You are on page 1of 18

Current Diabetes Reviews, 2006, 2, 195-211 195

Relationship Between Inflammation, Insulin Resistance and Type 2


Diabetes: ‘Cause or Effect’?

Jerry R. Greenfield1 and Lesley V. Campbell1,2,*

1
Diabetes and Obesity Research Program, Garvan Institute of Medical Research, Sydney, Australia
2
Diabetes Centre, St. Vincent’s Hospital, Sydney, Australia

Abstract: Inflammation has been implicated as an important aetiological factor in the development of both insulin
resistance and type 2 diabetes mellitus. This conclusion is predominantly drawn from studies demonstrating associations
between elevated (but ‘normal range’) levels of circulating acute phase inflammatory markers, typified by C-reactive
protein (CRP), and indices of insulin resistance and the development of type 2 diabetes. There is debate as to whether
these associations are independent of body fatness or, rather, an epiphenomenon of obesity, particularly central obesity, a
strong predictor of insulin resistance and type 2 diabetes and an important source of inflammatory cytokines, such as
interleukin-6. Some of this controversy and the inability to draw definitive conclusions from these studies relate to the fact
that most studies measure body fat and its distribution indirectly using anthropometric estimates, such as Body Mass
Index and waist circumference, rather than directly by dual-energy X-ray absorptiometry, computed tomography or
magnetic resonance imaging. Furthermore, use of the term inflammation may be inappropriate when describing mild
elevations of CRP in the ‘normal range’ in the absence of the other changes that characterise classical inflammatory
diseases, such as a reduction in levels (or evidence of consumption) of complement proteins. Debate as to whether obesity
mediates the association between circulating levels of inflammatory markers and insulin resistance can be resolved by
well-designed studies using body fat measured by gold-standard methods. In this review, we present evidence to support
the suggestion that body fat is the primary determinant of circulating inflammatory marker levels in the basal state and
that marginally elevated levels of circulating interleukin-6 and CRP in obesity are a consequence rather than a cause of
insulin resistance. The importance of genetic influences in determining both body fatness and circulating CRP levels will
also be discussed. The review will conclude with a discussion of possible mechanisms linking body fat and insulin
resistance to elevated circulating levels of inflammatory markers, including the possible role of the toll-like family of
immune receptors.

INTRODUCTION reduced levels) of complement proteins, the use of the term


inflammation may not be appropriate when describing
The Metabolic Syndrome, also known as the Insulin
mildly raised (but normal range) levels of inflammatory
Resistance Syndrome, Syndrome X, Cardiovascular
markers [9].
Dysmetabolic Syndrome and the ‘deadly quartet’, consists of
abdominal or visceral adiposity, insulin resistance, Recent interest in the hypothesis that inflammation plays
dyslipidaemia (hypertriglyceridaemia, low high-density an important aetiological role in the development and
lipoprotein [HDL] cholesterol and a predominance of small, progression of coronary artery disease [10] is derived from
dense low-density lipoprotein [LDL] cholesterol) and numerous studies demonstrating associations between
hypertension [1-5]. While there is debate about the baseline levels of inflammatory markers, particularly C-
terminology [6], the importance of identifying and treating reactive protein (CRP), and the development of
components of the syndrome lies in the associated increased cardiovascular disease in healthy subjects [11]. This
risk of type 2 diabetes and cardiovascular disease. hypothesis is supported by predominantly in vitro data
reporting that CRP may directly promote pro-atherogenic
The coexistence of type 2 diabetes and atherosclerotic
processes in vascular cells [12, 13]. Although the risk
cardiovascular disease has raised the possibility that attributable to a raised CRP level in humans may be less than
common aetiological mechanisms contribute to their previously reported [11, 14], it nonetheless remains an
development. Indeed, a number of authors have recently independent predictor of cardiovascular mortality and has
suggested that inflammation is a common aetiological factor
been proposed to be a potentially useful additional
unifying these metabolic disorders [7, 8]. However, as
prognostic indicator in individuals with intermediate
discussed later, in the absence of significantly elevated levels (Framingham risk score 10 – 20%) 10-year coronary heart
of circulating acute phase reactants and consumption (ie. disease risk [15].
However, it remains controversial whether inflammation
(usually represented by an elevated circulating CRP level) is
*Address correspondence to this author at the Diabetes and Obesity directly involved in the pathogenesis of insulin resistance
Research Program, Garvan Institute of Medical Research, 384 Victoria St,
Darlinghurst, Sydney, Australia; Tel: 61 2 9295 8202; Fax: 61 2 9295 8201; and type 2 diabetes. An alternative hypothesis that will be
E-mail: l.campbell@garvan.org.au discussed is that increased adiposity, itself a predictor of

1573-3998/06 $50.00+.00 © 2006 Bentham Science Publishers Ltd.


196 Current Diabetes Reviews, 2006, Vol. 2, No. 2 Greenfield and Campbell

Fig. (1). Hypothesised relationships between obesity, insulin resistance and pro- and anti-inflammatory cytokines. It has been suggested that
body fat-derived adipocytokines directly promote (interleukin-6 and TNF-α) or protect against (adiponectin) the development of insulin
resistance (panel A). An alternative hypothesis is that increased circulating levels of proinflammatory cytokines, and decreased levels of
antiinflammatory cytokines, are a consequence, rather than a cause, of insulin resistance, primarily driven by increased body fat (panel B).

diabetes risk and a source of inflammatory cytokines, Role of Fatty Acids in the Pathogenesis of Insulin
mediates these associations and interacts with insulin Resistance and Type 2 Diabetes
resistance to raise CRP levels in the circulation [8, 9, 16, 17]
(Fig. 1). Type 2 diabetes is characterised by insulin resistance and
decreased insulin secretion [23]. It is widely accepted that
This article will review the evidence for whether the accumulation of fatty acids in metabolically active (non-
inflammation plays a primary role in the pathogenesis of adipose) tissues, such as muscle and liver, plays a major
insulin resistance and type 2 diabetes and the contribution of aetiological role in insulin resistance, with much research
body fat to relationships between CRP and these metabolic directed towards the discovery of mechanisms by which lipid
disorders. Furthermore, we will propose possible biological oversupply inhibits insulin action [24-28]. Recent studies
mechanisms by which body fat and insulin resistance may postulate that primary or secondary deposition of muscle
lead to elevated circulating levels of inflammatory markers. triglyceride and impaired mitochondrial oxidative
Specific attention will be focused on CRP, as it is the most phosphorylation explain the early presence of insulin
commonly studied inflammatory marker in relation to resistance in genetically predisposed individuals [29].
diabetes and cardiovascular risks.
The major source of fatty acids in the circulation is
PATHOGENESIS OF INSULIN RESISTANCE AND adipose tissue. In contrast to peripheral or glutofemoral fat, a
TYPE 2 DIABETES MELLITUS – A POSSIBLE central pattern of fat distribution has been identified as being
INFLAMMATORY BASIS? particularly adverse with respect to metabolic risk, with
strong relationships reported between increased abdominal
The number of people affected by type 2 diabetes is fat and impaired insulin action in humans [30-34]. In
predicted to increase from 135 million in 1995 to 300 million addition to the importance of the amount of fat in the
in 2025 [18]. This is largely attributable to increases in the abdominal region, qualitative differences in adipocyte
prevalence of overweight and obesity, typified by developed biology and fat cell metabolism contribute to the depot-
countries such as the United States [19], Australia [20] and specific characteristics of central adiposity. In this regard,
the United Kingdom [21]. These disturbing trends have led omental adipocytes have been shown to be more responsive
to widespread interest in the contributory pathophysiological to the lipolytic effects of noradrenaline than peripheral
mechanisms and, hence, possible interventions, both adipocytes in men and women [35, 36]. Increased sensitivity
pharmacological and non-pharmacological, to prevent or at of omental vs peripheral adipocytes to noradrenaline-
least delay the development of these metabolic disorders mediated lipolysis is accompanied by resistance of omental
[22]. adipocytes to the anti-lipolytic effects of insulin [27, 37, 38].
Inflammation, Insulin Resistance and Type 2 Diabetes Current Diabetes Reviews, 2006, Vol. 2, No. 2 197

Table 1. Studies of Prospective Associations between C-reactive Protein and other Inflammatory Markers and the Development of
Type 2 Diabetes Mellitus

Risk of development of type 2 diabetes (adjusted Effect of (further) adjustment for adiposity*
Study
for factors other than adiposity in some studies) (and other factors in some studies)

ARIC study [47] OR extreme quartiles factor VII, 1.7; fibrinogen, 1.5; NS except for factor VIII in women
vWF, 1.5 in women (NS in men); factor VIII, 1.4 in
men, 2.2 in women.
ARIC study [48] OR extreme quartiles WBC count, 1.9; low albumin, WBC count and sialic acid: some attenuation;
1.3; fibrinogen, 1.2; sialic acid, 3.7; oromucoid, 7.9; oromucoid: minimally changed; remainder NS
α1-antitrypsin and haptoglobin, NS
Women’s Health Study [49] RR extreme quartiles CRP, 15.7; IL-6, 7.5 RR CRP, 4.2; IL-6 NS
th th
Cardiovascular Health Study [50] OR 75 vs 25 percentile CRP, 2.0; fibrinogen, Minimally changed
albumin, WBC count, platelet count and factor VIIIc
NS
IRAS [51] OR for 1SD increase in CRP, 1.4; fibrinogen, 1.2; CRP, fibrinogen: NS PAI-1: minimally changed
PAI-1, 1.99
th th
Pima Indians [52] RH 90 vs 10 percentile WBC count, 2.7 Minimally changed (after adjustment for percent
body fat)
WOSCOPS [53] HR extreme quintiles CRP, 6.1 Markedly attenuated but statistically significant
Mexico City Diabetes Study [54] OR extreme tertiles CRP, 5.5 (women only; NS in Minimally changed
men)
ARIC study [55] HR extreme quartiles CRP, 2.8; IL-6, 2.5; Only IL-6 remained statistically significant
orosomucoid, 2.3; sialic acid, 1.9
MONICA Augsburg Cohort Study [56] HR extreme quartiles CRP, 2.8 NS
Japanese Americans [57] Results prior to adjustment for adiposity not HR extreme quartiles CRP, 2.8 in men, 3.1 in
presented women
Hoorn Study [58] OR extreme tertiles CRP, 3.0 (men only; NS in No effect but substantially lowered after
women) adjusting for WHR
Pima Indians [59] CRP, IL-6, TNF-α, sE-selectin, CAMs and vWF not NA
significant predictors in Body Mass Index-matched
cases and controls
EPIC-Potsdam cohort [60] OR CRP ≥1.5 vs <1.5 mg/L, 3.5; extreme quartiles IL-6 NS; others markedly attenuated but
IL-6, 7.3; TNF-α, 2.3; IL-1β not a predictor statistically significant
Honk Kong Cardiovascular Risk Factors RR extreme quintiles CRP, 3.9 Attenuated but statistically significant
Prevalence Study [61]
Kuopio Ischaemic Heart Disease Risk Factor OR CRP ≥3.0 vs 0.1-0.99 mg/L, 4.1 Markedly attenuated but statistically significant
Study [62]
Nurses’ Health Study [63] OR extreme quintiles CRP, 7.1; IL-6, 3.3; TNF-αR2, Markedly attenuated but statistically significant
2.6
*Body Mass Index unless otherwise stated. ARIC: Atherosclerosis Risk in Communities Study. CAM: cellular adhesion molecules; CRP: C-reactive protein; EPIC: European
Prospective Investigation into Cancer and Nutrition; HR: hazard ratio; IL-6: interleukin-6; IRAS: Insulin Resistance Atherosclerosis Study; MONICA: Monitoring of Trends and
Determinants in Cardiovascular Disease; NA: not applicable; NS: not significant; OR: odds ratio; RH: relative hazard; RR: relative risk; TNF-αR2: tumour necrosis factor-α receptor
2; vWF: von Willebrand factor; WHR: waist-to-hip ratio; WOSCOPS: West of Scotland Coronary Prevention Study.

Increased lipolysis in omental adipocytes results in increased that the process of inflammation is a component of the
release of fatty acids into both the portal vein, which impairs Metabolic Syndrome [45, 46]. These conclusions
hepatic insulin extraction, stimulates gluconeogenesis and predominantly stem from the increasing number of studies
enhances triglyceride synthesis, as well as the peripheral reporting strong prospective associations between circulating
circulation, resulting in impaired insulin-stimulated glucose acute phase inflammatory markers, albeit at levels
uptake [28, 38-40]. It is important to note that the adverse significantly less than those observed during acute
consequences of increased lipolysis of abdominal adipocytes inflammation, and the development of type 2 diabetes [47-
are not restricted to the liver and that nonportal drainage 63] (Table 1). Although various inflammatory ‘markers’
affecting peripheral tissues is also important [41]. have been studied, including cytokines, chemokines and
acute phase reactants such as interleukin-6, white blood cell
Inflammation and CRP count, albumin, oromucoid, sialic acid, fibrinogen and other
coagulation factors, associations are variable and
Inflammation in general and CRP in particular have been inconsistent (Table 1). The most widely studied and pivotal
implicated as important additional factors in the inflammatory biomarker is CRP, a member of the pentraxin
pathogenesis of insulin resistance, glucose intolerance and family of proteins, consisting of five identical polypeptide
type 2 diabetes [42-44]. Some authors have even suggested
198 Current Diabetes Reviews, 2006, Vol. 2, No. 2 Greenfield and Campbell

Table 2. Heritability of C-Reactive Protein in Twin and Family Studies

2
Study Population Studied Heritability (h ) ICC in Monozygotic Twin Pairs

St. Thomas’ Twin Study [65] 186 postmenopausal twin pairs 0.45* 0.54
Norwegian Twin Panel [66] 78 male and female twin pairs Not calculated† 0.40
Elderly twin study [67] 282 elderly twin pairs 0.20‡ 0.30
NHLBI Family Heart study [68] 924 Caucasian sibling pairs 0.40 N/A
British Family study [69] 98 British Caucasian families 0.39 N/A
Diabetic Heart Study [70] 250 families 0.40# N/A
Japanese American Family Study [71] 68 extended Asian American kindreds 0.29¶ N/A
*Adjusted for Body Mass Index. †Formal heritability estimates not calculated as only monozygotic twins were recruited. ‡No difference after adjusting for Body Mass Index.
#Adjusted for duration of diabetes and statin and aspirin use (h2 = 0.49 before adjustment). ¶Adjusted for age, gender, Body Mass Index, smoking, hypertension, diabetes, myocardial
infarction and coronary artery disease and use of CRP lowering medications, oral contraceptives and hormone replacement therapy (h2 = 0.27 before adjustment). ICC: intra-class
correlation coefficient; NHLBI: National Heart, Lung, and Blood Institute.

subunits associated non-covalently in a symmetrical Furthermore, TNF-α inhibition had no effect on insulin
pentameric configuration [17]. In humans, CRP circulates in sensitivity in obese patients with [82, 83] and without [84]
mg/L concentrations, with a median level of 0.8 mg/L, 90th type 2 diabetes.
centile 3 mg/L and 99th centile 10 mg/L in healthy
populations [64]. Hence, levels <3 mg/L are considered Interleukin-6
‘normal range’ and levels >10 mg/L are suggestive of
Interleukin-6 is a pro-inflammatory cytokine secreted
inflammation [12]. Circulating CRP levels are stable over
time making them the most suitable markers of long-term predominantly from stimulated macrophages, fibroblasts and
vascular endothelial cells [85]. However, in the basal state,
vascular and metabolic risk [11, 17]. Part of this stability
up to 30% of circulating interleukin-6 is derived from
may relate to the significant heritability of CRP, which has
adipose tissue [80]. In addition, elegant studies have recently
been investigated in three twin [65-67] and four family [68-
shown that strongly exercising muscle releases interleukin-6
71] studies (Table 2). These studies have found that
in a beneficial glycogen ‘sparing’ manner, increasing hepatic
approximately 35% of the population variance in circulating
glucose output, independent of counter-regulatory hormone
CRP is heritable, even after correcting for Body Mass Index
production [85].
and other potentially confounding covariates.
Although interleukin-6 has been implicated in the
THE ROLE OF ADIPOCYTOKINES IN INFLAM- pathogenesis of insulin resistance, data are conflicting [86].
MATION Alternatively, it has been postulated that interleukin-6 may
be upregulated in insulin-resistant muscle as an adaptive
Although a number of fat-derived cytokines
(‘adipocytokines’) have been implicated as possible response aimed at reversing impaired glucose uptake and
inflammatory mediators of insulin resistance, the evidence is that interleukin-6 may even be a potential therapeutic agent
predominantly from in vitro studies and animal models [72, in type 2 diabetes [85]. Contrary to evidence that interleukin-
73]. Caution must be exercised in extrapolating from mouse 6 interferes with insulin signalling in hepatocyte cell lines
to man, considering that the main acute phase reactant in [87], Stouthard et al. reported that incubation of 3T3-L1
mice is serum amyloid P and that mouse CRP, which adipocytes with interleukin-6 enhanced basal and insulin-
circulates in trace concentrations, only peaks at 2 mg/L at the stimulated glucose uptake [88]. In mice, over-expression of
height of the animal’s inflammatory response [17, 74]. The interleukin-6 was found to reduce diabetes risk [89]; in
physiological relevance of some of these adipocytokines, another study, high-fat fed interleukin-6 deficient mice (with
such as tumour necrosis factor (TNF)-α and, particularly, a targeted null mutation in the interluekin-6 gene) developed
interleukin-6, to human insulin resistance has not been hyperglycaemia and impaired glucose disposal [90]. In
established [75, 76]. contrast, it was recently reported that interleukin-6 depletion
selectively improved hepatic insulin sensitivity in obese
TNF-α mice [91].
TNF-α, which has been shown to upregulate interleukin- In vivo studies of the role of interleukin-6 in human
6 gene expression [77] and downregulate adiponectin gene insulin resistance have also yielded conflicting results. In
expression [78] in 3T3-L1 adipocytes, attenuates insulin- one study, administration of recombinant human interleukin-
mediated suppression of hepatic glucose output and glucose 6 to healthy subjects increased glucose levels in a dose-
utilisation in rodents [79]. In contrast, however, there is no dependent fashion, without affecting insulin or C-peptide
convincing evidence that humans secrete physiologically levels [92]. In contrast, interleukin-6 increased the metabolic
significant amounts of TNF-α from adipose tissue [80]; clearance rate of glucose in patients with metastatic renal cell
instead, it is thought that TNF-α may have autocrine- cancer [93]. In another study, the acute administration of
paracrine, rather than endocrine, actions in man [73]. In a interleukin-6 to healthy human subjects had no effect on
recent study, plasma TNF-α level was unrelated to insulin muscle glucose uptake, whole-body glucose disposal or
sensitivity (by euglycaemic-hyperinsulinaemic clamp) [81]. endogenous glucose production [94]. Differences in dose and
Inflammation, Insulin Resistance and Type 2 Diabetes Current Diabetes Reviews, 2006, Vol. 2, No. 2 199

cohort characteristics may partly explain these opposing Circulating adiponectin levels are directly associated
findings. with insulin sensitivity (measured by the euglycaemic-
hyperinsulinaemic clamp), even after adjusting for percent
An alternative view is that circulating interleukin-6 level
body fat [102]. A possible role for decreased adiponectin in
is more likely to be an index of adiposity rather than a direct
the pathogenesis of insulin resistance was provided by a
regulator of insulin action in man [81]. Consistent with this
suggestion, a study of Pima Indians found that the inverse study by Hotta et al. in Rhesus monkeys, which reported that
relationship between fasting plasma IL-6 concentration and plasma levels of adiponectin decreased in parallel with the
insulin sensitivity (by euglycaemic-hyperinsulinaemic development of insulin resistance [107]. Furthermore,
clamp) was attenuated after correcting for percent body fat circulating adiponectin levels are lower in healthy subjects
who develop type 2 diabetes compared to those who do not
[95]. Consistent with this theory, in a study of Body Mass
[108, 109]. Vionnet et al. recently mapped a diabetes
Index-matched insulin-sensitive controls and insulin-
susceptibility locus to human chromosome 3q27 [110], a
resistant type 2 diabetic relatives, there was no significant
region where the gene encoding adiponectin is located [111].
difference in interleukin-6 (or TNF-α) between the groups,
Polymorphisms in the adiponectin gene ACDC have been
despite 60% lower rates of glucose infusion and insulin-
stimulated peripheral glucose uptake in the latter group reported to be associated with type 2 diabetes in a Japanese
during hyperinsulinaemic-euglycaemic clamp studies [29]. population [111]; however, this was not replicated in a recent
In a recent study, sustained reduction in free fatty acid levels study of Pima Indians [112].
by acipimox, an inhibitor of lipolysis, significantly enhanced Receptors for adiponectin have recently been identified;
hepatic and muscle insulin sensitivity, without affecting adiponectin receptor 1 (AdipoR1), although expressed
weight (and presumably fat mass) or plasma levels of TNF-α ubiquitously, is most abundant in muscle and has high-
and interleukin-6 [96]. Furthermore, there was no affinity for globular and low affinity for full-length
relationship between whole body glucose disposal and adiponectin [113]. In contrast, adiponectin receptor 2
plasma levels of these cytokines before and after treatment (AdipoR2) is predominantly expressed in liver and has
[96]. Taken together, these results suggest that the amount of intermediate affinity for both globular and full-length
body fat is an important determinant of circulating adiponectin [113]. This differential receptor expression may
interleukin-6 level and that body fat, when measured account for tissue-specific effects of various isoforms of
accurately, largely accounts for the association between circulating adiponectin.
increased circulating interleukin-6 and insulin resistance. In obese and lipoatrophic insulin-resistant mice,
treatment with adiponectin (particularly globular
Adiponectin
adiponectin) has been shown to improve insulin sensitivity
Adiponectin, also referred to as apM1 (adipose most and to enhance suppression of insulin-mediated hepatic
abundant gene transcript 1), Acrp 30 (adipocyte glucose output, reduce hepatic and skeletal muscle
complement-related protein of 30 kDa), adipoQ and GBP28 triglyceride content and increase fatty acid oxidation in
(gelatin binding protein of 28 kDa) is a large, abundant muscle [114, 115]. Although the mechanisms by which
collagen-like protein secreted exclusively from adipocytes adiponectin exerts these actions are debated, there is
[97, 98]. Adiponectin, the secretion of which is inhibited by evidence to suggest that increased tyrosine phosphorylation
TNF-α and interleukin-6 [73], is reported to have insulin- of the insulin receptor in muscle, activation of adenosine
sensitising, anti-atherosclerotic and anti-inflammatory monophosphate (AMP)-activated protein kinase in muscle
properties [99], the latter mediated, possibly, through down- and liver, increased activity of peroxisome proliferator
regulation of CRP production and synthesis [100]. In activated receptor (PPAR)-α and increased expression of
contrast to most other adipocytokines, plasma adiponectin genes encoding proteins involved in fatty acid transport and
concentration is paradoxically inversely associated with total oxidation (e.g. CD-36 and fatty acid transport protein-1) may
body and abdominal fat and reduced in obesity, due, most be involved [116, 117].
likely, to the strong and independent association between Despite this evidence in animals, similar studies using
adiponectin levels and insulin sensitivity [101-103]. Perhaps infused adiponectin have not been published in humans.
paradoxically, there is some evidence to suggest that omental Therefore, a definitive role for adiponectin in human insulin
fat may be a more important determinant of circulating resistance remains uncertain. Against adiponectin being an
adiponectin levels than subcutaneous abdominal fat, as important factor in human insulin resistance, we [9] and
adiponectin secretion from human omental (but not others [29] reported that insulin-sensitive and insulin-
subcutaneous) adipocytes increased with decreasing Body resistant relatives of subjects with type 2 diabetes, with
Mass Index and following incubation with insulin and/or similar Body Mass Index values, had similar levels of
insulin-sensitising drugs such as rosiglitazone (a circulating adiponectin. In another study, although plasma
thiazolidinedione) [104]. However, it should be noted that adiponectin was correlated with insulin-stimulated non-
adiponectin circulates in two main forms – a low molecular oxidative glucose disposal in a study of type 2 diabetic first-
weight trimer-dimer (hexamer) and a higher-order high degree relatives, levels did not change following acipimox
molecular weight (HMW) complex, comprising two or three treatment, despite a significant improvement in insulin action
hexamers [99, 105]. Measuring these distinct complexes, [96]. Until adiponectin is shown to have direct favourable
specifically the ratio of HMW-to-total adiponectin, has been effects on insulin sensitivity in man, the possibility that low
proposed to reflect changes in insulin sensitivity better than circulating adiponectin levels are a consequence rather than
total levels in some situations, such as with thiazolidinedione cause of insulin resistance cannot be excluded.
therapy [99, 106].
200 Current Diabetes Reviews, 2006, Vol. 2, No. 2 Greenfield and Campbell

ADIPOSITY IS AN IMPORTANT DETERMINANT OF (who developed diabetes) and controls (who did not develop
CIRCULATING INFLAMMATORY MARKERS AND diabetes) were matched for age, gender and adiposity (albeit
THEIR RELATIONSHIP WITH TYPE 2 DIABETES estimated by Body Mass Index and waist circumference),
RISK basal CRP levels were not associated with the risk of type 2
diabetes [59]. However, the use of indirect anthropometric
There is widespread debate as to whether associations estimates of body fatness in the majority of these studies
between inflammatory markers and type 2 diabetes are must limit conclusions regarding the role of obesity in the
independent of body fatness or, rather, an epiphenomenon of relationship between CRP and type 2 diabetes. Persistence of
obesity, particularly central obesity, which is a strong a significant relationship after adjusting for surrogate
predictor of insulin resistance and type 2 diabetes [30] and measures of body size does not indicate that relationships
an important source of inflammatory cytokines [80]. Some between CRP and type 2 diabetes are exclusive of body
of this controversy, together with the inability to draw fatness. This question can only be resolved using direct
definitive conclusions from most studies, relates to the fact adiposity measures. This important issue of residual
that body fat and its distribution are usually measured
confounding and its clinical implications has been discussed
indirectly using anthropometric estimates, such as Body
elsewhere in detail [127].
Mass Index, waist circumference and waist-to-hip ratio
[118]. The inaccuracy of anthropometric surrogates as Although CRP is primarily secreted by the liver in
measures of central abdominal fat stores was illustrated by a response to stimulation by pro-inflammatory cytokines,
recent study, which failed to detect a difference in waist particularly interleukin-6, a recent report suggested that CRP
circumference and waist-to-hip ratio between subjects with mRNA may be expressed in adipose tissue itself [100]. As
and without type 2 diabetes, despite a significant difference discussed later in the review, in the basal state, 25 – 30% of
in visceral fat area measured by magnetic resonance imaging total systemic interleukin-6 is derived from adipose tissue
[119]. [80]. Therefore, it is biologically plausible that adiposity is
The question as to whether obesity mediates the an important, and perhaps the primary, determinant of
association between circulating levels of inflammatory inflammatory cytokine production in healthy individuals and
markers and insulin resistance and type 2 diabetes can only that increased body fat accounts for the strong association
be resolved by studies which measure body fat directly using between basal CRP and subsequent risk of type 2 diabetes.
more accurate techniques, such as dual-energy X-ray Of note, production rates of interleukin-6 in omental fat are
absorptiometry, computed tomography (CT) and magnetic up to three times that of subcutaneous adipose tissue [128].
resonance imaging (MRI). CT and MRI represent ‘gold- Furthermore, recent evidence suggests that in addition to
standard’ methods for measuring visceral fat and have been adipocytes, macrophages in fat may account for a significant
validated against cadaveric studies in humans [120, 121]. proportion of inflammatory cytokine production [129].
Dual-energy X-ray absorptiometry, which is commonly used In order to investigate the specific role of adiposity in
to measure bone mineral density in clinical practice, is an determining circulating CRP levels, we recently investigated
accurate, rapid and validated technique for measuring body the relationship between total body and central abdominal fat
fat mass and its distribution. Dual-energy X-ray (measured by dual-energy X-ray absorptiometry) and
absorptiometry not only quantifies adipose tissue depots, but circulating levels of CRP in healthy female twins [130]. As
also includes fat content within other tissues such as muscle. already mentioned above, approximately 35% of the
Dual-energy X-ray absorptiometry is associated with population variance in circulating CRP is heritable [65-71].
minimal radiation exposure and provides an accurate Furthermore, our group has reported that genetic factors
measure of total body and central abdominal adiposity, but explain up to 60 – 70% of the population variance in central
does not measure intra-abdominal and subcutaneous abdominal fat [131-133]. Of note, some of this genetic
abdominal fat depots independently [122]. However, influence is shared with total body fat [132]. Therefore it is
distinguishing intra-abdominal from subcutaneous important to control for genetic factors when examining the
abdominal fat may be less important than previously relationship between adiposity and circulating CRP levels.
thought, as there is evidence that subcutaneous abdominal fat The main finding of our study was that levels of CRP were
(perhaps deep subcutaneous abdominal fat in particular strongly related to total body and central abdominal fat. CRP
[123]) has a similar or even stronger relationship with was also associated with other components of the Metabolic
directly-measured insulin sensitivity than intra-abdominal fat Syndrome, including blood pressure, HDL cholesterol and
[124-126]. Indeed, it has been suggested that because the triglyceride levels. In order to determine whether these
amount of subcutaneous abdominal fat is large in relationships persisted after controlling for genetic
comparison with other fat compartments (e.g. intra- influences, we: (i) examined relationships between intra-pair
abdominal fat), it may constitute the most metabolically differences in CRP and these variables; and (ii) in
active fat depot influencing insulin sensitivity [124, 126]. monozygotic twins discordant for body fatness or lipid
In a number of studies [47-49, 51, 53, 55, 56, 58, 60-63], values, compared CRP levels in twins with the higher values
controlling for adiposity and other metabolic factors of these Metabolic Syndrome components with CRP levels
markedly attenuated, to the point of insignificance in some in twins with the lower values of these Metabolic Syndrome
studies [47, 48, 51, 55, 56], the association between components. In both analyses, CRP was consistently related
inflammatory markers and type 2 diabetes risk (Table 1). to total body fat, central abdominal fat, HDL cholesterol and
Furthermore, in a study of Pima Indians, in which cases triglyceride levels, independent of genetic, age, gender and
environmental effects.
Inflammation, Insulin Resistance and Type 2 Diabetes Current Diabetes Reviews, 2006, Vol. 2, No. 2 201

Few other studies have examined the relationship the Insulin Resistance Atherosclerosis Study, the association
between CRP levels and direct measures of adiposity. Using between low insulin sensitivity (as determined by the
dual-energy X-ray absorptiometry, we and others have frequently sampled intravenous glucose tolerance test) and
demonstrated significant associations between CRP and total higher CRP levels was considerably attenuated after
body fat [9, 134-145] and trunk fat [144-146]. Two adjusting for waist circumference alone [154].
subsequent studies [9, 134], both from our group, confirm In contrast to the above reports, in a study of healthy
our finding of a strong relationship between circulating women with a wide range of body fatness, central fat and
levels of CRP and central abdominal fat, a stronger insulin resistance (albeit estimated by waist circumference
determinant of insulin resistance than total body or trunk fat and homeostasis model assessment [HOMA] respectively)
[30]. Our results are consistent with studies that report
were both independently associated with CRP level [155].
associations between CRP and abdominal fat measured by
Similarly, percent total body fat (by dual-energy X-ray
CT [137, 138, 140, 143, 147, 148]. However, these studies
absorptiometry) and insulin resistance (by HOMA) were
are limited by the use of single-slice scanning, which, as we
independent predictors of CRP in a study of subjects with
have reported, may result in misclassification when ranking
varying degrees of glycaemia [135], although it should be
or ordering subjects based on abdominal adiposity [122]. noted that HOMA is an unreliable measure of insulin
Taken together, these studies highlight the importance of resistance in subjects with glucose intolerance [156]. While
body fat in determining circulating CRP levels and indicate differences in cohort size and characteristics may explain
that its confounding effect must be properly accounted for in
some of the disparity between studies, imprecision in the
future studies. methods used to measure adiposity and insulin sensitivity in
some of these studies is very likely to have contributed
ADIPOSITY VS INSULIN RESISTANCE IN
[127].
DETERMINING CIRCULATING LEVELS OF
INFLAMMATORY MARKERS
Reductions in CRP Levels Following Weight Loss are
Controversy exists as to the relative importance of body More Closely Related to Changes in Adiposity Than
fat and the closely related phenotype, insulin sensitivity, in Changes in Insulin Sensitivity
determining circulating levels of inflammatory markers. In
Although weight loss has been shown to lead to a
particular, it is unclear whether insulin resistance is
reduction in circulating CRP levels in obese subjects
associated with increased CRP levels in the absence of
following caloric restriction [143, 157-161] or bariatric
increased adiposity. Most of the studies investigating this surgery [162-165], relatively few studies have
question are limited by the use of indirect measures of body
simultaneously measured changes in fat mass, insulin
fatness and insulin sensitivity. The evidence investigating
sensitivity and CRP. A recent study raised the possibility that
this question is summarised in the following paragraphs.
Body Mass Index and macronutrient composition may
modify the CRP response to a weight-loss diet [166],
Body Fat Mediates the Relationship between CRP Levels
although the results are preliminary and require
and Insulin Sensitivity
confirmation.
Some studies have found that insulin resistance is not In a study of weight loss following gastric surgery,
associated with levels of inflammatory markers in the reductions in CRP levels paralleled the decrease in Body
circulation, despite strong and significant relationships Mass Index but were unrelated to changes in insulin
between CRP levels and Body Mass Index and waist sensitivity [163]. Similarly, in a recent diet-induced weight
circumference [149, 150]. Other studies suggest that loss study, reductions in CRP were significantly related to
adiposity mediates or confounds the inverse relationship the amount of weight lost, but were not correlated with the
between CRP and insulin sensitivity. For example, change in insulin sensitivity [167]. In a study of weight loss
controlling for Body Mass Index, waist circumference [151] induced by diet and exercise in overweight and obese
or total body fat measured by dual-energy X-ray postmenopausal women, although changes in total fat,
absorptiometry [144] eliminated associations between CRP visceral fat, CRP and interleukin-6 were inversely related to
and components of the Metabolic Syndrome. Consistent with the improvement in insulin sensitivity in univariate analysis,
these results, in a study using direct measures of insulin only visceral fat and levels of interleukin-6 were significant
sensitivity (euglycaemic-hyperinsulinaemic clamp) and predictors of the change in insulin sensitivity in stepwise
adiposity (dual-energy X-ray absorptiometry and CT), the multiple regression analysis [168]. In other words, CRP was
relationship between CRP and insulin-stimulated glucose not an independent predictor of weight loss-induced
disposal was attenuated in premenopausal women, and was improvements in insulin sensitivity, independent of
eliminated in postmenopausal women, by covarying for total adiposity.
body or intra-abdominal fat [140]. This is supported by two
other studies, in which the inverse relationship between CRP Subjects Concordant for Body Fat but Discordant for
and insulin sensitivity became non-significant after adjusting Insulin Sensitivity Have Similar Levels of Circulating
for total fat mass [152, 153]. In a study of Pima Indians, CRP
although percentage total body fat (by dual energy X-ray
absorptiometry) and insulin-stimulated glucose disposal (by Using bioelectrical impedance analysis to measure body
euglycaemic-hyperinsulinaemic clamp) explained 71% of the fat, Romano et al. recently reported that obese insulin-
total variance in circulating CRP levels, adiposity was the resistant subjects had higher CRP levels than obese insulin-
2
dominant factor (r = 0.51 and 0.20 respectively) [141]. In sensitive subjects [169]. However, in the same study, the
202 Current Diabetes Reviews, 2006, Vol. 2, No. 2 Greenfield and Campbell

latter group of subjects (mean fat mass 38.1 ± 8.6 kg) had process, complement is usually consumed and circulating
higher CRP levels than lean individuals with similar insulin levels are expected to be reduced. Although insulin
sensitivity (mean fat mass 19.2 ± 5.3 kg), implicating resistance and type 2 diabetes are often described as
adiposity as the main determinant of CRP. Although two inflammatory diseases, the use of the term ‘inflammation’
studies found that obese insulin-resistant subjects had higher may be inappropriate in the context of mildly elevated CRP
CRP levels than insulin-sensitive subjects ‘matched’ for levels (within the ‘normal range’) and normal complement
Body Mass Index [135, 157], no conclusion can be made protein levels. The reported association between minimally
from these studies regarding the relative influence of insulin elevated CRP levels and type 2 diabetes does not necessarily
sensitivity and body fat on CRP concentrations, as no direct implicate inflammation in the aetiology of insulin resistance
or true measure of body fat was performed [118]. and type 2 diabetes.
Furthermore, the use of the modified insulin suppression test
To further explore this question, we recently studied
to measure insulin sensitivity in one of these studies [157]
relationships between insulin sensitivity (by euglycaemic-
limits the applicability of these results, considering that
hyperinsulinaemic clamp), total body and abdominal obesity
glucose recovery following insulin injection is dependent on
(by dual-energy X-ray absorptiometry and MRI
a number of factors including insulin dose, the extent of the respectively), inflammatory mediators (CRP and
counter-regulatory hormone response and adiposity itself. adiponectin) and circulating levels of complement proteins
In a recent study of obese postmenopausal women, which (C3, C4, C1q, Factor B and Factor D) in 19 normoglycaemic
used direct measures of body fat (dual-energy X-ray normolipidaemic non-obese first-degree relatives of type 2
absorptiometry and single-slice CT), levels of CRP, diabetic subjects (REL) and 22 Body Mass Index-matched
interleukin-6 and TNF-α were similar in insulin-resistant controls with no known family history of type 2 diabetes
subjects with the Metabolic Syndrome and insulin-sensitive (CON) [9]. Despite similar body fat measures, REL were
subjects without the Metabolic Syndrome, a consequence, 20% less insulin sensitive than CON subjects (Table 3).
most likely, of the fact that the groups had similar amounts However, REL and CON had comparable, ‘normal range’
of total body and subcutaneous abdominal fat [170]. Using a levels of CRP, adiponectin and proteins of both the classical
similar design, a study comparing inflammatory markers in (C1q and C4) and alternative (C3, factors B and D)
premenopausal women with and without polycystic ovary complement pathways (Table 3) [9]. In other words, there
syndrome (PCOS), matched for Body Mass Index, found that was no evidence of inflammation (ie. elevated CRP levels
the relatively insulin-resistant PCOS group had similar CRP and complement consumption) in the REL group, despite
levels to relatively insulin-sensitive controls [171]. In the significantly lower insulin sensitivity.
whole group, controlling for Body Mass Index significantly
In analyses including the whole group, CRP was directly
attenuated the inverse relationship between insulin
related to measures of total body and central abdominal fat
sensitivity and CRP level. In contrast, controlling for insulin
and inversely associated with insulin sensitivity and
sensitivity had no effect on the relationship between Body
adiponectin levels (Table 4) [9]. CRP levels were related to
Mass Index and CRP levels. Consistent with this result, in
levels of acute-phase components of the alternative pathway
another study of women with PCOS, the relationship
(Factor B and C3) but not Factor D, a non-acute phase
between insulin resistance (estimated by HOMA) and CRP alternative pathway component. Of the classical complement
level was attenuated after adjusting for Body Mass Index pathway components, CRP was significantly associated with
[172]. Finally, controlling for Body Mass Index greatly
C1q only (Table 4). In multiple regression analysis with
attenuated associations of PCOS and CRP with carotid
glucose infusion rate as the dependent variable, CRP was not
intima-media wall thickness, raising the possibility that these
significantly related to insulin sensitivity, independently of
associations are also mediated by increased body fatness
total body fat. Similar results were found when total body fat
[173].
was replaced by either central abdominal fat or subcutaneous
abdominal fat [9].
IS INFLAMMATION ASSOCIATED WITH INSULIN
RESISTANCE IN THE ABSENCE OF OBESITY? In summary, our results suggest that, in the absence of
obesity, insulin resistance is not associated with the
Taken together, the studies described above suggest that recognised processes of inflammation and that the use of the
body fat is the primary determinant of circulating CRP levels term may be need to be revised when referring to
in the basal state and increase the likelihood that marginally associations between circulating inflammatory markers,
elevated levels of circulating CRP in insulin resistance are insulin resistance and type 2 diabetes.
mediated by increased body fat. However, it is still not clear
whether inflammation is a mediator of insulin resistance in POSSIBLE PATHOPHYSIOLOGICAL MECHANISMS
obesity, or, rather, whether levels of pro-inflammatory LINKING BODY FAT AND INSULIN RESISTANCE
cytokines and inflammatory markers, such as interleukin-6 TO ELEVATED CIRCULATING LEVELS OF
and CRP, are increased in obese subjects as a consequence of INFLAMMATORY MARKERS
insulin resistance [42].
Although the pathophysiological links between adiposity
At the height of an inflammatory response, circulating and insulin resistance and elevated levels of systemic
CRP levels may rise up to 10,000 fold [17]. CRP activates inflammatory markers remain unknown, possible
the classical complement pathway by binding to C1q,
mechanisms include: (1) cytokine production by adipocytes
leading to the activation of the terminal membrane attack
and immune cells within adipose tissue; (2) chronic over-
complex, C5 – C9 [174]. Hence, during an inflammatory stimulation of the immune response by toll-like receptors
Inflammation, Insulin Resistance and Type 2 Diabetes Current Diabetes Reviews, 2006, Vol. 2, No. 2 203

Table 3. Cohort Characteristics and Biochemical Variables in Relatives of Type 2 Diabetic Subjects (REL) and Healthy Controls
(CON)

Variable REL CON P-value

Age (y) 30.7 ± 1.6 31.3 ± 1.6 0.80


Weight (kg) 73.4 ± 4.1 73.7 ± 3.5 0.90
2
Body Mass Index (kg/m ) 25.8 ± 1.1 24.3 ± 0.8 0.29
Waist (cm) 80.0 ± 3.2 78.7 ± 2.6 0.76
Waist-to-hip ratio 0.79 ± 0.02 0.82 ± 0.02 0.38
Total body fat (kg)* 25.5 ± 2.5 20.9 ± 2.0 0.15
Fat free mass (FFM, kg)* 47.7 ± 2.6 52.9 ± 2.9 0.20
Central abdominal fat (kg)* 1.39 ± 0.15 1.21 ± 0.13 0.37
Subcutaneous abdominal fat (L)† 2.23 ± 0.29 1.69 ± 0.25 0.17
Visceral abdominal fat (L)† 0.59 ± 0.07 0.61 ± 0.11 0.88
Fasting plasma glucose (mmol/L) 5.0 ± 0.1 4.9 ± 0.1 0.35
2-hr post-challenge glucose (mmol/L)‡ 5.8 ± 0.2 5.2 ± 0.3 0.12
Fasting plasma insulin (mU/L) 10.6 ± 0.8 8.5 ± 0.9 0.08
Glucose infusion rate (µmol/min/kgFFM) 51.8 ± 3.9 64.9 ± 4.6 0.04
Fasting total cholesterol (mmol/L) 4.08 ± 0.22 4.19 ± 0.18 0.71
Fasting HDL cholesterol (mmol/L) 0.89 ± 0.07 0.90 ± 0.06 0.93
Fasting triglycerides (mmol/L) 0.81 ± 0.11 0.68 ± 0.05 0.27
Fasting non-esterified fatty acids (mmol/L) 0.46 ± 0.04 0.42 ± 0.05 0.58
C-reactive protein (mg/L) 1.55 ± 0.23 1.68 ± 0.46 0.33
Adiponectin (µg/mL) 14.6 ± 1.7 15.1 ± 1.4 0.82
C1q (mg/L) 103 ± 4 101 ± 2 0.67
C4 (mg/l) 218 ± 15 217 ± 16 0.96
C3 (g/L) 1.21 ± 0.07 1.05 ± 0.05 0.06
Factor B (mg/L) 263 ± 17 227 ± 13 0.10
Factor D (µg/mL) 1.39 ± 0.09 1.33 ± 0.06 0.58
*by dual-energy X-ray absorptiometry; †by magnetic resonance imaging in n=36 subjects (scans unavailable in one CON subject due to claustrophobia and
four REL subjects due to technical difficulties); ‡following 75 g oral glucose tolerance test. Copyright © 2004 American Diabetes Association. Reprinted from
[9] with permission from the American Diabetes Association.

(TLR); (3) an inherited acute phase ‘hyper-responsiveness’ adipocytes [128]. A series of elegant and detailed studies
in obese subjects; (4) loss of insulin-mediated inhibition of recently demonstrated that the number of macrophages in
cytokine-stimulated CRP production in insulin resistance; adipose tissue increases with increasing obesity and that
and (5) mitochondrial dysfunction. Each of these macrophages are responsible for a significant proportion of
mechanisms will be discussed briefly below. inflammatory cytokine production from fat tissue [175, 176].
However, the factors that attract macrophages to adipose
Cytokine Production by Adipose Tissue tissue, and the specific factors that stimulate these immune
cells to produce inflammatory cytokines in obesity, remain
Adipose tissue has been reported to produce
to be determined [129].
approximately 30% of systemic interleukin-6 in healthy
subjects [80]. Recent studies have demonstrated that Toll-Like Receptors
inflammatory cytokines originate not only from adipocytes,
but also from immune cells, specifically macrophages, Emerging evidence for a possible role of TLRs,
within adipose tissue [129]. Earlier evidence that cells other specifically TLR-4, may be relevant to this discussion. This
than adipocytes may produce inflammatory cytokines in group of recently discovered pattern-recognition receptors,
human adipose tissue was provided by Fried et al. who 10 of which have been described in humans, provide a
observed that only 10% of total tissue production of potential crucial link between innate immunity and
interleukin-6 by adipose tissue could be attributed to atherogenesis [177]. It is possible that stimulation of
204 Current Diabetes Reviews, 2006, Vol. 2, No. 2 Greenfield and Campbell

Table 4. Simple Linear Regression Analyses between C-reactive Protein Levels and Adiposity Measures, Metabolic Variables,
Adiponectin and Complement Proteins in Relatives of Type 2 Diabetic Subjects and Healthy Controls

Variable r-value P-value

Age (y) 0.12 0.45


Weight (kg) 0.34 0.03
2
Body Mass Index (kg/m ) 0.41 0.008
Waist (cm) 0.36 0.02
Waist-to-hip ratio 0.12 0.46
Total body fat (kg)* 0.35 0.03
Fat free mass (kg)* 0.15 0.35
Central abdominal fat (kg)* 0.43 0.005
Subcutaneous abdominal fat (L)† 0.59 0.0002
Visceral abdominal fat (L)† 0.38 0.02
Fasting plasma glucose (mmol/L) 0.32 0.045
2-hr post-challenge glucose (mmol/L)‡ 0.03 0.88
Fasting plasma insulin (mU/L) 0.26 0.11
Glucose Infusion Rate (µmol/min/kgFFM) -0.33 0.04
Fasting total cholesterol (mmol/L) 0.22 0.17
Fasting HDL cholesterol (mmol/L) -0.20 0.21
Fasting triglycerides (mmol/L) 0.39 0.01
Fasting non-esterified fatty acids (mmol/L) 0.17 0.30
Adiponectin (µg/mL) -0.34 0.03
C1q (mg/L) 0.34 0.03
C4 (mg/L) 0.30 0.06
C3 (g/L) 0.41 0.009
Factor B (mg/L) 0.43 0.005
Factor D (µg/mL) 0.10 0.56

*by dual-energy X-ray absorptiometry; †by magnetic resonance imaging in n = 36 subjects (scans unavailable in one CON subject due to claustrophobia and
four REL subjects due to technical difficulties); ‡following 75 g oral glucose tolerance test. Copyright © 2004 American Diabetes Association. Reprinted from
[9] with permission from the American Diabetes Association.

macrophages in fat to produce inflammatory cytokines is the density, associated with hypertriglyceridaemia and low HDL
result of TLR-4 activation. Stimulation of TLR-bearing concentrations in the Metabolic Syndrome, increase its
immune cells is known to directly regulate innate immune sensitivity to atherogenic oxidative modification [180].
genes, resulting in the secretion of pro-inflammatory Binding of these modified lipoproteins to TLR-4 on
cytokines and chemokines, including interleukin-6 [178, macrophages may be a potential mechanism stimulating
179]. cytokine production in obesity, particularly central obesity.
Although the specific ligands that trigger this immune
Acute Phase ‘Hyper-Responsiveness’
cascade in adipose tissue remain uncertain, lipoproteins may
be important candidates. Reports that oxidized LDL Polymorphisms in the TLR gene are associated with
(oxLDL) and mildly oxidized (minimally modified) LDL reduced circulating levels of pro-inflammatory cytokines, a
(mmLDL) are possible endogenous ligands for TLR-4 [177- diminished response to endotoxin-derived lipopoly-
179], raise the possibility that this modified lipoprotein, or saccharide (LPS), and greater susceptibility to acute bacterial
related lipid moieties, may contribute to the production of infection, but relative protection from atherosclerosis [178,
pro-inflammatory cytokines in obesity. Although absolute 181]. On the other hand, it could be hypothesised that acute
LDL concentration is often ‘normal’ in obesity, relationships phase ‘hyper-responsiveness’, due to increased sensitivity of
between insulin resistance, a component of the Metabolic the TLR to exogenous ligands such as LPS, and, eventually,
Syndrome, and LDL cholesterol level have been reported to some endogenous ligands, results in excessive cytokine
[32]. Furthermore, qualitative changes in LDL size and and chemokine production. Indeed, it has been suggested
Inflammation, Insulin Resistance and Type 2 Diabetes Current Diabetes Reviews, 2006, Vol. 2, No. 2 205

Fig. (2). Acute phase ‘hyper-responsiveness’ hypothesis. Increased levels of a broad range of inflammatory markers in obese individuals,
typified by C-reactive protein (CRP), are a function of obesity-driven cytokine production due to genetically determined immune hyper-
responsiveness. It is important to note that the amount of body fat, as well as its distribution, are also under significant genetic control. CRP:
C-reactive protein; hsp60: heat shock protein 60; LPS: lipopolysaccharide; mmLDL: minimally modified low-density lipoprotein; TLR: toll-
like receptor.

that the normally protective process of binding of LPS to cytokine production in individuals with the Metabolic
lipoproteins, which buffers its toxicity, is ‘overcome’ by Syndrome, the closely related phenotype, insulin resistance,
chronic stimulation, making LDL immunogenic and toxic to is likely to play an important mediating role in obesity.
endothelial cells [86]. Whilst hyper-responsiveness to Insulin has been reported to have anti-inflammatory
bacterial infection, together with an enhanced ability to store properties, including suppression of cytokine-mediated
fat, may have conferred a ‘survival advantage’ over many acute-phase protein gene expression [184] and reduction in
centuries, it could be expected to be potentially detrimental intranuclear factor κB, reactive oxygen species, plasma
in a modern, obese, sedentary population, particularly in soluble intercellular adhesion molecule-1, monocyte
relation to endothelial function and cardiovascular risk. Very chemoattractant protein-1 and plasminogen activator
recent evidence suggests that the immune response to LPS is inhibitor-1 [185]. This has led to the notion that insulin may
largely genetically determined, with heritability estimates of have ‘anti-inflammatory’ properties [86, 186, 187] and hence
0.53 for TNF-α, 0.57 for interleukin-6 and 0.86 for that insulin resistance increases levels of inflammatory
interleukin-1β [182]. Using factor analysis, it has been markers by attenuating this effect [165]. We therefore
shown that obesity, albeit estimated by anthropometric suggest that increased levels of CRP are likely to be a
estimates, clusters with inflammatory markers [183], consequence, and not necessarily the cause, of insulin
suggesting that obesity and circulating levels of resistance. Consistent with previous suggestions [188], these
inflammatory markers may be mechanistically linked and results increase the likelihood that circulating CRP levels are
influenced by common genetic factors. In other words, a balance between adipose tissue-derived interleukin-6-
increased levels of a broad range of inflammatory markers in mediated up-regulation of hepatic CRP production and the
obese individuals may be a function of genetically ‘anti-inflammatory’ or suppressive effect of insulin on the
determined immune hyper-responsiveness, stimulated by any liver, which is impaired in insulin-resistant subjects.
of a variety of putative ligands related to increased adiposity,
This hypothesis is illustrated in Fig. (3). In scenario (a),
itself under significant genetic control. This hypothesis is
in non-obese insulin-sensitive subjects, there is minimal
illustrated in Fig. (2).
stimulation of hepatic CRP production by interleukin-6 due
to relatively low levels of body fat; hence circulating levels
Impaired Anti-Inflammatory Effect of Insulin
of CRP are in the low-normal range. In scenario (b), in non-
As discussed, although the evidence presented in this obese insulin-resistant subjects, although insulin inhibition
review argues that body fat may be the primary stimulus for of interleukin-6-mediated hepatic CRP production is
206 Current Diabetes Reviews, 2006, Vol. 2, No. 2 Greenfield and Campbell

Fig. (3). Hypothesised interaction between stimulation of hepatic C-reactive protein (CRP) production by fat-derived interleukin-6 and
insulin-mediated inhibition of this process in insulin-sensitive and insulin-resistant obese and non-obese subjects. The thickness of the arrows
relate to the quantity of interleukin-6 (IL-6) or CRP secreted from fat and the liver respectively. The width of the T-bar indicates the degree of
inhibition by insulin of hepatic CRP production. Copyright © 2004 American Diabetes Association. Reprinted from [9] with permission from
the American Diabetes Association.

impaired due to insulin resistance, CRP levels remain in the may be a potential unifying pathomechanism linking insulin
low-normal range because of minimal stimulation of CRP resistance with increased levels of inflammatory markers,
secretion by interleukin-6. In scenario (c), in obese insulin- causing lipid accumulation in muscle and being one of the
sensitive subjects, the tendency for CRP levels to be up- possible precedents of hsp60-induced TLR-4-mediated
regulated in proportion to body fatness is somewhat cytokine release.
attenuated by insulin, and hence circulating levels are
relatively normal. In scenario (d), the same positive stimulus CONCLUDING REMARKS
to CRP secretion exists; however, because inhibition of CRP
production by insulin is impaired, CRP levels are in the In conclusion, there is a significant body of evidence to
high-normal range. suggest that the relationship between inflammatory markers
and both insulin resistance and the development of type 2
Mitochondrial Dysfunction diabetes is mediated by increased adiposity. Although
inflammation has been purported to induce insulin
It has been argued that insulin resistance is more resistance, contrary evidence suggests that in the absence of
important than insulin deficiency in mediating the acute increased adiposity, insulin resistance per se is not
phase response [189]. As discussed earlier, it has been associated with systemic elevation of CRP and reduced
postulated that muscle triglyceride accumulation and complement protein levels. As insulin has anti-inflammatory
impaired mitochondrial activity accompany insulin properties, it is possible that levels of inflammatory markers
resistance early in its development in genetically predisposed can be mildly increased in insulin resistance as a
subjects [29]. It is interesting to note that heat shock protein consequence of impaired suppression of cytokine-stimulated
60 (hsp60), a mitochondrial ‘housekeeping’ protein, is hepatic CRP production. Recent evidence that TLRs are
released into the extracellular space by impaired possible mediators of the inflammatory response and that
mitochondrial metabolism [190]. Hsp60 is an endogenous macrophages are responsible for a significant proportion of
ligand of TLR-4, which activates proinflammatory gene cytokine production in adipose tissue have provided
targets [179] (Fig. 2). Therefore, mitochondrial dysfunction biologically plausible mechanisms for these associations.
Inflammation, Insulin Resistance and Type 2 Diabetes Current Diabetes Reviews, 2006, Vol. 2, No. 2 207

Given the evidence presented in this review, it is imperative [22] American Diabetes Association National Institute of Diabetes and
that future studies both consider and control for the Digestive Kidney Diseases. Prevention or delay of type 2 diabetes.
Diabetes Care 2004; 27:S47-54.
confounding effect of body fat when formulating [23] Lyssenko V, Almgren P, Anevski D, et al. Predictors of and
mechanistic hypotheses regarding inflammatory markers and longitudinal changes in insulin sensitivity and secretion preceding
metabolic disease. Failure to properly adjust for adiposity, in onset of type 2 diabetes. Diabetes 2005; 54:166-74.
part a consequence of the use of indirect estimates of body [24] Kraegen EW, Cooney GJ. The role of free fatty acids in muscle
insulin resistance. Diabetes Annual 1999; 12:141-59.
fat in some studies [191, 192], must limit the validity of their [25] Hegarty BD, Furler SM, Ye J, Cooney GJ, Kraegen EW. The role
findings. of intramuscular lipid in insulin resistance. Acta Physiol Scand
2003; 178:373-83.
REFERENCES [26] Schmitz-Peiffer C. Signalling aspects of insulin resistance in
skeletal muscle: mechanisms induced by lipid oversupply. Cell
[1] Chisholm DJ, Campbell LV, Kraegen EW. Pathogenesis of the Signal 2000; 12:583-94.
insulin resistance syndrome (syndrome X). Clin Exp Pharmacol [27] Lewis GF, Carpentier A, Adeli K, Giacca A. Disordered fat storage
Physiol 1997; 24:782-4. and mobilization in the pathogenesis of insulin resistance and type
[2] DeFronzo RA, Ferrannini E. Insulin resistance. A multifaceted 2 diabetes. Endocr Rev 2002; 23:201-29.
syndrome responsible for NIDDM, obesity, hypertension, [28] Arner P. Insulin resistance in type 2 diabetes: role of fatty acids.
dyslipidemia, and atherosclerotic cardiovascular disease. Diabetes Diabetes Metab Res Rev 2002; 18 Suppl 2:S5-9.
Care 1991; 14:173-94. [29] Petersen KF, Dufour S, Befroy D, Garcia R, Shulman GI. Impaired
[3] Kaplan NM. The deadly quartet. Upper-body obesity, glucose mitochondrial activity in the insulin-resistant offspring of patients
intolerance, hypertriglyceridemia, and hypertension. Arch Intern with type 2 diabetes. N Engl J Med 2004; 350:664-71.
Med 1989; 149:1514-20. [30] Carey DG, Jenkins AB, Campbell LV, Freund J, Chisholm DJ.
[4] Grundy SM, Brewer HB Jr, Cleeman JI, Smith SC Jr, Lenfant C. Abdominal fat and insulin resistance in normal and overweight
Definition of metabolic syndrome: Report of the National Heart, women: Direct measurements reveal a strong relationship in
Lung, and Blood Institute/American Heart Association conference subjects at both low and high risk of NIDDM. Diabetes 1996;
on scientific issues related to definition. Circulation 2004; 109:433- 45:633-8.
8. [31] Greenfield JR, Samaras K, Chisholm DJ. Insulin resistance, intra-
[5] Grundy SM. Obesity, metabolic syndrome, and cardiovascular abdominal fat, cardiovascular risk factors, and androgens in healthy
disease. J Clin Endocrinol Metab 2004; 89:2595-600. young women with type 1 diabetes mellitus. J Clin Endocrinol
[6] Kahn R, Buse J, Ferrannini E, Stern M. The metabolic syndrome: Metab 2002; 87:1036-40.
time for a critical appraisal. Joint statement from the American [32] Poynten AM, Gan SK, Kriketos AD, Campbell LV, Chisholm DJ.
Diabetes Association and the European Association for the Study Circulating fatty acids, non-High Density Lipoprotein cholesterol,
of Diabetes. Diabetologia 2005; 48:1684-99. and insulin-infused fat oxidation acutely influence whole-body
[7] Stern MP. Diabetes and cardiovascular disease. The "common soil" insulin sensitivity in nondiabetic men. J Clin Endocrinol Metab
hypothesis. Diabetes 1995; 44:369-74. 2004; 90:1035-40.
[8] Pradhan AD, Ridker PM. Do atherosclerosis and type 2 diabetes [33] Gan SK, Kriketos AD, Poynten AM, et al. Insulin action, regional
share a common inflammatory basis? Eur Heart J 2002; 23:831-4. fat, and myocyte lipid: altered relationships with increased
[9] Kriketos AD, Greenfield JR, Peake PW, et al. Inflammation, adiposity. Obes Res 2003; 11:1295-305.
insulin resistance, and adiposity. A study of first-degree relatives of [34] Kriketos AD, Furler SM, Gan SK, Poynten AM, Chisholm DJ,
type 2 diabetic subjects. Diabetes Care 2004; 27:2033-40. Campbell LV. Multiple indexes of lipid availability are
[10] Ross R. Atherosclerosis is an inflammatory disease. Am Heart J independently related to whole body insulin action in healthy
1999; 138:S419-20. humans. J Clin Endocrinol Metab 2003; 88:793-8.
[11] Danesh J, Wheeler JG, Hirschfield GM, et al. C-reactive protein [35] Rebuffé-Scrive M, Enk L, Crona N, et al. Fat cell metabolism in
and other circulating markers of inflammation in the prediction of different regions in women. Effect of menstrual cycle, pregnancy,
coronary heart disease. N Engl J Med 2004; 350:1387-97. and lactation. J Clin Invest 1985; 75:1973-6.
[12] Jialal I, Devaraj S, Venugopal SK. C-reactive protein: risk marker [36] Rebuffé-Scrive M, Lonnroth P, Marin P, Wesslau C, Bjorntorp P,
or mediator in atherothrombosis? Hypertension 2004; 44:6-11. Smith U. Regional adipose tissue metabolism in men and
[13] Yeh ETH, Willerson JT. Coming of age of C-reactive protein. postmenopausal women. Int J Obes 1987; 11:347-55.
Using inflammation markers in cardiology. Circulation 2003; [37] Zierath JR, Livingston JN, Thorne A, et al. Regional difference in
107:370-2. insulin inhibition of non-esterified fatty acid release from human
[14] Pai JK, Pischon T, Ma J, et al. Inflammatory markers and the risk adipocytes: relation to insulin receptor phosphorylation and
of coronary heart disease in men and women. N Engl J Med 2004; intracellular signalling through the insulin receptor substrate-1
351:2599-610. pathway. Diabetologia 1998; 41:1343-54.
[15] Pearson TA, Mensah GA, Alexander RW, et al. Markers of [38] Ruderman N, Chisholm D, Pi-Sunyer X, Schneider S. The
inflammation and cardiovascular disease: application to clinical metabolically obese, normal-weight individual revisited. Diabetes
and public health practice: A statement for healthcare professionals 1998; 47:699-713.
from the Centers for Disease Control and Prevention and the [39] Roden M, Price TB, Perseghin G, et al. Mechanism of free fatty
American Heart Association. Circulation 2003; 107:499-511. acid-induced insulin resistance in humans. J Clin Invest 1996;
[16] Haffner SM. Insulin resistance, inflammation, and the prediabetic 97:2859-65.
state. Am J Cardiol 2003; 92 (suppl):18J-26J. [40] Chalkley SM, Hettiarachchi M, Chisholm DJ, Kraegen EW. Five-
[17] Pepys MB, Hirschfield GM. C-reactive protein: a critical update. J hour fatty acid elevation increases muscle lipids and impairs
Clin Invest 2003; 111:1805-12. glycogen synthesis in the rat. Metabolism 1998; 47:1121-6.
[18] King H, Aubert RE, Herman WH. Global burden of diabetes, 1995- [41] Campbell LV, Carey DG, Chisholm DJ. Measurement of central
2025: prevalence, numerical estimates, and projections. Diabetes adiposity. A bet each way? Diabetes Care 1996; 19:1033-4.
Care 1998; 21:1414-31. [42] Pickup JC. Inflammation and activated innate immunity in the
[19] Flegal KM, Carroll MD, Kuczmarski RJ, Johnson CL. Overweight pathogenesis of type 2 diabetes. Diabetes Care 2004; 27:813-23.
and obesity in the United States: prevalence and trends, 1960-1994. [43] Pickup JC, Crook MA. Is type II diabetes mellitus a disease of the
Int J Obes Relat Metab Disord 1998; 22:39-47. innate immune system? Diabetologia 1998; 41:1241-8.
[20] Cameron AJ, Welborn TA, Zimmet PZ, et al. Overweight and [44] Lee YH, Pratley RE. The evolving role of inflammation in obesity
obesity in Australia: the 1999-2000 Australian Diabetes, Obesity and the metabolic syndrome. Curr Diab Rep 2005; 5:70-5.
and Lifestyle Study (AusDiab). Med J Aust 2003; 178:427-32. [45] Nesto R. C-reactive protein, its role in inflammation, Type 2
[21] Prentice AM, Jebb SA. Obesity in Britain: gluttony or sloth? BMJ diabetes and cardiovascular disease, and the effects of insulin-
1995; 311:437-9. sensitizing treatment with thiazolidinediones. Diabet Med 2004;
21:810-7.
208 Current Diabetes Reviews, 2006, Vol. 2, No. 2 Greenfield and Campbell

[46] Festa A, D'Agostino R Jr, Howard G, Mykkanen L, Tracy RP, [67] de Maat MP, Bladbjerg EM, Hjelmborg JB, Bathum L, Jespersen J,
Haffner SM. Chronic subclinical inflammation as part of the insulin Christensen K. Genetic influence on inflammation variables in the
resistance syndrome: the Insulin Resistance Atherosclerosis Study elderly. Arterioscler Thromb Vasc Biol 2004; 24:2168-73.
(IRAS). Circulation 2000; 102:42-7. [68] Pankow JS, Folsom AR, Cushman M, et al. Familial and genetic
[47] Duncan BB, Schmidt MI, Offenbacher S, Wu KK, Savage PJ, determinants of systemic markers of inflammation: the NHLBI
Heiss G. Factor VIII and other hemostasis variables are related to family heart study. Atherosclerosis 2001; 154:681-9.
incident diabetes in adults. The Atherosclerosis Risk in [69] Vickers MA, Green FR, Terry C, et al. Genotype at a promoter
Communities (ARIC) Study. Diabetes Care 1999; 22:767-72. polymorphism of the interleukin-6 gene is associated with baseline
[48] Schmidt MI, Duncan BB, Sharrett AR, et al. Markers of levels of C-reactive protein. Cardiovasc Res 2002; 53:1029-34.
inflammation and prediction of diabetes mellitus in adults [70] Lange LA, Bowden DW. Familial aggregation of C-reactive
(Atherosclerosis Risk in Communities study): a cohort study. protein in type 2 diabetes. Circulation 2003; 107:22.
Lancet 1999; 353:1649-52. [71] Austin MA, Zhang C, Humphries SE, et al. Heritability of C-
[49] Pradhan AD, Manson JE, Rifai N, Buring JE, Ridker PM. C- reactive protein and association with apolipoprotein E genotypes in
reactive protein, interleukin 6, and risk of developing type 2 Japanese Americans. Ann Hum Genet 2004; 68:179-88.
diabetes mellitus. JAMA 2001; 286:327-34. [72] Moller DE. Potential role of TNF-alpha in the pathogenesis of
[50] Barzilay JI, Abraham L, Heckbert SR, et al. The relation of insulin resistance and type 2 diabetes. Trends Endocrinol Metab
markers of inflammation to the development of glucose disorders 2000; 11:212-7.
in the elderly: the Cardiovascular Health Study. Diabetes 2001; [73] Guerre-Millo M. Adipose tissue and adipokines: for better or
50:2384-9. worse. Diabetes Metab 2004; 30:13-9.
[51] Festa A, D'Agostino R Jr, Tracy RP, Haffner SM. Elevated levels [74] Pepys MB, Baltz M, Gomer K, Davies AJ, Doenhoff M. Serum
of acute-phase proteins and plasminogen activator inhibitor-1 amyloid P-component is an acute-phase reactant in the mouse.
predict the development of type 2 diabetes: the Insulin Resistance Nature 1979; 278:259-61.
Athersclerosis Study. Diabetes 2002; 51:1131-7. [75] Rajala MW, Scherer PE. Minireview: The adipocyte-at the
[52] Vozarova B, Weyer C, Lindsay RS, Pratley RE, Bogardus C, crossroads of energy homeostasis, inflammation, and
Tataranni PA. High white blood cell count is associated with a atherosclerosis. Endocrinology 2003; 144:3765-73.
worsening of insulin sensitivity and predicts the development of [76] Beltowski J. Adiponectin and resistin - new hormones of white
type 2 diabetes. Diabetes 2002; 51:455-61. adipose tissue. Med Sci Monit 2003; 9:RA55-61.
[53] Freeman DJ, Norrie J, Caslake MJ, et al. C-reactive protein is an [77] Fasshauer M, Klein J, Lossner U, Paschke R. Interleukin (IL)-6
independent predictor of risk for the development of diabetes in the mRNA expression is stimulated by insulin, isoproterenol, tumour
West Scotland Coronary Prevention Study. Diabetes 2002; necrosis factor alpha, growth hormone, and IL-6 in 3T3-L1
51:1596-600. adipocytes. Horm Metab Res 2003; 35:147-52.
[54] Han TS, Sattar N, Williams K, Gonzalez-Villalpando C, Lean ME, [78] Fasshauer M, Klein J, Neumann S, Eszlinger M, Paschke R.
Haffner SM. Prospective study of C-reactive protein in relation to Hormonal regulation of adiponectin gene expression in 3T3-L1
the development of diabetes and metabolic syndrome in the Mexico adipocytes. Biochem Biophys Res Commun 2002; 290:1084-9.
City Diabetes Study. Diabetes Care 2002; 25:2016-21. [79] Lang CH, Dobrescu C, Bagby GJ. Tumor necrosis factor impairs
[55] Duncan BB, Schmidt MI, Pankow JS, et al. Low-grade systemic insulin action on peripheral glucose disposal and hepatic glucose
inflammation and the development of type 2 diabetes. The output. Endocrinology 1992; 130:43-52.
Atherosclerosis Risk in Communities Study. Diabetes 2003; [80] Mohamed-Ali V, Goodrick S, Rawesh A, et al. Subcutaneous
52:1799-805. adipose tissue releases interleukin-6, but not tumor necrosis factor-
[56] Thorand B, Lowel H, Schneider A, et al. C-reactive protein as a alpha, in vivo. J Clin Endocrinol Metab 1997; 82:4196-200.
predictor for incident diabetes mellitus among middle-aged men. [81] Carey AL, Bruce CR, Sacchetti M, et al. Interleukin-6 and tumor
Results from the MONICA Ausburg Cohort Study. Arch Intern necrosis factor-alpha are not increased in patients with Type 2
Med 2003; 163:93-9. diabetes: evidence that plasma interleukin-6 is related to fat mass
[57] Nakanishi S, Yamane K, Kamel N, Okubo M, Kohno N. Elevated and not insulin responsiveness. Diabetologia 2004; 47:1029-37.
C-reactive protein is a risk factor for the development of type 2 [82] Ofei F, Hurel S, Newkirk J, Sopwith M, Taylor R. Effects of an
diabetes in Japanese Americans. Diabetes Care 2003; 26:2754-7. engineered human anti-TNF-alpha antibody (CDP571) on insulin
[58] Snijder BM, Dekker JM, Visser M, et al. Prospective relation of C- sensitivity and glycemic control in patients with NIDDM. Diabetes
reactive protein with type 2 diabetes. Diabetes Care 2003; 26:1656- 1996; 45:881-5.
7. [83] Dominguez H, Storgaard H, Rask-Madsen C, et al. Metabolic and
[59] Krakoff J, Funahashi T, Stehouwer CDA, et al. Inflammatory Vascular Effects of Tumor Necrosis Factor-alpha Blockade with
markers, adiponectin, and risk of type 2 diabetes in the Pima Etanercept in Obese Patients with Type 2 Diabetes. J Vasc Res
Indian. Diabetes Care 2003; 26:1745-51. 2005; 42:517-25.
[60] Spranger J, Kroke A, Mohlig M, et al. Inflammatory cytokines and [84] Paquot N, Castillo MJ, Lefebvre PJ, Scheen AJ. No increased
the risk to develop type 2 diabetes. Results of the Prospective insulin sensitivity after a single intravenous administration of a
Population based European Prospective Investigation into Cancer recombinant human tumor necrosis factor receptor: Fc fusion
and Nutrition (EPIC)-Potsdam Study. Diabetes 2003; 52:812-7. protein in obese insulin-resistant patients. J Clin Endocrinol Metab
[61] Tan KC, Wat NM, Tam SC, Janus ED, Lam TH, Lam KS. C- 2000; 85:1316-9.
reactive protein predicts the deterioration of glycemia in chinese [85] Febbraio MA, Pedersen BK. Muscle-derived interleukin-6:
subjects with impaired glucose tolerance. Diabetes Care 2003; mechanisms for activation and possible biological roles. FASEB J
26:2323-8. 2002; 16:1335-47.
[62] Laaksonen DE, Niskanen L, Nyyssonen K, et al. C-reactive protein [86] Fernandez-Real JM, Ricart W. Insulin resistance and chronic
and the development of the metabolic syndrome and diabetes in cardiovascular inflammatory syndrome. Endocr Rev 2003; 24:278-
middle-aged men. Diabetologia 2004; 47:1403-10. 301.
[63] Hu FB, Meigs JB, Li TY, Rifai N, Manson JE. Inflammatory [87] Senn JJ, Klover PJ, Nowak IA, et al. Suppressor of cytokine
markers and risk of developing type 2 diabetes in women. Diabetes signaling-3 (SOCS-3), a potential mediator of interleukin-6-
2004; 53:693-700. dependent insulin resistance in hepatocytes. J Biol Chem 2003;
[64] Shine B, de Beer FC, Pepys MB. Solid phase radioimmunoassays 278:13740-6.
for human C-reactive protein. Clin Chim Acta 1981; 117:13-23. [88] Stouthard JM, Oude Elferink RP, Sauerwein HP. Interleukin-6
[65] MacGregor AJ, Gallimore JR, Spector TD, Pepys MB. Genetic enhances glucose transport in 3T3-L1 adipocytes. Biochem
effects on baseline values of C-reactive protein and Serum Biophys Res Commun 1996; 220:241-5.
Amyloid A protein. A comparison of monozygotic and dizygotic [89] DiCosmo BF, Picarella D, Flavell RA. Local production of human
twins. Clin Chem 2004; 50:130-4. IL-6 promotes insulitis but retards the onset of insulin-dependent
[66] Retterstol L, Eikvar L, Berg K. A twin study of C-Reactive Protein diabetes mellitus in non-obese diabetic mice. Int Immunol 1994;
compared to other risk factors for coronary heart disease. 6:1829-37.
Atherosclerosis 2003; 169:279-82. [90] Wallenius V, Wallenius K, Ahren B, et al. Interleukin-6-deficient
mice develop mature-onset obesity. Nat Med 2002; 8:75-9.
Inflammation, Insulin Resistance and Type 2 Diabetes Current Diabetes Reviews, 2006, Vol. 2, No. 2 209

[91] Klover PJ, Clementi AH, Mooney RA. Interleukin-6 depletion [113] Yamauchi T, Kamon J, Ito Y, et al. Cloning of adiponectin
selectively improves hepatic insulin action in obesity. receptors that mediate antidiabetic metabolic effects. Nature 2003;
Endocrinology 2005; 146:3417-27. 423:762-9.
[92] Tsigos C, Papanicolaou DA, Kyrou I, Defensor R, Mitsiadis CS, [114] Yamauchi T, Kamon J, Waki H, et al. The fat-derived hormone
Chrousos GP. Dose-dependent effects of recombinant human adiponectin reverses insulin resistance associated with both
interleukin-6 on glucose regulation. J Clin Endocrinol Metab 1997; lipoatrophy and obesity. Nat Med 2001; 7:941-6.
82:4167-70. [115] Berg AH, Combs TP, Du X, Brownlee M, Scherer PE. The
[93] Stouthard JM, Romijn JA, Van der Poll T, et al. Endocrinologic adipocyte-secreted protein Acrp30 enhances hepatic insulin action.
and metabolic effects of interleukin-6 in humans. Am J Physiol Nat Med 2001; 7:947-53.
1995; 268:E813-9. [116] Maeda N, Shimomura I, Kishida K, et al. Diet-induced insulin
[94] Steensberg A, Fischer CP, Sacchetti M, et al. Acute interleukin-6 resistance in mice lacking adiponectin/ACRP30. Nat Med 2002;
administration does not impair muscle glucose uptake or whole- 8:731-7.
body glucose disposal in healthy humans. J Physiol 2003; 548:631- [117] Chandran M, Phillips SA, Ciaraldi T, Henry RR. Adiponectin:
8. more than just another fat cell hormone? Diabetes Care 2003;
[95] Vozarova B, Weyer C, Hanson K, Tataranni PA, Bogardus C, 26:2442-50.
Pratley RE. Circulating interleukin-6 in relation to adiposity, [118] Jenkins AB, Campbell LV. Does relative leptinemia predict weight
insulin action, and insulin secretion. Obes Res 2001; 9:414-7. gain in humans? Obes Res 2003; 11:373-4.
[96] Bajaj M, Suraamornkul S, Kashyap S, Cusi K, Mandarino L, [119] Diamant M, Lamb HJ, van de Ree MA, et al. The association
DeFronzo RA. Sustained reduction in plasma free fatty acid between abdominal visceral fat and carotid stiffness is mediated by
concentration improves insulin action without altering plasma circulating inflammatory markers in uncomplicated type 2 diabetes.
adipocytokine levels in subjects with strong family history of type J Clin Endocrinol Metab 2005; 'in press'.
2 diabetes. J Clin Endocrinol Metab 2004; 89:4649-55. [120] Rössner S, Bo WJ, Hiltbrandt E, et al. Adipose tissue
[97] Havel PJ. Update on adipocyte hormones: regulation of energy determinations in cadavers--a comparison between cross- sectional
balance and carbohydrate/lipid metabolism. Diabetes 2004; 53 planimetry and computed tomography. Int J Obes 1990; 14:893-
Suppl 1:S143-51. 902.
[98] Kershaw EE, Flier JS. Adipose tissue as an endocrine organ. J Clin [121] Abate N, Burns D, Peshock RM, Garg A, Grundy SM. Estimation
Endocrinol Metab 2004; 89:2548-56. of adipose tissue mass by magnetic resonance imaging: validation
[99] Trujillo ME, Scherer PE. Adiponectin - journey from an adipocyte against dissection in human cadavers. J Lipid Res 1994; 35:1490-6.
secretory protein to biomarker of the metabolic syndrome. J Intern [122] Greenfield JR, Samaras K, Chisholm DJ, Campbell LV. Regional
Med 2005; 257:167-75. intra-subject variability in abdominal adiposity limits usefulness of
[100] Ouchi N, Kihara S, Funahashi T, et al. Reciprocal association of C- computed tomography. Obes Res 2002; 10:260-5.
reactive protein with adiponectin in blood stream and adipose [123] Kelley DE, Thaete FL, Troost F, Huwe T, Goodpaster BH.
tissue. Circulation 2003; 107:671-4. Subdivisions of subcutaneous abdominal adipose tissue and insulin
[101] Kriketos AD, Gan SK, Poynten AM, Furler SM, Chisholm DJ, resistance. Am J Physiol Endocrinol Metab 2000; 278:E941-48.
Campbell LV. Exercise increases adiponectin levels and insulin [124] Abate N, Garg A, Peshock RM, Stray-Gundersen J, Grundy SM.
sensitivity in humans. Diabetes Care 2004; 27:629-30. Relationships of generalized and regional adiposity to insulin
[102] Tschritter O, Fritsche A, Thamer C, et al. Plasma adiponectin sensitivity in men. J Clin Invest 1995; 96:88-98.
concentrations predict insulin sensitivity of both glucose and lipid [125] Abate N, Garg A, Peshock RM, Stray-Gundersen J, Adams-Huet B,
metabolism. Diabetes 2003; 52:239-43. Grundy SM. Relationship of generalized and regional adiposity to
[103] Bacha F, Saad R, Gungor N, Arslanian SA. Adiponectin in youth: insulin sensitivity in men with NIDDM. Diabetes 1996; 45:1684-
relationship to visceral adiposity, insulin sensitivity, and beta-cell 93.
function. Diabetes Care 2004; 27:547-52. [126] Garg A. Regional adiposity and insulin resistance. J Clin
[104] Motoshima H, Wu X, Sinha MK, et al. Differential regulation of Endocrinol Metab 2004; 89:4206-10.
adiponectin secretion from cultured human omental and [127] Davey Smith G, Phillips AN. Confounding in epidemiological
subcutaneous adipocytes: effects of insulin and rosiglitazone. J Clin studies: why "independent" effects may not be all they seem. BMJ
Endocrinol Metab 2002; 87:5662-7. 1992; 305:757-9.
[105] Pajvani UB, Du X, Combs TP, et al. Structure-function studies of [128] Fried SK, Bunkin DA, Greenberg AS. Omental and subcutaneous
the adipocyte-secreted hormone Acrp30/adiponectin. Implications adipose tissues of obese subjects release interleukin-6: depot
fpr metabolic regulation and bioactivity. J Biol Chem 2003; difference and regulation by glucocorticoid. J Clin Endocrinol
278:9073-85. Metab 1998; 83:847-50.
[106] Pajvani UB, Hawkins M, Combs TP, et al. Complex distribution, [129] Wellen KE, Hotamisligil GS. Obesity-induced inflammatory
not absolute amount of adiponectin, correlates with changes in adipose tissue. J Clin Invest 2003; 112:1785-8.
thiazolidinedione-mediated improvement in insulin sensitivity. J [130] Greenfield JR, Samaras K, Jenkins AB, et al. Obesity is an
Biol Chem 2004; 279:12152-62. important determinant of baseline C-reactive protein concentration
[107] Hotta K, Funahashi T, Bodkin NL, et al. Circulating concentrations in monozygotic twins, independent of genetic influences.
of the adipocyte protein adiponectin are decreased in parallel with Circulation 2004; 109:3022-8.
reduced insulin sensitivity during the progression to type 2 diabetes [131] Samaras K, Spector TD, Nguyen TV, Baan K, Campbell LV, Kelly
in rhesus monkeys. Diabetes 2001; 50:1126-33. PJ. Independent genetic factors determine the amount and
[108] Spranger J, Kroke A, Mohlig M, et al. Adiponectin and protection distribution of fat in women after the menopause. J Clin Endocrinol
against type 2 diabetes mellitus. Lancet 2003; 361:226-8. Metab 1997; 82:781-5.
[109] Lindsay RS, Funahashi T, Hanson RL, et al. Adiponectin and [132] Samaras K, Nguyen TV, Jenkins AB, et al. Clustering of insulin
development of type 2 diabetes in the Pima Indian population. resistance, total and central abdominal fat: same genes or same
Lancet 2002; 360:57-8. environment? Twin Res 1999; 2:218-25.
[110] Vionnet N, Hani El H, Dupont S, et al. Genomewide search for [133] Carey DG, Nguyen TV, Campbell LV, Chisholm DJ, Kelly P.
type 2 diabetes-susceptibility genes in French whites: evidence for Genetic influences on central abdominal fat: a twin study. Int J
a novel susceptibility locus for early-onset diabetes on chromosome Obes Relat Metab Disord 1996; 20:722-6.
3q27-qter and independent replication of a type 2-diabetes locus on [134] Greenfield JR, Samaras K, Hayward CS, Chisholm DJ, Campbell
chromosome 1q21-q24. Am J Hum Genet 2000; 67:1470-80. LV. Beneficial postprandial effect of a small amount of alcohol on
[111] Hara K, Boutin P, Mori Y, et al. Genetic variation in the gene diabetes and cardiovascular risk factors: modification by insulin
encoding adiponectin is associated with an increased risk of type 2 resistance. J Clin Endocrinol Metab 2005; 90:661-72.
diabetes in the Japanese population. Diabetes 2002; 51:536-40. [135] Putz DM, Goldner WS, Bar RS, Haynes WG, Sivitz WI.
[112] de Courten BV, Hanson RL, Funahashi T, et al. Common Adiponectin and C-reactive protein in obesity, type 2 diabetes, and
polymorphisms in the adiponectin gene ACDC are not associated monodrug therapy. Metabolism 2004; 53:1454-61.
with diabetes in PIMA Indians. Diabetes 2005; 54:284-9. [136] Maachi M, Pieroni L, Bruckert E, et al. Systemic low-grade
inflammation is related to both circulating and adipose tissue
210 Current Diabetes Reviews, 2006, Vol. 2, No. 2 Greenfield and Campbell

TNFalpha, leptin and IL-6 levels in obese women. Int J Obes Relat [157] McLaughlin T, Abbasi F, Lamendola C, et al. Differentiation
Metab Disord 2004; 28:993-7. between obesity and insulin resistance in the association with C-
[137] Barinas-Mitchell E, Cushman M, Meilahn EN, Tracy RP, Kuller reactive protein. Circulation 2002; 106:2908-12.
LH. Serum levels of C-reactive protein are associated with obesity, [158] Heilbronn LK, Noakes M, Clifton PM. Energy restriction and
weight gain, and hormone replacement therapy in healthy weight loss on very-low-fat diets reduce C-reactive protein
postmenopausal women. Am J Epidemiol 2001; 153:1094-101. concentrations in obese, healthy women. Arterioscler Thromb Vasc
[138] Forouhi NG, Sattar N, McKeigue PM. Relation of C-reactive Biol 2001; 21:968-70.
protein to body fat distribution and features of the metabolic [159] Esposito K, Pontillo A, Di Palo C, et al. Effect of weight loss and
syndrome in Europeans and South Asians. Int J Obes Relat Metab lifestyle changes on vascular inflammatory markers in obese
Disord 2001; 25:1327-31. women: a randomized trial. JAMA 2003; 289:1799-804.
[139] Stauffer BL, Hoetzer GL, Smith DT, DeSouza CA. Plasma C- [160] Esposito K, Pontillo A, Giugliano F, et al. Association of low
reactive protein is not elevated in physically active postmenopausal interleukin-10 levels with the metabolic syndrome in obese women.
women taking hormone replacement therapy. J Appl Physiol 2004; J Clin Endocrinol Metab 2003; 88:1055-8.
96:143-8. [161] Dansinger ML, Gleason JA, Griffith JL, Selker HP, Schaefer EJ.
[140] Sites CK, Toth MJ, Cushman M, et al. Menopause-related Comparison of the Atkins, Ornish, Weight Watchers, and Zone
differences in inflammation markers and their relationship to body diets for weight loss and heart disease risk reduction: a randomized
fat distribution and insulin stimulated-glucose disposal. Fertil Steril trial. JAMA 2005; 293:43-53.
2002; 77:128-35. [162] Hanusch-Enserer U, Cauza E, Spak M, et al. Acute-phase response
[141] Weyer C, Yudkin JS, Stehouwer CDA, Schalkwijk CG, Pratley RE, and immunological markers in morbid obese patients and patients
Tataranni PA. Humoral markers of inflammation and endothelial following adjustable gastric banding. Int J Obes Relat Metab
dysfunction in relation to adiposity and in vivo insulin action in Disord 2003; 27:355-61.
Pima Indians. Atherosclerosis 2002; 161:233-42. [163] Kopp HP, Kopp CW, Festa A, et al. Impact of weight loss on
[142] Bastard J-P, Jardel C, Delattre J, Hainque B, Bruckert E, Oberlin F. inflammatory proteins and their association with the insulin
Evidence for a link between adipose tissue interleukin-6 content resistance syndrome in morbidly obese patients. Arterioscler
and serum C-reactive protein concentrations in obese subjects. Thromb Vasc Biol 2003; 23:1042-7.
Circulation 1999; 99:2221-2. [164] Laimer M, Ebenbichler CF, Kaser S, et al. Markers of chronic
[143] Tchernof A, Nolan A, Sites CK, Ades PA, Poehlman ET. Weight inflammation and obesity: a prospective study on the reversibility
loss reduces C-reactive protein levels in obese postmenopausal of this association in middle-aged women undergoing weight loss
women. Circulation 2002; 105:564-9. by surgical intervention. Int J Obes Relat Metab Disord 2002;
[144] Bo M, Raspo S, Morra F, et al. Body fat and C-reactive protein 26:659-62.
levels in healthy non-obese men. Nutr Metab Cardiovasc Dis 2004; [165] Vazquez LA, Pazos F, Berrazueta JR, et al. Effects of changes in
14:66-72. body weight and insulin resistance on inflammation and endothelial
[145] Axelsson J, Rashid Qureshi A, Suliman ME, et al. Truncal fat mass function in morbid obesity after bariatric surgery. J Clin Endocrinol
as a contributor to inflammation in end-stage renal disease. Am J Metab 2005; 90:316-22.
Clin Nutr 2004; 80:1222-9. [166] Seshadri P, Iqbal N, Stern L, et al. A randomized study comparing
[146] Manns PJ, Williams DP, Snow CM, Wander R. Physical activity, the effects of a low-carbohydrate diet and a conventional diet on
body fat, and serum C-reactive protein in postmenopausal women lipoprotein subfractions and C-reactive protein levels in patients
with and without hormone replacement. Am J Hum Biol 2003; with severe obesity. Am J Med 2004; 117:398-405.
15:91-100. [167] O'Brien KD, Brehm BJ, Seeley RJ, et al. Diet-induced weight loss
[147] Saijo Y, Kiyota N, Kawasaki Y, et al. Relationship between C- is associated with decreases in plasma serum amyloid a and C-
reactive protein and visceral adipose tissue in healthy Japanese reactive protein independent of dietary macronutrient composition
subjects. Diabetes Obes Metab 2004; 6:249-58. in obese subjects. J Clin Endocrinol Metab 2005; 90:2244-9.
[148] Lemieux I, Pascot A, Prud'homme D, et al. Elevated C-reactive [168] Ryan AS, Nicklas BJ. Reductions in plasma cytokine levels with
protein: another component of the atherothrombotic profile of weight loss improve insulin sensitivity in overweight and obese
abdominal obesity. Arterioscler Thromb Vasc Biol 2001; 21:961-7. postmenopausal women. Diabetes Care 2004; 27:1699-705.
[149] Weiss R, Dziura J, Burgert TS, et al. Obesity and the metabolic [169] Romano M, Guagnano MT, Pacini G, et al. Association of
syndrome in children and adolescents. N Engl J Med 2004; inflammation markers with impaired insulin sensitivity and
350:2362-74. coagulative activation in obese healthy women. J Clin Endocrinol
[150] Vikram NK, Misra A, Pandey Rm R, Dwivedi M, Luthra K. Metab 2003; 88:5321-6.
Adiponectin, insulin resistance, and C-reactive protein in [170] You T, Ryan AS, Nicklas BJ. The metabolic syndrome in obese
postpubertal Asian Indian adolescents. Metabolism 2004; 53:1336- postmenopausal women: relationship to body composition, visceral
41. fat, and inflammation. J Clin Endocrinol Metab 2004; 89:5517-22.
[151] Hak AE, Stehouwer CD, Bots ML, et al. Associations of C-reactive [171] Escobar-Morreale HF, Villuendas G, Botella-Carretero JI, Sancho
protein with measures of obesity, insulin resistance, and subclinical J, San Millan JL. Obesity, and not insulin resistance, is the major
atherosclerosis in healthy, middle-aged women. Arterioscler determinant of serum inflammatory cardiovascular risk markers in
Thromb Vasc Biol 1999; 19:1986-91. pre-menopausal women. Diabetologia 2003; 46:625-33.
[152] Marcell TJ, McAuley KA, Traustadottir T, Reaven PD. Exercise [172] Mohlig M, Spranger J, Osterhoff M, et al. The polycystic ovary
training is not associated with improved levels of C-reactive syndrome per se is not associated with increased chronic
protein or adiponectin. Metabolism 2005; 54:533-41. inflammation. Eur J Endocrinol 2004; 150:525-32.
[153] Behre CJ, Fagerberg B, Hulten LM, Hulthe J. The reciprocal [173] Talbott EO, Zborowski JV, Boudreaux MY, McHugh-Pemu KP,
association of adipocytokines with insulin resistance and C-reactive Sutton-Tyrrell K, Guzick DS. The relationship between C-reactive
protein in clinically healthy men. Metabolism 2005; 54:439-44. protein and carotid intima-media wall thickness in middle-aged
[154] Haffner SM, D'Agostino RB Jr, Festa A, et al. Low insulin women with polycystic ovary syndrome. J Clin Endocrinol Metab
sensitivity (Si=0) in diabetic and nondiabetic subjects in the Insulin 2004; 89:6061-7.
Resistance Atherosclerosis Study. Diabetes Care 2003; 26:2796- [174] Volanakis JE. Complement activation by C-reactive protein
803. complexes. Ann N Y Acad Sci 1982; 389:235-50.
[155] Pannacciulli N, Cantatore FP, Minenna A, Bellacicco M, Giorgino [175] Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL,
R, De Pergola G. C-reactive protein is independently associated Ferrante AW Jr. Obesity is associated with macrophage
with total body fat, central fat, and insulin resistance in adult accumulation in adipose tissue. J Clin Invest 2003; 112:1796-808.
women. Int J Obes Relat Metab Disord 2001; 25:1416-20. [176] Xu H, Barnes GT, Yang Q, et al. Chronic inflammation in fat plays
[156] Tripathy D, Almgren P, Tuomi T, Groop L. Contribution of a crucial role in the development of obesity-related insulin
insulin-stimulated glucose uptake and basal hepatic insulin resistance. J Clin Invest 2003; 112:1821-30.
sensitivity to surrogate measures of insulin sensitivity. Diabetes [177] Miller YI, Chang MK, Binder CJ, Shaw PX, Witztum JL. Oxidized
Care 2004; 27:2204-10. low density lipoprotein and innate immune receptors. Curr Opin
Lipidol 2003; 14:437-45.
Inflammation, Insulin Resistance and Type 2 Diabetes Current Diabetes Reviews, 2006, Vol. 2, No. 2 211

[178] Kiechl S, Lorenz E, Reindl M, et al. Toll-like receptor 4 [186] Dandona P, Aljada A. A rational approach to pathogenesis and
polymorphisms and atherogenesis. N Engl J Med 2002; 347:185- treatment of type 2 diabetes mellitus, insulin resistance,
92. inflammation, and atherosclerosis. Am J Cardiol 2002;
[179] Michelsen KS, Doherty TM, Shah PK, Arditi M. TLR signaling: an 90(suppl):27G-33G.
emerging bridge from innate immunity to atherogenesis. J Immunol [187] Chaudhuri A, Janicke D, Wilson MF, et al. Anti-inflammatory and
2004; 173:5901-7. profibrinolytic effect of insulin in acute ST-segment-elevation
[180] Griffin BA. Lipoprotein atherogenicity: an overview of current myocardial infarction. Circulation 2004; 109:849-54.
mechanisms. Proc Nutr Soc 1999; 58:163-9. [188] Hak AE, Pols HA, Stehouwer CD, et al. Markers of inflammation
[181] Arbour NC, Lorenz E, Schutte BC, et al. TLR4 mutations are and cellular adhesion molecules in relation to insulin resistance in
associated with endotoxin hyporesponsiveness in humans. Nat nondiabetic elderly: the Rotterdam study. J Clin Endocrinol Metab
Genet 2000; 25:187-91. 2001; 86:4398-405.
[182] de Craen AJ, Posthuma D, Remarque EJ, van den Biggelaar AH, [189] Recasens M, Lopez-Bermejo A, Ricart W, Vendrell J, Casamitjana
Westendorp RG, Boomsma DI. Heritability estimates of innate R, Fernandez-Real JM. An inflammation score is better associated
immunity: an extended twin study. Genes Immun 2005. with basal than stimulated surrogate indexes of insulin resistance. J
[183] Austin MA, Edwards KL, McNeely MJ, et al. Heritability of Clin Endocrinol Metab 2005; 90:112-6.
multivariate factors of the metabolic syndrome in nondiabetic [190] Stöllberger C, Finsterer J. Atherosclerosis: infection-induced
Japanese Americans. Diabetes 2004; 53:1166-9. involvement of mitochondrial chaperonins. Lancet 2003; 362:1949-
[184] Campos SP, Baumann H. Insulin is a prominent modulator of the 50.
cytokine-stimulated expression of acute-phase plasma protein [191] Kaushal K, Heald AH, Siddals KW, et al. The impact of
genes. Mol Cell Biol 1992; 12:1789-92. abnormalities in IGF and inflammatory systems on the metabolic
[185] Dandona P, Aljada A, Mohanty P, et al. Insulin inhibits syndrome. Diabetes Care 2004; 27:2682-8.
intranuclear nuclear factor kappaB and stimulates IkappaB in [192] Schulze MB, Rimm EB, Li T, Rifai N, Stampfer MJ, Hu FB. C-
mononuclear cells in obese subjects: evidence for an anti- reactive protein and incident cardiovascular events among men
inflammatory effect? J Clin Endocrinol Metab 2001; 86:3257-65. with diabetes. Diabetes Care 2004; 27:889-94.

Received: 02 August, 2005 Revised: 30 September, 2005 Accepted: 14 October, 2005

You might also like