effects of resistin on
tribute to the development of a pro-inflammatory milieu the vasculature have arisen through studies examining hu-
associated with insulin resistance. Overall, it is apparent man aortic endothelial cells. One study reported that re-
that the relationships between obesity, inflammation sistin up-regulated the expression of adhesion molecules,
and resistin expression are complex. Further studies are including VCAM-1 and ICAM-1, as well as long PTX3
clearly required to explain the role of the adipocyte as a (pentraxin 3), a marker of inflammation [100–102].
site of an immune response, potentially contributing to Therefore, as adhesion molecules may represent the initial
coupling of immunity and metabolic dysregulation. stages of atherosclerotic lesions and PTX3 expression is
In addition to resistin, adiponectin may possess anti- increased in atherosclerotic lesions, resistin may exert a
inflammatory properties. Recent studies have shown that pro-inflammatory effect on vascular endothelial cells,
adiponectin suppressed cytokine production in macro- promoting the onset of atherosclerosis [100]. In contrast
phages [95] and disrupted the activation of NF-κB with resistin, adiponectin, known to enhance insulin sen-
(nuclear factor κB) in an aortic endothelial cell model sitivity and reduce atherosclerotic plaques, suppressed a
[96]. Ajuwon and Spurlock [96] reported that adiponectin resistin-mediated rise in VCAM-1 and ICAM-1 [103].
attenuated LPS-induced NF-κB activation and IL-6 These observations coincide well with a study showing


C 2005 The Biochemical Society
252 C. M. Kusminski, P. G. McTernan and S. Kumar

that adiponectin suppressed the TNF-α-stimulated ex- CONCLUSIONS


pression of E-selectin, VCAM-1 and ICAM-1 in human
endothelial cells [104–106]. This suggests further that It is apparent that the pathogenesis of T2DM and CVD
adiponectin may be vasoprotective and negatively modu- is complex and multifactorial in which several adipo-
late the atherogenic processes. These findings raise the cytokines have been implicated. Over the last 4 years our
possibility that resistin, in conjunction with other adipo- understanding of the role of resistin in this process has led
cytokines, may influence the degree of inflammation some to question its relative importance in man. Recent
in the vasculature via direct modulation of endothelial studies have shed more light on differences between
function. However, further studies are required to fully rodents and humans, indicating the diversity of resistin
elucidate the relationship of resistin and adiponectin with action in these species. Further studies are required to
inflammatory-related CVD. establish the relevance of resistin to human diabetes, in
particular its effects on the central nervous system and
β-cell function that have not been characterized. It is
HUMAN GENETIC STUDIES OF RESISTIN
also clear that further work is required to understand the
Several SNPs (single nucleotide polymorphisms) have basis for formation of different higher and lower mol-
been identified in the Retn gene, but only few have minor ecular-mass forms of resistin in the circulation and their
allele frequencies over 5 % and are associated with dis- effects on function. Clearly, identifying a receptor for
ease risk [104–106]. Therefore further confusion ensues resistin would represent a major advance and could lead
when reviewing genetic studies examining associations to a clearer understanding of its function. However, our
between resistin and disease. In a study of non- growing knowledge of the role of pro-inflammatory mol-
diabetic French Canadians in Quebec [104], two Retn ecules generated within adipose tissue and their link to
5 -flanking SNPs (− 537 and − 420) were associated diabetes means that interventions that reduce the pro-
with increased BMI. Furthermore, a resistin genotype duction of pro-inflammatory cytokines, including re-
at nucleotide + 299 (IVS2 + 181G → A) and obesity was sistin, may prove to have therapeutic potential.
a significant determinant of T2DM risk among Type II
diabetic Caucasians in Boston (MA, U.S.A.) [107].
ACKNOWLEDGMENTS
Additionally, the − 420C → G SNP (− 180 relative to
putative transcription start site) was associated with We thank Diabetes UK and the Dunhill Medical Trust for
higher resistin mRNA levels in abdominal fat of obese providing funding of a PhD Studentship and a Research
subjects [51]. Conversely, Mattevi et al. [105] showed Fellowship respectively.
an association between the − 420C → G polymorphism
with lower BMI in non-diabetic individuals from a
Brazilian population of European descent, although, REFERENCES
among non-diabetic Caucasians in Sicily and Gargano
(Italy), an ATG triplet repeat in the 3 -untranslated region 1 Mohamed-Ali, V., Pinkney, J. H. and Coppack, S. W.
(1998) Adipose tissue as an endocrine and paracrine
of the resistin gene was associated with a decreased organ. Int. J. Obes. Relat. Metab. Disord. 22, 1145–1158
risk of insulin resistance [108]. 2 Holcomb, I. N., Kabakoff, R. C., Chan, B. et al. (2000)
FIZZ1, a novel cysteine-rich secreted protein associated
Genetic analysis of resistin using a Japanese population with pulmonary inflammation, defines a new gene family.
demonstrated that a − 420G/G genotype was associated EMBO J. 19, 4046–4055
with T2DM and could accelerate the onset of disease 3 Steppan, C. M., Bailey, S. T., Bhat, S. et al. (2001) The
hormone resistin links obesity to diabetes.
by 4.9 years [106]; moreover, the genotype itself was a Nature (London) 409, 307–312
primary variant determining T2DM susceptibility [106]. 4 Kim, K. H., Lee, K., Moon, Y. S. and Sul, H. S. (2001)
A cysteine-rich adipose tissue-specific secretory factor
Consistent with these findings, elevated levels of serum inhibits adipocyte differentiation. J. Biol. Chem. 276,
resistin were reported in T2DM subjects carrying the 11252–11256
5 Banerjee, R. R. and Lazar, M. A. (2001) Dimerization of
− 420G/G genotype [109]. In contrast, studies in a resistin and resistin-like molecules is determined by a
Japanese obese population reported the − 638G → A, single cysteine. J. Biol. Chem. 276, 25970–25973
− 420C → G, and − 358G → A SNPs, which although 6 Patel, S. D., Rajala, M. W., Rossetti, L., Scherer, P. E. and
Shapiro, L. (2004) Disulfide-dependent multimeric
associated with serum resistin, did not confer any associ- assembly of resistin family hormones. Science 304,
ation with obesity or insulin resistance [110,111]. These 1154–1158
7 Steppan, C. M., Brown, E. J., Wright, C. M. et al. (2001)
genetic studies highlight the discrepancies in resistin A family of tissue-specific resistin-like molecules.
SNP analysis examining the association with obesity- Proc. Natl. Acad. Sci. U.S.A. 98, 502–506
related insulin resistance; these may partly be explained 8 Gerstmayer, B., Kusters, D., Gebel, S. et al. (2003)
Identification of RELM-γ , a novel resistin-like molecule
by different genetic backgrounds or environmental with a distinct expression pattern. Genomics 81, 588–595
conditions of the populations studied. Further studies 9 Rajala, M. W., Obici, S., Scherer, P. E. and Rossetti, L.
(2003) Adipose-derived resistin and gut-derived
on the physiology of resistin and genetic implications for resistin-like molecule-β selectively impair insulin action
the development of this disease are therefore crucial. on glucose production. J. Clin. Invest. 111, 225–230


C 2005 The Biochemical Society
Role of resistin in obesity, insulin resistance and Type II diabetes 253

10 Stejskal, D., Proskova, J., Adamovska, S., Jurakova, R. 30 Steppan, C. M. and Lazar, M. A. (2002) Resistin and
and Bartek, J. (2002) Preliminary experience with resistin obesity-associated insulin resistance. Trends Endocrinol.
assessment in common population. Biomed. Pap. Med. Metab. 13, 18–23
Fac. Palacky. Univ., Olomouc. 146, 47–49 31 Makimura, H., Mizuno, T. M., Bergen, H. and Mobbs,
11 Azuma, K., Katsukawa, F., Oguchi, S. et al. (2003) C. V. (2002) Adiponectin is stimulated by adrenalectomy
Correlation between serum resistin level and adiposity in in ob/ob mice and is highly correlated with resistin
obese individuals. Obes. Res. 11, 997–1001 mRNA. Am. J. Physiol. Endocrinol. Metab. 283,
12 Valsamakis, G., McTernan, P. G., Chetty, R. et al. (2004) E1266–E1271
Modest weight loss and reduction in waist circumference 32 Le Lay, S., Boucher, J., Rey, A. et al. (2001) Decreased
after medical treatment are associated with favourable resistin expression in mice with different sensitivities to a
changes in serum adipocytokines. Metab. Clin. Exp. 53, high-fat diet. Biochem. Biophys. Res. Commun. 289,
430–434 564–567
13 Nagaev, I. and Smith, U. (2001) Insulin resistance and 33 Lee, J. H., Bullen, Jr, J. W., Stoyneva, V. L. and
type 2 diabetes are not related to resistin expression in Mantzoros, C. S. (2005) Circulating resistin in lean, obese
human fat cells or skeletal muscle. Biochem. Biophys. and insulin-resistant mouse models: lack of association
Res. Commun. 285, 561–564 with insulinemia and glycemia. Am. J. Physiol.
14 Janke, J., Engeli, S., Gorzelniak, K., Luft, F. C. and Endocrinol. Metab. 288, E625–E632
Sharma, A. M. (2002) Resistin gene expression in human 34 Rajala, M. W., Qi, Y., Patel, H. R. et al. (2004) Regulation
adipocytes is not related to insulin resistance. Obes. Res. of resistin expression and circulating levels in obesity,
10, 1–5 diabetes, and fasting. Diabetes 53, 1671–1679
15 Savage, D. B., Sewter, C. P., Klenk, E. S. et al. (2001) 35 Asensio, C., Cettour-Rose, P., Theander-Carrillo, C.,
Resistin/Fizz3 expression in relation to obesity and Rohner-Jeanrenaud, F. and Muzzin, P. (2004) Changes in
peroxisome proliferator-activated receptor-γ action in glycemia by leptin administration or high-fat feeding in
humans. Diabetes 50, 2199–2202 rodent models of obesity/type 2 diabetes suggest a link
16 McTernan, C. L., McTernan, P. G., Harte, A. L., Levick, between resistin expression and control of glucose
P. L., Barnett, A. H. and Kumar, S. (2002) Resistin, central homeostasis. Endocrinology 145, 2206–2213
obesity, and type 2 diabetes. Lancet 359, 46–47 36 Haugen, F., Jorgensen, A., Drevon, C. A. and Trayhurn, P.
17 Patel, L., Buckels, A. C., Kinghorn, I. J. et al. (2003) (2001) Inhibition by insulin of resistin gene expression in
Resistin is expressed in human macrophages and directly 3T3-L1 adipocytes. FEBS Lett. 507, 105–108
regulated by PPARγ activators. Biochem. Biophys. 37 Kim, K. H., Zhao, L., Moon, Y., Kang, C. and Sul, H. S.
Res. Commun. 300, 472–476 (2004) Dominant inhibitory adipocyte-specific secretory
18 Fain, J. N., Cheema, P. S., Bahouth, S. W. and factor (ADSF)/resistin enhances adipogenesis and
Lloyd Hiler, M. (2003) Resistin release by human adipose improves insulin sensitivity. Proc. Natl. Acad. Sci. U.S.A.
tissue explants in primary culture. Biochem. Biophys. 101, 6780–6785
Res. Commun. 300, 674–678 38 Vendrell, J., Broch, M., Vilarrasa, N. et al. (2004) Resistin,
19 Nohira, T., Nagao, K., Kameyama, K. et al. (2004) adiponectin, ghrelin, leptin, and proinflammatory
Identification of an alternative splicing transcript for the cytokines: relationships in obesity. Obes. Res. 12, 962–971
39 Schaffler, A., Buchler, C., Muller-Ladner, U. et al. (2004)
resistin gene and distribution of its mRNA in human
Identification of variables influencing resistin serum levels
tissue. Eur. J. Endocrinol. 151, 151–154
in patients with type 1 and type 2 diabetes mellitus.
20 Del Arco, A., Peralta, S., Carrascosa, J. M., Ros, M.,
Horm. Metab. Res. 36, 702–707
Andres, A. and Arribas, C. (2003) Alternative splicing
40 Degawa-Yamauchi, M., Bovenkerk, J. E., Juliar, B. E. et al.
generates a novel non-secretable resistin isoform in Wistar (2003) Serum resistin (FIZZ3) protein is increased in
rats. FEBS Lett. 555, 243–249 obese humans. J. Clin. Endocrinol. Metab. 88, 5452–5455
21 Steppan, C. M. and Lazar, M. A. (2004) The current 41 Vozarova de Courten, B., Degawa-Yamauchi, M.,
biology of resistin. J. Intern. Med. 255, 439–447 Considine, R. V., Tataranni, P. A. and Volarova de
22 Ghosh, S., Singh, A. K., Aruna, B., Mukhopadhyay, S. and Courten, B. (2004) High serum resistin is associated with
Ehtesham, N. Z. (2003) The genomic organization of an increase in adiposity but not a worsening of insulin
mouse resistin reveals major differences from the human resistance in Pima Indians. Diabetes 53, 1279–1284
resistin: functional implications. Gene 305, 27–34 42 Yannakoulia, M., Yiannakouris, N., Bluher, S., Matalas,
23 Gabriely, I., Ma, X. H., Yang, X. M. et al. (2002) Removal A. L., Klimis-Zacas, D. and Mantzoros, C. S. (2003) Body
of visceral fat prevents insulin resistance and glucose fat mass and macronutrient intake in relation to
intolerance of aging: an adipokine-mediated process? circulating soluble leptin receptor, free leptin index,
Diabetes 51, 2951–2958 adiponectin, and resistin concentrations in healthy
24 Levy, J. R., Davenport, B., Clore, J. N. and Stevens, W. humans. J. Clin. Endocrinol. Metab. 88, 1730–1736
(2002) Lipid metabolism and resistin gene expression in 43 McTernan, P. G., Fisher, F. M., Valsamakis, G.
insulin-resistant Fischer 344 rats. Am. J. Physiol. et al. (2003) Resistin and type 2 diabetes: regulation of
Endocrinol. Metab. 282, E626–E633 resistin expression by insulin and rosiglitazone and the
25 McTernan, P. G., McTernan, C. L., Chetty, R. et al. (2002) effects of recombinant resistin on lipid and glucose
Increased resistin gene and protein expression in human metabolism in human differentiated adipocytes. J. Clin.
abdominal adipose tissue. J. Clin. Endocrinol. Metab. 87, Endocrinol. Metab. 88, 6098–6106
2407 44 Maebuchi, M., Machidori, M., Urade, R., Ogawa, T. and
26 Zhang, J., Qin, Y., Zheng, X. et al. (2002) The relationship Moriyama, T. (2003) Low resistin levels in adipose tissues
between human serum resistin level and body fat content, and serum in high-fat fed mice and genetically obese mice:
plasma glucose as well as blood pressure. Zhonghua Yi development of an ELISA system for quantification of
Xue Za Zhi 82, 1609–1612 resistin. Arch. Biochem. Biophys. 416, 164–170
27 Way, J. M., Gorgun, C. Z., Tong, Q. et al. (2001) 45 Huang, L., Wang, Z. and Li, C. (2001) Modulation of
Adipose tissue resistin expression is severely suppressed circulating leptin levels by its soluble receptor.
in obesity and stimulated by peroxisome J. Biol. Chem. 276, 6343–6349
proliferator-activated receptor γ agonists. J. Biol. Chem. 46 Lahlou, N., Clement, K., Carel, J. C. et al. (2000) Soluble
276, 25651–25653 leptin receptor in serum of subjects with complete
28 Fukui, Y. and Motojima, K. (2002) Expression of resistin resistance to leptin: relation to fat mass. Diabetes 49,
in the adipose tissue is modulated by various factors 1347–1352
including peroxisome proliferator-activated receptor α. 47 Winkler, G., Kiss, S., Keszthelyi, L. et al. (2003)
Diabetes Obes. Metab. 4, 342–345 Expression of tumor necrosis factor (TNF)-α protein in
29 Rajala, M. W., Lin, Y., Ranalletta, M. et al. (2002) Cell the subcutaneous and visceral adipose tissue in correlation
type-specific expression and coregulation of murine with adipocyte cell volume, serum TNF-α, soluble serum
resistin and resistin-like molecule-α in adipose tissue. TNF-receptor-2 concentrations and C-peptide level.
Mol. Endocrinol. 16, 1920–1930 Eur. J. Endocrinol. 149, 129–135


C 2005 The Biochemical Society
254 C. M. Kusminski, P. G. McTernan and S. Kumar

48 Lee, J. H., Chan, J. L., Yiannakouris, N. et al. (2003) 67 Bajaj, M., Suraamornkul, S., Hardies, L. J.,
Circulating resistin levels are not associated with obesity Pratipanawatr, T. and DeFronzo, R. A. (2004) Plasma
or insulin resistance in humans and are not regulated by resistin concentration, hepatic fat content, and hepatic and
fasting or leptin administration: cross-sectional and peripheral insulin resistance in pioglitazone-treated
interventional studies in normal, insulin-resistant, and type II diabetic patients. Int. J. Obes. Relat. Metab.
diabetic subjects. J. Clin. Endocrinol. Metab. 88, Disord. 28, 783–789
4848–4856 68 Lehrke, M., Reilly, M. P., Millington, S. C., Iqbal, N.,
49 Silha, J. V., Krsek, M., Skrha, J. V., Sucharda, P., Nyomba, Rader, D. J. and Lazar, M. A. (2004) An inflammatory
B. L. and Murphy, L. J. (2003) Plasma resistin, adiponectin cascade leading to hyperresistinemia in humans. PLoS
and leptin levels in lean and obese subjects: correlations Med. 1, e45
with insulin resistance. Eur. J. Endocrinol. 149, 331–335 69 Aranda, A. and Pascual, A. (2001) Nuclear hormone
50 Heilbronn, L. K., Rood, J., Janderova, L. et al. (2004) receptors and gene expression. Physiol. Rev. 81,
Relationship between serum resistin concentrations and 1269–1304
insulin resistance in nonobese, obese, and obese diabetic 70 Ricote, M., Li, A. C., Willson, T. M., Kelly, C. J. and
subjects. J. Clin. Endocrinol. Metab. 89, 1844–1848 Glass, C. K. (1998) The peroxisome proliferator-activated
51 Smith, S. R., Bai, F., Charbonneau, C., Janderova, L. and receptor-γ is a negative regulator of macrophage
Argyropoulos, G. (2003) A promoter genotype and activation. Nature (London) 391, 79–82
oxidative stress potentially link resistin to human insulin 71 Hong, G., Davis, B., Khatoon, N., Baker, S. F. and
resistance. Diabetes 52, 1611–1618 Brown, J. (2003) PPAR γ -dependent anti-inflammatory
52 Fujinami, A., Obayashi, H., Ohta, K. et al. (2004) action of rosiglitazone in human monocytes: suppression
Enzyme-linked immunosorbent assay for circulating of TNF α secretion is not mediated by PTEN regulation.
human resistin: resistin concentrations in normal subjects Biochem. Biophys. Res. Commun. 303, 782–787
and patients with type 2 diabetes. Clin. Chim. Acta. 339, 72 Nogueiras, R., Gallego, R., Gualillo, O. et al. (2003)
57–63 Resistin is expressed in different rat tissues and is
53 Pfutzner, A., Langenfeld, M., Kunt, T., Lobig, M. and regulated in a tissue- and gender-specific manner.
Forst, T. (2003) Evaluation of human resistin assays with FEBS Lett. 548, 21–27
serum from patients with type 2 diabetes and different 73 Nogueiras, R., Gualillo, O., Caminos, J. E., Casanueva,
degrees of insulin resistance. Clin. Lab. 49, 571–576 F. F. and Dieguez, C. (2003) Regulation of resistin by
54 Youn, B. S., Yu, K. Y., Park, H. J. et al. (2004) Plasma gonadal, thyroid hormone, and nutritional status.
resistin concentrations measured by enzyme-linked Obes. Res. 11, 408–414
immunosorbent assay using a newly developed 74 Gui, Y., Silha, J. V. and Murphy, L. J. (2004) Sexual
monoclonal antibody are elevated in individuals with type dimorphism and regulation of resistin, adiponectin, and
2 diabetes mellitus. J. Clin. Endocrinol. Metab. 89, leptin expression in the mouse. Obes. Res. 12, 1481–1491
150–156 75 Hu, E., Liang, P. and Spiegelman, B. M. (1996) AdipoQ is
55 Yang, J., Li, M., Wu, C. Y., Wang, H., Xu, Q. S. and Deng, a novel adipose-specific gene dysregulated in obesity.
J. Y. (2003) Reduced resistin levels in patients with type 2 J. Biol. Chem. 271, 10697–10703
diabetes mellitus. Zhonghua Yi Xue Za Zhi 83, 1471–1474 76 Weyer, C., Funahashi, T., Tanaka, S. et al. (2001)
56 Hirosumi, J., Tuncman, G., Chang, L. et al. (2002) A Hypoadiponectinemia in obesity and type 2 diabetes:
central role for JNK in obesity and insulin resistance. close association with insulin resistance and
Nature (London) 420, 333–336 hyperinsulinemia. J. Clin. Endocrinol. Metab. 86,
57 Juan, C. C., Au, L. C., Fang, V. S. et al. (2001) Suppressed 1930–1935
gene expression of adipocyte resistin in an insulin- 77 Yang, W. S., Lee, W. J., Funahashi, T. et al. (2001) Weight
resistant rat model probably by elevated free fatty acids. reduction increases plasma levels of an adipose-derived
Biochem. Biophys. Res. Commun. 289, 1328–1333 anti-inflammatory protein, adiponectin. J. Clin.
58 Stejskal, D., Adamovska, S., Bartek, J., Jurakova, R. and Endocrinol Metab. 86, 3815–3819
Proskova, J. (2003) Resistin concentrations in persons 78 Spranger, J., Kroke, A., Mohlig, M. et al. (2003)
with type 2 diabetes mellitus and in individuals with acute Adiponectin and protection against type 2 diabetes
inflammatory disease. Biomed. Pap. Med. Fac. Palacky. mellitus. Lancet 361, 226–228
Univ., Olomouc. 147, 63–69 79 Yu, J. G., Javorschi, S., Hevener, A. L. et al. (2002) The
59 Hegele, R. A., Kraw, M. E., Ban, M. R., Miskie, B. A., effect of thiazolidinediones on plasma adiponectin levels
Huff, M. W. and Cao, H. (2003) Elevated serum in normal, obese, and type 2 diabetic subjects. Diabetes
C-reactive protein and free fatty acids among nondiabetic 51, 2968–2974
carriers of missense mutations in the gene encoding lamin 80 Motoshima, H., Wu, X., Sinha, M. K. et al. (2002)
A/C (LMNA) with partial lipodystrophy. Arterioscler., Differential regulation of adiponectin secretion from
Thromb., Vasc. Biol. 23, 111–116 cultured human omental and subcutaneous adipocytes:
60 Moon, B., Kwan, J. J., Duddy, N., Sweeney, G. and effects of insulin and rosiglitazone. J. Clin.
Begum, N. (2003) Resistin inhibits glucose uptake in L6 Endocrinol. Metab. 87, 5662–5667
cells independently of changes in insulin signaling and 81 Yang, W. S., Jeng, C. Y., Wu, T. J. et al. (2002) Synthetic
GLUT4 translocation. Am. J. Physiol. Endocrinol. peroxisome proliferator-activated receptor-γ agonist,
Metab. 285, E106–E115 rosiglitazone, increases plasma levels of adiponectin in
61 Steppan, C. M., Wang, J., Whiteman, E. L., Birnbaum, type 2 diabetic patients. Diabetes Care 25, 376–380
M. J. and Lazar, M. A. (2005) Activation of SOCS-3 by 82 Delporte, M. L., Funahashi, T., Takahashi, M.,
resistin. Mol. Cell. Biol. 25, 1569–1575 Matsuzawa, Y. and Brichard, S. M. (2002) Pre- and
62 Banerjee, R. R., Rangwala, S. M., Shapiro, J. S. et al. (2004) post-translational negative effect of β-adrenoceptor
Regulation of fasted blood glucose by resistin. Science agonists on adiponectin secretion: in vitro and in vivo
303, 1195–1198 studies. Biochem. J. 367, 677–685
63 Rangwala, S. M., Rich, A. S., Rhoades, B. et al. (2004) 83 Fasshauer, M., Kralisch, S., Klier, M. et al. (2003)
Abnormal glucose homeostasis due to chronic Adiponectin gene expression and secretion is inhibited by
hyperresistinemia. Diabetes. 53, 1937–1941 interleukin-6 in 3T3-L1 adipocytes. Biochem. Biophys.
64 Moore, G. B., Chapman, H., Holder, J. C. et al. (2001) Res. Commun. 301, 1045–1050
Differential regulation of adipocytokine mRNAs by 84 Dandona, P., Aljada, A. and Bandyopadhyay, A. (2004)
rosiglitazone in db/db mice. Biochem. Biophys. Inflammation: the link between insulin resistance, obesity
Res. Commun. 286, 735–741 and diabetes. Trends Immunol. 25, 4–7
65 Shojima, N., Sakoda, H., Ogihara, T. et al. (2002) 85 Fernandez-Real, J. M. and Ricart, W. (2003) Insulin
Humoral regulation of resistin expression in 3T3-L1 and resistance and chronic cardiovascular inflammatory
mouse adipose cells. Diabetes 51, 1737–1744 syndrome. Endocr. Rev. 24, 278–301
66 Fujita, H., Fujishima, H., Morii, T. et al. (2002) Effect of 86 Lu, S. C., Shieh, W. Y., Chen, C. Y., Hsu, S. C. and Chen,
metformin on adipose tissue resistin expression in db/db H. L. (2002) Lipopolysaccharide increases resistin gene
mice. Biochem. Biophys. Res. Commun. 298, 345–349 expression in vivo and in vitro. FEBS Lett. 530, 158–162


C 2005 The Biochemical Society
Role of resistin in obesity, insulin resistance and Type II diabetes 255

87 Harsch, I. A., Koebnick, C., Wallaschofski, H. et al. 108 Pizzuti, A., Argiolas, A., Di Paola, R. et al. (2002)
(2004) Resistin levels in patients with obstructive sleep An ATG repeat in the 3 -untranslated region of the
apnoea syndrome: the link to subclinical inflammation? human resistin gene is associated with a decreased risk of
Med. Sci. Monit. 10, CR510–CR515 insulin resistance. J. Clin. Endocrinol. Metab. 87,
88 Kaser, S., Kaser, A., Sandhofer, A., Ebenbichler, C. F., 4403–4406
Tilg, H. and Patsch, J. R. (2003) Resistin messenger-RNA 109 Azuma, K., Oguchi, S., Matsubara, Y. et al. (2004) Novel
expression is increased by pro-inflammatory cytokines resistin promoter polymorphisms: association with serum
in vitro. Biochem. Biophys. Res. Commun. 309, 286–290 resistin level in Japanese obese individuals.
89 Wellen, K. E. and Hotamisligil, G. S. (2003) Horm. Metab. Res. 36, 564–570
Obesity-induced inflammatory changes in adipose tissue. 110 Osawa, H., Onuma, H., Murakami, A. et al. (2002)
J. Clin. Invest. 112, 1785–1788 Systematic search for single nucleotide polymorphisms in
90 Charriere, G., Cousin, B., Arnaud, E. et al. (2003) the resistin gene: the absence of evidence for the
Preadipocyte conversion to macrophage. Evidence of association of three identified single nucleotide
plasticity. J. Biol. Chem. 278, 9850–9855 polymorphisms with Japanese type 2 diabetes. Diabetes
91 Rajala, M. W. and Scherer, P. E. (2003) The adipocyte: at 51, 863–866
the crossroads of energy homeostasis, inflammation, and 111 Ochi, M., Osawa, H., Onuma, H. et al. (2003) The
atherosclerosis. Endocrinology 144, 3765–3773 absence of evidence for major effects of the frequent SNP
92 Haffner, S. M. (2003) Insulin resistance, inflammation, + 299G > A in the resistin gene on susceptibility to insulin
and the prediabetic state. Am. J. Cardiol. 92, 18J–26J resistance syndrome associated with Japanese type 2
93 Wulster-Radcliffe, M. C., Ajuwon, K. M., Wang, J., diabetes. Diabetes Res. Clin. Pract. 61, 191–198
Christian, J. A. and Spurlock, M. E. (2004) Adiponectin 112 Montague, C. T., Prins, J. B., Sanders, L., Digby, J. E.
differentially regulates cytokines in porcine macrophages. and O’Rahilly, S. (1997) Depot- and sex-specific
Biochem. Biophys. Res. Commun. 316, 924–929 differences in human leptin mRNA expression:
94 Yokota, T., Oritani, K., Takahashi, I. et al. (2000) implications for the control of regional fat distribution.
Adiponectin, a new member of the family of soluble Diabetes 46, 342–347
defense collagens, negatively regulates the growth 113 Lonnqvist, F., Nordfors, L., Jansson, M., Thorne, A.,
of myelomonocytic progenitors and the functions of Schalling, M. and Arner, P. (1997) Leptin secretion from
macrophages. Blood 96, 1723–1732 adipose tissue in women. Relationship to plasma levels
95 Jhun, B. S., Jin, Q., Oh, Y. T. et al. (2004) and gene expression. J. Clin. Invest. 99, 2398–2404
5-Aminoimidazole-4-carboxamide riboside suppresses 114 Kim, Y. W. and Scarpace, P. J. (2003) Repeated
lipopolysaccharide-induced TNF-α production through fasting/refeeding elevates plasma leptin without
inhibition of phosphatidylinositol 3-kinase/Akt increasing fat mass in rats. Physiol. Behav. 78, 459–464
activation in RAW 264.7 murine macrophages. 115 Hube, F. and Hauner, H. (1999) The role of TNF-α in
Biochem. Biophys. Res. Commun. 318, 372–380 human adipose tissue: prevention of weight gain at the
96 Ajuwon, K. M. and Spurlock, M. E. (2005) Adiponectin expense of insulin resistance? Horm. Metab. Res. 31,
inhibits LPS-induced NFκB activation and IL-6 626–631
production, and increases PPARγ 2 expression in 116 Grossi, S. G. (2001) Treatment of periodontal disease and
adipocytes. Am. J. Physiol. Regulatory Integrative control of diabetes: an assessment of the evidence and
Comp. Physiol. 288, R1220–R1225 need for future research. Ann. Periodontol. 6, 138–145
97 Cooke, J. P. and Oka, R. K. (2002) Does leptin cause 117 Fruhbeck, G., Gomez-Ambrosi, J., Muruzabal, F. J. and
vascular disease? Circulation 106, 1904–1905 Burrell, M. A. (2001) The adipocyte: a model for
98 Reilly, M. P., Lehrke, M., Wolfe, M. L., Rohatgi, A., integration of endocrine and metabolic signaling in energy
Lazar, M. A. and Rader, D. J. (2005) Resistin is an metabolism regulation. Am. J. Physiol. Endocrinol.
inflammatory marker of atherosclerosis in humans. Metab. 280, E827–E847
Circulation 111, 932–939 118 Bullo, M., Garcia-Lorda, P., Peinado-Onsurbe, J. et al.
99 Verma, S., Li, S. H., Wang, C. H. et al. (2003) Resistin (2002) TNF-α expression of subcutaneous adipose tissue
promotes endothelial cell activation: further evidence of in obese and morbid obese females: relationship to
adipokine–endothelial interaction. Circulation 108, adipocyte LPL activity and leptin synthesis. Int. J. Obes.
736–740 Relat. Metab. Disord. 26, 652–658
100 Kawanami, D., Maemura, K., Takeda, N. et al. (2004) 119 Hotamisligil, G. S., Arner, P., Caro, J. F., Atkinson, R. L.
Direct reciprocal effects of resistin and adiponectin on and Spiegelman, B. M. (1995) Increased adipose tissue
vascular endothelial cells: a new insight into expression of tumor necrosis factor-α in human obesity
adipocytokine-endothelial cell interactions. and insulin resistance. J. Clin. Invest. 95, 2409–2415
Biochem. Biophys. Res. Commun. 314, 415–419 120 Hotamisligil, G. S., Shargill, N. S. and Spiegelman, B. M.
101 Rolph, M. S., Zimmer, S., Bottazzi, B., Garlanda, C., (1993) Adipose expression of tumor necrosis factor-α:
Mantovani, A. and Hansson, G. K. (2002) Production of direct role in obesity-linked insulin resistance. Science
the long pentraxin PTX3 in advanced atherosclerotic 259, 87–91
plaques. Arterioscler., Thromb., Vasc. Biol. 22, e10–e14 121 Mohamed-Ali, V., Goodrick, S., Rawesh, A. et al. (1997)
102 Libby, P. (2002) Inflammation in atherosclerosis. Nature Subcutaneous adipose tissue releases interleukin-6, but
(London) 420, 868–874 not tumor necrosis factor-α, in vivo. J. Clin.
103 Hotta, K., Funahashi, T., Arita, Y. et al. (2000) Plasma Endocrinol. Metab. 82, 4196–4200
concentrations of a novel, adipose-specific protein, 122 Xing, Z., Gauldie, J., Cox, G. et al. (1998) IL-6 is an
adiponectin, in type 2 diabetic patients. Arterioscler., antiinflammatory cytokine required for controlling local
Thromb., Vasc. Biol. 20, 1595–1599 or systemic acute inflammatory responses. J. Clin. Invest.
104 Engert, J. C., Vohl, M. C., Williams, S. M. et al. (2002) 101, 311–320
5 flanking variants of resistin are associated with obesity. 123 Krogh-Madsen, R., Moller, K., Dela, F., Kronborg, G.,
Diabetes 51, 1629–1634 Jauffred, S. and Pedersen, B. K. (2004) Effect of
105 Mattevi, V. S., Zembrzuski, V. M. and Hutz, M. H. (2004) hyperglycemia and hyperinsulinemia on the response of
A resistin gene polymorphism is associated with body IL-6, TNF-α, and FFAs to low-dose endotoxemia in
mass index in women. Hum. Genet. 115, 208–212 humans. Am. J. Physiol. Endocrinol. Metab. 286,
106 Osawa, H., Yamada, K., Onuma, H. et al. (2004) The G/G E766–E772
genotype of a resistin single-nucleotide polymorphism at 124 Febbraio, M. A. and Pedersen, B. K. (2002)
− 420 increases type 2 diabetes mellitus susceptibility by Muscle-derived interleukin-6: mechanisms for activation
inducing promoter activity through specific binding of and possible biological roles. FASEB J. 16, 1335–1347
Sp1/3. Am. J. Hum. Genet. 75, 678–686 125 Ostrowski, S. R., Plomgaard, P., Fischer, C. P. et al. (2005)
107 Ma, X., Warram, J. H., Trischitta, V. and Doria, A. (2002) Interleukin-6 infusion during human endotoxaemia
Genetic variants at the resistin locus and risk of type 2 inhibits in vitro release of the urokinase receptor from
diabetes in Caucasians. J. Clin. Endocrinol. Metab. 87, peripheral blood mononuclear cells. Scand. J. Immunol.
4407–4410 61, 197–206


C 2005 The Biochemical Society
256 C. M. Kusminski, P. G. McTernan and S. Kumar

126 Juhan-Vague, I. and Alessi, M. C. (1997) PAI-1, obesity, 143 Liu, Y. M., Moldes, M., Bastard, J. P. et al. (2004)
insulin resistance and risk of cardiovascular events. Adiponutrin: a new gene regulated by energy balance in
Thromb. Haemostasis 78, 656–660 human adipose tissue. J. Clin. Endocrinol. Metab. 89,
127 Van Harmelen, V., Ariapart, P., Hoffstedt, J., 2684–2689
Lundkvist, I., Bringman, S. and Arner, P. (2000) Increased 144 Atzmon, G., Yang, X. M., Muzumdar, R., Ma, X. H.,
adipose angiotensinogen gene expression in human Gabriely, I. and Barzilai, N. (2002) Differential gene
obesity. Obes. Res. 8, 337–341 expression between visceral and subcutaneous fat depots.
128 Fried, S. K. and Kral, J. G. (1987) Sex differences in Horm. Metab. Res. 34, 622–628
regional distribution of fat cell size and lipoprotein lipase 145 Morash, B. A., Ur, E., Wiesner, G., Roy, J. and
activity in morbidly obese patients. Int. J. Obes. 11, Wilkinson, M. (2004) Pituitary resistin gene expression:
129–140 effects of age, gender and obesity. Neuroendocrinology
129 Fruebis, J., Tsao, T. S., Javorschi, S. et al. (2001) 79, 149–156
Proteolytic cleavage product of 30-kDa adipocyte 146 Morash, B. A., Willkinson, D., Ur, E. and Wilkinson, M.
complement-related protein increases fatty acid oxidation (2002) Resistin expression and regulation in mouse
in muscle and causes weight loss in mice. Proc. Natl. pituitary. FEBS Lett. 526, 26–30
Acad. Sci. U.S.A. 98, 2005–2010 147 Milan, G., Granzotto, M., Scarda, A. et al. (2002)
130 Arita, Y., Kihara, S., Ouchi, N. et al. (1999) Paradoxical Resistin and adiponectin expression in visceral fat
decrease of an adipose-specific protein, adiponectin, in of obese rats: effect of weight loss. Obes. Res. 10,
obesity. Biochem. Biophys. Res. Commun. 257, 79–83 1095–1103
131 Fisher, F. M., McTernan, P. G., Valsamakis, G. et al. (2002) 148 Dietze, D., Koenen, M., Rohrig, K., Horikoshi, H.,
Differences in adiponectin protein expression: effect of fat Hauner, H. and Eckel, J. (2002) Impairment of
depots and type 2 diabetic status. Horm. Metab. Res. 34, insulin signaling in human skeletal muscle cells by
650–654 co-culture with human adipocytes. Diabetes 51,
132 Samad, F. and Loskutoff, D. J. (1996) Tissue distribution 2369–2376
and regulation of plasminogen activator inhibitor-1 in 149 Minn, A. H., Patterson, N. B., Pack, S. et al. (2003)
obese mice. Mol. Med. 2, 568–582 Resistin is expressed in pancreatic islets. Biochem.
133 Montague, C. T., Prins, J. B., Sanders, L. et al. (1998) Biophys. Res. Commun. 310, 641–645
Depot-related gene expression in human subcutaneous 150 Yura, S., Sagawa, N., Itoh, H. et al. (2003) Resistin is
and omental adipocytes. Diabetes 47, 1384–1391 expressed in the human placenta. J. Clin.
134 Lefebvre, A. M., Laville, M., Vega, N. et al. (1998) Endocrinol. Metab. 88, 1394–1397
Depot-specific differences in adipose tissue gene 151 Delhanty, P. J., Mesotten, D., McDougall, F. and
expression in lean and obese subjects. Diabetes 47, 98–103 Baxter, R. C. (2002) Growth hormone rapidly induces
135 Faraj, M., Lu, H. L. and Cianflone, K. (2004) Diabetes, resistin gene expression in white adipose tissue of
lipids, and adipocyte secretagogues. Biochem. Cell. Biol. spontaneous dwarf (SDR) rats. Endocrinology 143,
82, 170–190 2445–2448
136 Kojima, M., Hosoda, H., Date, Y., Nakazato, M., 152 Ling, C., Kindblom, J., Wennbo, H. and Billig, H. (2001)
Matsuo, H. and Kangawa, K. (1999) Ghrelin is a Increased resistin expression in the adipose tissue of male
growth-hormone-releasing acylated peptide from prolactin transgenic mice and in male mice with elevated
stomach. Nature (London) 402, 656–660 androgen levels. FEBS Lett. 507, 147–150
137 Tschop, M., Smiley, D. L. and Heiman, M. L. (2000) 153 Kochan, Z. and Karbowska, J. (2004)
Ghrelin induces adiposity in rodents. Nature (London) Dehydroepiandrosterone up-regulates resistin gene
407, 908–913 expression in white adipose tissue. Mol. Cell. Endocrinol.
138 Tschop, M., Weyer, C., Tataranni, P. A., Devanarayan, V., 218, 57–64
Ravussin, E. and Heiman, M. L. (2001) Circulating 154 Huang, S. W., Seow, K. M., Ho, L. T. et al. (2005) Resistin
ghrelin levels are decreased in human obesity. Diabetes 50, mRNA levels are downregulated by estrogen in vivo and
707–709 in vitro. FEBS Lett. 579, 449–454
139 Wren, A. M., Small, C. J., Ward, H. L. et al. (2000) The 155 Iglesias, P., Alvarez Fidalgo, P., Codoceo, R. and Diez,
novel hypothalamic peptide ghrelin stimulates food intake J. J. (2003) Serum concentrations of adipocytokines in
and growth hormone secretion. Endocrinology 141, patients with hyperthyroidism and hypothyroidism
4325–4328 before and after control of thyroid function.
140 Poykko, S. M., Kellokoski, E., Horkko, S., Kauma, H., Clin. Endocrinol. 59, 621–629
Kesaniemi, Y. A. and Ukkola, O. (2003) Low plasma 156 Yuzuriha, H., Inui, A., Goto, K., Asakawa, A.,
ghrelin is associated with insulin resistance, hypertension, Fujimiya, M. and Kasuga, M. (2003)
and the prevalence of type 2 diabetes. Diabetes 52, Intracerebroventricular administration of NPY stimulates
2546–2553 resistin gene expression in mice. Int. J. Mol. Med. 11,
141 Baulande, S., Lasnier, F., Lucas, M. and Pairault, J. (2001) 675–676
Adiponutrin, a transmembrane protein corresponding to 157 Fasshauer, M., Klein, J., Neumann, S., Eszlinger, M. and
a novel dietary- and obesity-linked mRNA specifically Paschke, R. (2001) Isoproterenol inhibits resistin gene
expressed in the adipose lineage. J. Biol. Chem. 276, expression through a GS -protein-coupled pathway in
33336–33344 3T3-L1 adipocytes. FEBS Lett. 500, 60–63
142 Polson, D. A. and Thompson, M. P. (2003) Adiponutrin 158 Zhong, Q., Lin, C. Y., Clarke, K. J., Kemppainen, R. J.,
mRNA expression in white adipose tissue is rapidly Schwartz, D. D. and Judd, R. L. (2002) Endothelin-1
induced by meal-feeding a high-sucrose diet. inhibits resistin secretion in 3T3-L1 adipocytes.
Biochem. Biophys. Res. Commun. 301, 261–266 Biochem. Biophys. Res. Commun. 296, 383–387

Received 25 February 2005/1 April 2005; accepted 6 April 2005


Published on the Internet 24 August 2005, doi:10.1042/CS20050078


C 2005 The Biochemical Society

You might also like