Hostname: page-component-8448b6f56d-wq2xx Total loading time: 0 Render date: 2024-04-19T10:52:30.072Z Has data issue: false hasContentIssue false

Cellular hypoxia and adipose tissue dysfunction in obesity

Symposium on ‘Frontiers in adipose tissue biology’

Published online by Cambridge University Press:  24 August 2009

I. Stuart Wood*
Affiliation:
Obesity Biology Research Unit, School of Clinical Sciences, University of Liverpool, Duncan Building, Liverpool L69 3GA, UK
Fátima Pérez de Heredia
Affiliation:
Obesity Biology Research Unit, School of Clinical Sciences, University of Liverpool, Duncan Building, Liverpool L69 3GA, UK
Bohan Wang
Affiliation:
Obesity Biology Research Unit, School of Clinical Sciences, University of Liverpool, Duncan Building, Liverpool L69 3GA, UK
Paul Trayhurn
Affiliation:
Obesity Biology Research Unit, School of Clinical Sciences, University of Liverpool, Duncan Building, Liverpool L69 3GA, UK
*
*Corresponding author: Dr I. Stuart Wood, fax +44 151 706 5802, email i.s.wood@liverpool.ac.uk
Rights & Permissions [Opens in a new window]

Abstract

Expansion of adipose tissue mass, the distinctive feature of obesity, is associated with low-grade inflammation. White adipose tissue secretes a diverse range of adipokines, a number of which are inflammatory mediators (such as TNFα, IL-1β, IL-6, monocyte chemoattractant protein 1). The production of inflammatory adipokines is increased with obesity and these adipokines have been implicated in the development of insulin resistance and the metabolic syndrome. However, the basis for the link between increased adiposity and inflammation is unclear. It has been proposed previously that hypoxia may occur in areas within adipose tissue in obesity as a result of adipocyte hypertrophy compromising effective O2 supply from the vasculature, thereby instigating an inflammatory response through recruitment of the transcription factor, hypoxic inducible factor-1. Studies in animal models (mutant mice, diet-induced obesity) and cell-culture systems (mouse and human adipocytes) have provided strong support for a role for hypoxia in modulating the production of several inflammation-related adipokines, including increased IL-6, leptin and macrophage migratory inhibition factor production together with reduced adiponectin synthesis. Increased glucose transport into adipocytes is also observed with low O2 tension, largely as a result of the up-regulation of GLUT-1 expression, indicating changes in cellular glucose metabolism. Hypoxia also induces inflammatory responses in macrophages and inhibits the differentiation of preadipocytes (while inducing the expression of leptin). Collectively, there is strong evidence to suggest that cellular hypoxia may be a key factor in adipocyte physiology and the underlying cause of adipose tissue dysfunction contributing to the adverse metabolic milieu associated with obesity.

Type
Research Article
Copyright
Copyright © The Authors 2009

Abbreviations:
HIF

hypoxic inducible factor

MCT

monocarboxylate transporters

The biology of adipose tissue has long surpassed the boundaries that defined it as merely a storage site for metabolic fuel, in addition to its role in thermal insulation and mechanical support. White adipose tissue has been recognised to release (and take up) NEFA and secrete the enzyme lipoprotein lipase. However, its role as an important endocrine organ was realised in 1994 following the identification of leptin, the protein product of the ob gene that is mutated in the genetically-obese (ob/ob) mouse(Reference Zhang, Proenca and Maffei1). This secreted protein was found to be synthesised and secreted by the adipocyte (the lipid-containing cell of adipose tissue) and thereby became one of the founder members of an extensive family of proteins, i.e. the adipokines, which are defined as proteins released from white adipocytes(Reference Trayhurn and Beattie2Reference Rosen and Spiegelman5).

Adipocytes secrete in excess of seventy-five adipokines, providing a means by which adipose tissue communicates with other organs, both centrally (brain) and peripherally (liver, skeletal muscle, etc.). The adipokines exhibit diverse functional roles, including in energy balance (e.g. leptin), insulin sensitivity and glucose metabolism (e.g. adiponectin), inflammation (e.g. TNFα), immunity (e.g. adipsin), lipid metabolism (e.g. cholesteryl ester transfer protein), blood pressure control (e.g. angiotensinogen), haemostasis (e.g. plasminogen-activating inhibitor-1) and angiogenesis (e.g. vascular epithelial growth factor)(Reference Trayhurn and Beattie2, Reference Trayhurn and Wood3). Maintaining an appropriate amount of adipose tissue is essential for optimal health. Outside ‘normal’ levels the resultant imbalance in adipokine production leads to adverse health consequences; a result of having too little (anorexia, cachexia), too much (obesity) or an inappropriate redistribution of adipose tissue (lipodystrophy).

The excessive expansion of adipose tissue mass is the distinctive feature of obesity. There has been an unprecedented rise in obesity, the incidence increasing >4-fold in the UK alone over the last three decades. The recently-published Foresight report from the UK Government has indicated that at the present rate of increase (including the rise in childhood obesity) between 50% and 60% of the population could be expected to be seen as obese by 2050(Reference McPherson, Marsh and Brown6). To compound the immediate problems of obesity per se (shortness of breath, back pain etc.), there is an increase in the risk of the co-morbidities associated with obesity, and more particularly of central obesity. These co-morbidities include insulin resistance (predisposing to type 2 diabetes mellitus), hypertension and CVD, which together form the cornerstones of the metabolic syndrome(Reference Phillips and Prins7). There also appears to be a greater risk of developing certain cancers, including breast and colon cancer(8).

Obesity is characterised by a state of chronic low-grade inflammation(Reference Engstrom, Hedblad and Stavenow9Reference Yudkin, Stehouwer and Emeis12). The elevated levels of circulatory pro-inflammatory cytokines (such as IL-6, TNFα) and acute-phase proteins (such as C-reactive protein) have been suggested as a causative link between obesity and its secondary complications, particularly insulin resistance. Importantly, some of these inflammatory mediators are also recognised as adipokines, thus consolidating an earlier observation that adipose tissue may have an inflammation-related role in obesity-linked insulin resistance(Reference Hotamisligil, Shargill and Spiegelman13). Adipose tissue is a heterogeneous organ at the cellular level, with adipocytes comprising ⩽60% of the total cell content. The remaining cell types consist of preadipocytes, resident macrophages, fibroblasts, histiocytes and endothelial cells. This non-adipocyte fraction is also recognised to secrete a number of inflammatory mediators(Reference Fain14), although the contribution from each cell type is difficult to assess accurately.

The majority of adipokines exhibit increased expression and secretion in the obese state; adiponectin is the notable exception, showing a decrease, which is consistent with its reported insulin-sensitising and anti-inflammatory roles(Reference Arita, Kihara and Ouchi15Reference Hotta, Funahashi and Arita17). The inflamed status of adipose tissue in obesity is further exacerbated by the recruitment and infiltration of bone marrow-derived macrophages into adipose tissue, possibly as a result of monocyte chemoattractant protein 1 release from adipocytes(Reference Weisberg, McCann and Desai18, Reference Xu, Barnes and Yang19). It is suggested that the recruitment of macrophages may represent a key event in obesity-derived metabolic disorders(Reference Bouloumie, Curat and Sengenes20). Leptin has also been suggested to play a chemotactic role in macrophage recruitment(Reference Curat, Miranville and Sengenes21). It is evident that adipose tissue may make a major contribution to the inflamed status associated with obesity. However, the sequence of events responsible for initiation of the inflammation response is less certain.

The hypoxia hypothesis

White adipose tissue is generally considered to be poorly vascularised(Reference Fleischmann, Kurz and Niedermayr22). In 2002 it was demonstrated that angiogenesis is required for adipose tissue expansion(Reference Rupnick, Panigrahy and Zhang23). The following year, a study using a mouse cell line suggested that a hypoxic environment may be responsible for promoting the expression of angiogenic genes (vascular epithelial growth factor), thus promoting the formation of new blood vessels during adipose tissue growth(Reference Lolmede, Durand de Saint Front and Galitzky24). Furthermore, studies from two independent groups demonstrated that the human leptin gene is activated by hypoxia(Reference Ambrosini, Nath and Sierra-Honigmann25Reference Grosfeld, Zilberfarb and Turban27). These earlier observations formed the basis of a hypothesis proposed in 2004 that hypoxia is the key initiator in adipokine dysregulation in obesity, thereby inducing an inflammation response within adipose tissue(Reference Trayhurn and Wood3). More precisely, localised hypoxia was suggested to develop in expanding adipocytes furthest removed from the blood supply. This proposal was enhanced by the observation that blood flow to white adipose tissue is not elevated in obese subjects(Reference Kabon, Nagele and Reddy28), nor in contrast to lean subjects is it increased postprandially(Reference Karpe, Fielding and Ilic29). Importantly, hypertrophic adipocytes in obese subjects can reach a size of approximately 150–200 μm in diameter(Reference Skurk, Alberti-Huber and Herder30), thus exceeding the normal O2 diffusion distance of 100–200 μm(Reference Brahimi-Horn and Pouyssegur31).

Hypoxia, defined as a deficiency of O2 in tissues, is more often associated with high altitude, ischaemic injury or tumour development. Cellular adaption to hypoxia is a highly-conserved evolutionary defence mechanism adapted by cells to conserve O2 for vital metabolic functions when levels are low or dramatically reduced. This response to low O2 levels is accomplished through the activation of specific transcription factors. The best known of these factors is hypoxic inducible factor (HIF)-1(Reference Semenza and Wang32, Reference Wang and Semenza33). The key element of this heterodimer is the subunit HIF-1α, which is considered to be the molecular oxygen sensor(Reference Semenza34). When cellular O2 levels are sufficient, this protein is continuously synthesised but is immediately targeted for proteasome degradation by the involvement of activated prolyl hydroxylase domain dioxygenases, which hydroxylate HIF-1α to provide sites for binding of the von Hippel-Lindau protein. Further hydroxylation occurs and prevents binding of the cAMP-binding protein-binding protein/p300 subunit co-factor. The presence of von Hippel-Lindau protein enables ubiquitination, thus targeting HIF-1α to the proteasome (summarised in Fig. 1). Under low O2 levels the prolyl hydroxylase domain enzymes are inactivated, thus stabilising HIF-1α by prevention of von Hippel-Lindau protein binding and enabling binding of the cAMP-binding protein-binding protein/p300 subunit co-factor.

Fig. 1. Overview of the regulation of hypoxic inducible factor (HIF)-1α under normoxia and hypoxia conditions. PHD, propyl hydrogenase domain proteins; VHL, von Hippel-Lindau protein; Ub, ubiquitin; HIF-1β, HIF-1β subunit; CBP/p300, cAMP-binding protein-binding protein/p300 subunit. , Increase; , decrease.

The second subunit, HIF-1β, a constitutively-expressed protein that is O2 insensitive, is able to bind to HIF-1α and form the active transcription factor. HIF-1 is involved in the activation of over seventy genes directly via binding to cis-acting hypoxic response elements; these genes involve a multiplicity of functions including angiogenesis, glucose metabolism, apoptosis, cellular stress, extracellular matrix re-modelling and inflammation(Reference Semenza35).

Three α subunits have been described, each being derived from a different gene. Whereas the main focus has been on HIF-1α, attention has recently been given to HIF-2α. The 2α subunit forms the transcription factor HIF-2, expression of which appears to be more tissue selective than HIF-1 and may respond at different cellular O2 levels(Reference Patel and Simon36). Little is known about HIF-3α and its splice variants, but there is some suggestion that one of these variants may act as a negative regulator(Reference Bardos and Ashcroft37). In addition to HIF-2, other transcription factors have been shown to mediate responses to low cellular O2 levels, including NF-κB and cAMP-response element-binding protein(Reference Leonard, Howell and Madden38, Reference Taylor39). NF-κB is more readily recognised as mediating inflammatory signalling pathways, such as those associated with TNFα, but would also appear to play a role in hypoxia, particularly intermittent hypoxia(Reference Taylor39).

Hypoxia and adipose tissue

Support for the hypoxia hypothesis was provided in 2007 by the demonstration that adipose tissue is in a hypoxic state in mouse models of obesity, both genetic and diet-induced(Reference Hosogai, Fukuhara and Oshima40, Reference Ye, Gao and Yin41). It was shown using O2 electrodes that the interstitial O2 partial pressure is lower in vivo in two different fat depots of ob/ob mice(Reference Ye, Gao and Yin41). The levels of O2 partial pressure reported for both lean and obese adipose tissue (48 and 15 mmHg respectively) are within the range of values described for general tissue oxygenation (40–50 mmHg) and tissue considered to be existing in a hypoxic state (e.g. retina; 2–15 mmHg)(Reference Brahimi-Horn and Pouyssegur31).

A second technique that was used to establish the presence of hypoxia is that of a pimonidazole hydrochloride stain, again in mouse obesity models. This dye is able to perfuse into tissue and forms protein adjuncts in areas in which the O2 tension is <10 mmHg (about 1% (v/v) O2). It was found that pimonidazole hydrochloride staining of adipose tissue from ob/ob mice is apparent when compared with lean littermates, as is the increased abundance of the probe measured by immunoblotting techniques indicating the presence of hypoxia(Reference Hosogai, Fukuhara and Oshima40, Reference Ye, Gao and Yin41). Importantly, there is a substantial induction of HIF-1α protein in the hypoxic adipose tissue of the obese animals(Reference Hosogai, Fukuhara and Oshima40, Reference Ye, Gao and Yin41). A very recent report has suggested that mild hypoxia is also evident in adipose tissue of obese human subjects(Reference Pasarica, Sereda and Redman42).

Hypoxia and adipocytes

In order to address the concept that there is a link between hypoxia and inflammatory-related adipokines, studies in Liverpool have focused on human adipocytes using primary culture systems. Exposure of mature adipocytes to 1% (v/v) O2 has been found to induce the expression of HIF-1α protein as early as 4 h, and importantly this induction is rapidly reversible; HIF-1α protein levels return to basal levels within 10 min of transferring adipocytes to control O2 conditions (21% (v/v) O2)(Reference Wang, Wood and Trayhurn43). Both the mRNA and protein levels of a key hypoxia marker, the facilitative glucose transporter GLUT-1, have been found to substantially increase, indicating that these cells are indeed responsive to a hypoxic environment(Reference Wang, Wood and Trayhurn43, Reference Wood, Wang and Lorente-Cebrian44). Using a candidate gene approach mRNA levels of fasting-induced adipose factor/angiopoietin-like protein 4, IL-6, leptin, macrophage migration inhibitory factor, plasminogen-activating inhibitor-1 and vascular endothelial growth factor have been found to increase substantially(Reference Wang, Wood and Trayhurn43). These increased mRNA levels are accompanied by increased protein secretion in the case of IL-6, leptin, macrophage migration inhibitory factor and vascular epithelial growth factor. Furthermore, adiponectin gene expression and protein secretion decrease.

Similar results have generally been observed when cells are treated with the hypoxia mimetic CoCl2, which is able to stabilise HIF-1α under normal O2 levels by inactivating the cellular prolyl hydroxylase domain hydroxylases(Reference Wang, Wood and Trayhurn43). Each of the hypoxia-sensitive genes has been found to be dependent on HIF-1 activation, with the exception of IL-6, which appears to be HIF-1 independent. Interestingly, expression of the key pro-inflammatory cytokine TNFα is not hypoxia sensitive. Overall, these findings on human adipocytes parallel those observed for mouse 3T3-L1 and 3T3-F442A fat cells(Reference Hosogai, Fukuhara and Oshima40, Reference Ye, Gao and Yin41, Reference Chen, Lam and Wang45) and imply that hypoxia may be the underlying cause of tissue inflammation and cellular dysfunction in obesity.

The response of human adipocytes to hypoxia has been further investigated using a commercial high-throughput real-time PCR strategy (PCR arrays). A collection of eighty-four genes related to hypoxia signalling were used to screen human adipocytes subjected to hypoxia for 24 h(Reference Wang, Wood and Trayhurn46). Of the twelve genes that were found to be up regulated, including those genes identified using the candidate gene approach described earlier (leptin, vascular epithelial growth factor etc.), the largest change that was observed was that for a previously-unreported adipocyte-related gene, metallothionein-3, the mRNA level of which was found to increase >600-fold over 24 h compared with untreated cells. This response is selective for this specific member of the metallothionein gene family, as the related gene, MT-2A, is minimally affected by hypoxia(Reference Wang, Wood and Trayhurn46). The role of metallothionein-3 in adipocytes is still unclear, but the gene is highly expressed in the brain and is thought to protect the cells against hypoxic damage(Reference Tanji, Irie and Uchida47).

Hypoxia and adipocyte metabolic function

The metabolic consequence of reduced cellular O2 levels within the adipocyte extends beyond that of altered adipokine expression. One of the initial steps in the response of cells to hypoxia is adaptation at the mitochondrial level by both reducing the amount of the high O2-consuming process of oxidative phosphorylation and simultaneously improving its efficiency(Reference Semenza48). The main site of O2 consumption is at complex IV, comprising cytochrome c oxidase subunit 4. However, this process is not fully efficient and some leakage can occur at complex III, resulting in the generation of reactive oxygen species. Under low O2 conditions the mitochondrial protease LON is up regulated and acts to degrade the cytochrome c oxidase subunit 4–1 subunit, which is subsequently replaced through the up-regulation of cytochrome c oxidase subunit 4–2; this subunit is more efficient for the consumption of O2(Reference Semenza48). These events have also been demonstrated with human adipocytes (B Wang, IS Wood and P Trayhurn, unpublished results).

As a result of reduced oxidative phosphorylation the cell switches to anaerobic glycolysis for its energy production. However, in order to compensate for the lower efficiency of ATP production, there is an increase in demand for glucose via up-regulation of GLUT-1 expression and insertion into the plasma membrane. As mentioned earlier, GLUT-1 is considered to be a molecular marker for cellular hypoxia. Indeed, the levels of GLUT-1 mRNA and protein have been shown to increase in human adipocytes in response to low O2 tension(Reference Wang, Wood and Trayhurn43, Reference Wood, Wang and Lorente-Cebrian44). Furthermore, an increase in glucose transport has been observed in hypoxic human adipocytes(Reference Wood, Wang and Lorente-Cebrian44), and more recently also in murine fat cells(Reference Regazzetti, Peraldi and Gremeaux49, Reference Yin, Gao and He50). What is not clear from the measurement of glucose uptake is the extent to which other glucose transporters may be involved in the response to hypoxia. Adipocytes have been shown to express a range of other members of the facilitative glucose transporter (GLUT) family(Reference Wood, Hunter and Trayhurn51, Reference Wood and Trayhurn52). Of these transporters, gene expression has been found to be increased for GLUT-3 and GLUT-5 (fructose transporter) in human adipocytes cultured in 1% (v/v) O2 for 24 h. On the other hand, GLUT-4, GLUT-10 and GLUT-12 mRNA levels are unchanged(Reference Wood, Wang and Lorente-Cebrian44).

Western blot analysis has shown that the protein abundance for GLUT-4 is also unchanged(Reference Wood, Wang and Lorente-Cebrian44). However, this outcome may reflect the presence of GLUT-4 mainly as intracellular stores that readily move to the plasma membrane in response to insulin. Hence, determination of total cellular GLUT-4 abundance in adipocyte cultures will not reveal whether GLUT-4 is involved in hypoxia-induced glucose uptake. Studies in muscle cells have shown that hypoxia can induce rapid translocation of GLUT-4 to the cell surface using mechanisms distinct from the insulin signalling pathway but similar to that observed with exercise-induced translocation of GLUT-4(Reference Douen, Ramlal and Rastogi53, Reference Zierath, Tsao and Stenbit54). However, initial studies in the authors' laboratory, using subcellular fractionation, have suggested that in human adipocytes GLUT-4 remains within the intracellular stores for between 1 and ⩽24 h of hypoxia treatment; furthermore, there is no acute increase in glucose transport following ⩽4 h of hypoxia (IS Wood and P Trayhurn, unpublished results).

A further consequence of the switch to anaerobic glycolysis is an increase in lactic acid production, which has to be rapidly exported from the cell; in the case of human adipocytes it has recently been found that hypoxia leads to a stimulation in lactate release (F Pérez de Heredia, IS Wood and P Trayhurn, unpublished results). The monocarboxylate transporters (MCT) are a large gene family comprising fourteen members, four of which (MCT-1–MCT-4) are recognised to transport lactate(Reference Meredith and Christian55). A recently reported study that used gene promoter analysis has found that MCT-4, but not MCT-1, transcription is increased under hypoxic conditions in HeLa cells(Reference Ullah, Davies and Halestrap56). Studies have found that several MCT are expressed in human adipocytes and that expression of both MCT-4 and MCT-1 is increased in response to hypoxia and is apparently HIF-1 dependent; interestingly, MCT-2 mRNA levels are decreased (F Pérez de Heredia, IS Wood and P Trayhurn, unpublished results). This finding would indicate that MCT are an important component of the increased glucose utilisation that occurs during the adaptation of adipocytes to a hypoxic environment.

Hypoxia and insulin resistance

The inflammatory state associated with obesity is considered to be a causal event in the development of insulin resistance(Reference de Luca and Olefsky57), to which adipose tissue dysfunction is considered to be a major contributor. Two adipokines implicated in the development of insulin resistance are IL-6 and adiponectin, the production of which increases and falls respectively in obesity and in response to hypoxia(Reference Hosogai, Fukuhara and Oshima40, Reference Ye, Gao and Yin41, Reference Wang, Wood and Trayhurn43). This finding has raised the question of whether hypoxia per se may be an underlying cause of insulin resistance. Indeed, recent studies have found that in mouse adipocytes subjected to 1% (v/v) O2 for 16–24 h insulin-stimulated glucose transport is attenuated(Reference Regazzetti, Peraldi and Gremeaux49, Reference Yin, Gao and He50). Moreover, the insulin receptor-β and insulin receptor substrate-1 protein levels are reduced in obese mice and 3T3-L1 cells(Reference Yin, Gao and He50). Phosphorylation of insulin receptor-β and insulin receptor substrate-1 has been shown to be reduced in both mouse and human adipocytes via HIF-1-dependent mechanisms(Reference Regazzetti, Peraldi and Gremeaux49). These findings provide strong evidence for a central role for adipose tissue hypoxia in the induction of insulin resistance.

A closer examination has been carried out of the role of GLUT-4 in the induction of insulin resistance, based on the primary observations that in obese subjects GLUT-4 protein levels decrease in adipose tissue, although not in skeletal muscle(Reference Shepherd and Kahn58), and that ablation of GLUT-4 in adipose tissue-specific knock-out mice results in whole-body insulin resistance (muscle and liver)(Reference Abel, Peroni and Kim59). The effects of prolonged (chronic) exposure of adipocytes in culture to hypoxia have been investigated with the view that this approach would better represent the situation for adipose tissue in obesity. Previous studies have found that adipocyte GLUT-4 mRNA and protein levels are unchanged for ⩽24 h exposure to 1% (v/v) O2(Reference Wood, Wang and Lorente-Cebrian44). However, by 48 h GLUT-4 mRNA levels fall(Reference Pérez de Heredia, Wood and Trayhurn60) and initial indications are that GLUT-4 protein abundance is reduced in both mouse and human adipocytes following 2–4 d exposure to 1% (v/v) O2 (F Pérez de Heredia, IS Wood and P Trayhurn, unpublished results). Further studies are underway to determine whether disturbance of GLUT-4 levels in adipose tissue as a result of hypoxia may contribute to the insulin resistance state associated with obesity.

Hypoxia and non-adipocyte cells

Other cellular components of adipose tissue include resident macrophages and preadipocytes, and these components may both contribute to the inflammatory response observed in adipose tissue. Indeed, preadipocytes have been found to exhibit a substantial response to inflammatory stimuli such as lipopolysaccharide(Reference Chung, Lapoint and Martinez61), with the increased secretion of inflammatory adipokines (e.g. IL-6, monocyte chemoattractant protein 1). They have also been found to secrete increased levels of IL-1β and TNFα in response to leptin(Reference Simons, van den Pangaart and van Roomen62). Hypoxia has been shown to inhibit differentiation of mouse preadipocytes, probably as a result of a decrease in PPARγ levels(Reference Yun, Maecker and Johnson63Reference Zhou, Lechpammer and Greenberger66). Consistent with this outcome are findings in human preadipocytes in which exposure to hypoxic conditions results in a fall in PPARγ gene expression, together with increased HIF-1α protein and GLUT-1 expression(Reference Wang, Wood and Trayhurn67). The overall response of preadipocytes to hypoxia appears to be blunted compared with that of adipocytes, there being some marked differences such as a lack of response in angiopoietin-like protein 4 and IL-6 expression to low O2 tension.

Perhaps the most interesting finding from the study on preadipocytes is that hypoxia induces the expression and secretion of leptin(Reference Wang, Wood and Trayhurn67); these cells are considered not to express leptin, which is a differentiation-dependent gene in adipocytes. However, hypoxia has been shown previously to increase leptin expression in other cell types(Reference Grosfeld, Andre and Hauguel-De Mouzon26, Reference Sagawa, Yura and Itoh68), including adipocytes(Reference Grosfeld, Zilberfarb and Turban27, Reference Hosogai, Fukuhara and Oshima40, Reference Wang, Wood and Trayhurn43, Reference Wang, Wood and Trayhurn46). These data imply that preadipocytes may contribute to a hypoxia-induced increase in leptin production by adipose tissue in obesity.

While the recruitment and infiltration of macrophages into obese adipose tissue have been recognised for several years, the mechanisms involved are still largely unknown. These cells are capable of increasing their inflammatory response in the face of hypoxia(Reference Lewis, Lee and Underwood69, Reference Oda, Hirota and Nishi70), and more recently in the specific context of adipose tissue(Reference Ye, Gao and Yin41). The finding that macrophages are present around areas of hypoxic adipocytes in adipose tissue(Reference Rausch, Weisberg and Vardhana71) may suggest that tissue hypoxia could provide a means of macrophage recruitment. The increased production of monocyte chemoattractant protein 1 (chemoattractant) and macrophage migration inhibitory factor-1 (prevention of macrophage release from tissue) have been proposed as key determinants of macrophage infiltration and enhanced inflammatory response(Reference Skurk, Herder and Kraft72, Reference Trayhurn, Wang and Wood73). Indeed, leptin has also been suggested as a macrophage chemoattractant(Reference Curat, Miranville and Sengenes21, Reference Gruen, Hao and Piston74).

In addition to an endocrine role, it is evident that the majority of adipokines produced by adipocytes may also act in a paracrine and/or autocrine manner within adipose tissue(Reference Trayhurn and Wood3, Reference Trayhurn and Wood75). Several studies have reported considerable inflammatory responses when cultured adipocytes and/or preadipocytes are treated with conditioned media used to culture macrophages(Reference Constant, Gagnon and Landry76Reference Permana, Zhang and Wabitsch79). It is highly likely that induction of adipose tissue hypoxia may elicit an inflammatory response from each of the different cells types that is amplified and promulgated through cellular cross talk (see Fig. 2).

Fig. 2. Cross talk between cell types within adipose tissue under hypoxia. Possible interactions occurring within adipose tissue that may be responsible for amplifying the inflammatory response observed during hypoxia. MIF-1, macrophage migration inhibitory factor-1; MMP9, matrix metallopeptidase-9; VEGF, vascular endothelial growth factor; PAI-1, plasminogen-activating factor-1; angptl4, angiopoietin-like protein-4; MT-3, metallothionein-3. , Increase; , decrease; , cross talk.

Implications and perspectives

It is evident that hypoxia can induce inflammatory responses in the different cell types contained within adipose tissue and that collectively these responses may underlie tissue dysfunction giving rise to the metabolic complications observed with obesity, consistent with the original hypothesis(Reference Trayhurn and Wood3, Reference Trayhurn and Wood75). However, it is recognised that hypoxia may not necessarily be the only factor involved. It should be noted that alternative mechanisms have been proposed linking cellular stress and inflammation, i.e. oxidative stress (mitochondrial reactive oxygen species generation)(Reference Houstis, Rosen and Lander80) and endoplasmic reticulum stress(Reference Gregor and Hotamisligil81, Reference Ozcan, Cao and Yilmaz82). Interestingly, hypoxia has also been shown to stimulate these two processes, suggesting that the various postulated mechanisms underlying inflammation are not necessary mutually exclusive. Indeed, a modest increase in reactive oxygen species production, through mitochondrial leakage within the electron transport chain when O2 levels initially fall below optimal levels, has been suggested as one possible mechanism to inhibit the prolyl hydroxylase domain enzymes and induce HIF-1 stability in order to protect against further rises in reactive oxygen species production(Reference Semenza48). Hypoxia has been shown to induce the expression of the CCAAT-enhancer-binding protein homologous protein and glucose-related protein 78 genes; these proteins are two markers of endoplasmic reticulum stress(Reference Hosogai, Fukuhara and Oshima40).

Obstructive sleep apnoea has received increased interest recently, mostly for its association with systemic inflammation and the development of insulin resistance (particularly in CVD), but also because obesity is recognised as a risk factor(Reference Williams and Scharf83). The cessation of breathing for brief periods during sleep induces bouts of chronic intermittent hypoxia that has marked mechanistic similarities to that found with continuous hypoxia (as described earlier). The response to intermittent hypoxia appears to be mediated through the transcription factor NF-κB, typically recognised as a major regulator of the inflammatory response, and has been suggested as a link between obstructive sleep apnoea and insulin resistance(Reference Williams and Scharf83). It is of interest to investigate whether systemic intermittent hypoxia has an influence on adipose tissue hypoxia.

The majority of studies have focused on hypoxia associated with increased fat mass through obesity. It is expected that hypoxia may also occur in other instances in which increased adiposity occurs, such as fat wrapping (associated with areas of mucosal inflammation in patients with Crohn's disease)(Reference Sheehan, Warren and Gear84) and pregnancy. These areas may provide opportunities to explore further the role of hypoxia under conditions not necessarily linked to overnutrition.

Therapeutic intervention in obesity has focused on the direct causes of the positive energy balance. However, targeting the consequences of obesity is important and may be more fruitful. From this perspective inflammation in adipose tissue may be a key target for treating the consequences of obesity. The recognition that hypoxia is found in obese adipose tissue presents new opportunities to reduce the adverse health consequences of a chronic inflammatory status. However, careful dissection of the intricate pathways will be required to provide specific targets to avoid unwanted interference with other essential systems.

Acknowledgements

We are grateful to the Biotechnology and Biological Sciences Research Council for financial support. F. P. de H. is funded by Fundación Alfonso Martín Escudero, Spain. P. T. is a member of the Management Committee of European Cooperation in the field of Scientific and Technical Research (COST) Action BM0602 (Adipose tissue: A key target for prevention of the metabolic syndrome). The authors declare no conflicts of interest. I. S. W. wrote the draft of the article, with subsequent revisions from F. P. deH., B. W. and P. T.; I. S. W. prepared the final version.

References

1.Zhang, Y, Proenca, R, Maffei, M et al. (1994) Positional cloning of the mouse obese gene and its human homologue. Nature 372, 425432.CrossRefGoogle ScholarPubMed
2.Trayhurn, P & Beattie, JH (2001) Physiological role of adipose tissue: white adipose tissue as an endocrine and secretory organ. Proc Nutr Soc 60, 329339.CrossRefGoogle ScholarPubMed
3.Trayhurn, P & Wood, IS (2004) Adipokines: inflammation and the pleiotropic role of white adipose tissue. Br J Nutr 92, 347355.CrossRefGoogle ScholarPubMed
4.Fantuzzi, G (2005) Adipose tissue, adipokines, and inflammation. J Allergy Clin Immunol 115, 911919.CrossRefGoogle ScholarPubMed
5.Rosen, ED & Spiegelman, BM (2006) Adipocytes as regulators of energy balance and glucose homeostasis. Nature 444, 847853.CrossRefGoogle ScholarPubMed
6.McPherson, K, Marsh, T & Brown, M (2007) Foresight. Tackling Obesities: Future Choices – Modelling Future Trends in Obesity and the Impact on Health, 2nd ed. London: The Stationery Office.Google Scholar
7.Phillips, LK & Prins, JB (2008) The link between abdominal obesity and the metabolic syndrome. Curr Hypertens Rep 10, 156164.CrossRefGoogle ScholarPubMed
8.Cancer Research UK (2009) Bodyweight and risk of cancer in the UK. http://info.cancerresearchuk.org/cancerstats/causes/lifestyle/bodyweight/Google Scholar
9.Engstrom, G, Hedblad, B, Stavenow, L et al. (2003) Inflammation-sensitive plasma proteins are associated with future weight gain. Diabetes 52, 20972101.CrossRefGoogle ScholarPubMed
10.Festa, A, D'Agostino, R Jr, Williams, K et al. (2001) The relation of body fat mass and distribution to markers of chronic inflammation. Int J Obes Relat Metab Disord 25, 14071415.CrossRefGoogle ScholarPubMed
11.Yudkin, JS (2003) Adipose tissue, insulin action and vascular disease: inflammatory signals. Int J Obes 27, Suppl. 3, S25S28.CrossRefGoogle ScholarPubMed
12.Yudkin, JS, Stehouwer, CD, Emeis, JJ et al. (1999) C-reactive protein in healthy subjects: associations with obesity, insulin resistance, and endothelial dysfunction: a potential role for cytokines originating from adipose tissue? Arterioscler Thromb Vasc Biol 19, 972978.CrossRefGoogle ScholarPubMed
13.Hotamisligil, GS, Shargill, NS & Spiegelman, BM (1993) Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science 259, 8791.CrossRefGoogle ScholarPubMed
14.Fain, JN (2006) Release of interleukins and other inflammatory cytokines by human adipose tissue is enhanced in obesity and primarily due to the nonfat cells. Vitam Horm 74, 443477.CrossRefGoogle Scholar
15.Arita, Y, Kihara, S, Ouchi, N et al. (1999) Paradoxical decrease of an adipose-specific protein, adiponectin, in obesity. Biochem Biophys Res Commun 257, 7983.CrossRefGoogle ScholarPubMed
16.Ouchi, N, Kihara, S, Arita, Y et al. (1999) Novel modulator for endothelial adhesion molecules: adipocyte-derived plasma protein adiponectin. Circulation 100, 24732476.CrossRefGoogle ScholarPubMed
17.Hotta, K, Funahashi, T, Arita, Y et al. (2000) Plasma concentrations of a novel, adipose-specific protein, adiponectin, in type 2 diabetic patients. Arterioscler Thromb Vasc Biol 20, 15951599.CrossRefGoogle ScholarPubMed
18.Weisberg, SP, McCann, D, Desai, M et al. (2003) Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest 112, 17961808.CrossRefGoogle ScholarPubMed
19.Xu, H, Barnes, GT, Yang, Q et al. (2003) Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J Clin Invest 112, 18211830.CrossRefGoogle Scholar
20.Bouloumie, A, Curat, CA, Sengenes, C et al. (2005) Role of macrophage tissue infiltration in metabolic diseases. Curr Opin Clin Nutr Metab Care 8, 347354.CrossRefGoogle ScholarPubMed
21.Curat, CA, Miranville, A, Sengenes, C et al. (2004) From blood monocytes to adipose tissue-resident macrophages: induction of diapedesis by human mature adipocytes. Diabetes 53, 12851292.CrossRefGoogle ScholarPubMed
22.Fleischmann, E, Kurz, A, Niedermayr, M et al. (2005) Tissue oxygenation in obese and non-obese patients during laparoscopy. Obes Surg 15, 813819.CrossRefGoogle ScholarPubMed
23.Rupnick, MA, Panigrahy, D, Zhang, CY et al. (2002) Adipose tissue mass can be regulated through the vasculature. Proc Natl Acad Sci USA 99, 1073010735.CrossRefGoogle ScholarPubMed
24.Lolmede, K, Durand de Saint Front, V, Galitzky, J et al. (2003) Effects of hypoxia on the expression of proangiogenic factors in differentiated 3T3-F442A adipocytes. Int J Obes Relat Metab Disord 27, 11871195.CrossRefGoogle ScholarPubMed
25.Ambrosini, G, Nath, AK, Sierra-Honigmann, MR et al. (2002) Transcriptional activation of the human leptin gene in response to hypoxia. Involvement of hypoxia-inducible factor 1. J Biol Chem 277, 3460134609.CrossRefGoogle ScholarPubMed
26.Grosfeld, A, Andre, J, Hauguel-De Mouzon, S et al. (2002) Hypoxia-inducible factor 1 transactivates the human leptin gene promoter. J Biol Chem 277, 4295342957.CrossRefGoogle ScholarPubMed
27.Grosfeld, A, Zilberfarb, V, Turban, S et al. (2002) Hypoxia increases leptin expression in human PAZ6 adipose cells. Diabetologia 45, 527530.CrossRefGoogle ScholarPubMed
28.Kabon, B, Nagele, A, Reddy, D et al. (2004) Obesity decreases perioperative tissue oxygenation. Anesthesiology 100, 274280.CrossRefGoogle ScholarPubMed
29.Karpe, F, Fielding, BA, Ilic, V et al. (2002) Impaired postprandial adipose tissue blood flow response is related to aspects of insulin sensitivity. Diabetes 51, 24672473.CrossRefGoogle ScholarPubMed
30.Skurk, T, Alberti-Huber, C, Herder, C et al. (2007) Relationship between adipocyte size and adipokine expression and secretion. J Clin Endocrinol Metab 92, 10231033.CrossRefGoogle ScholarPubMed
31.Brahimi-Horn, MC & Pouyssegur, J (2007) Oxygen, a source of life and stress. FEBS Lett 581, 35823591.CrossRefGoogle ScholarPubMed
32.Semenza, GL & Wang, GL (1992) A nuclear factor induced by hypoxia via de novo protein synthesis binds to the human erythropoietin gene enhancer at a site required for transcriptional activation. Mol Cell Biol 12, 54475454.Google Scholar
33.Wang, GL & Semenza, GL (1993) General involvement of hypoxia-inducible factor 1 in transcriptional response to hypoxia. Proc Natl Acad Sci USA 90, 43044308.CrossRefGoogle ScholarPubMed
34.Semenza, GL (1998) Hypoxia-inducible factor 1: master regulator of O2 homeostasis. Curr Opin Genet Dev 8, 588594.CrossRefGoogle ScholarPubMed
35.Semenza, GL (2003) Targeting HIF-1 for cancer therapy. Nat Rev Cancer 3, 721732.CrossRefGoogle ScholarPubMed
36.Patel, SA & Simon, MC (2008) Biology of hypoxia-inducible factor-2alpha in development and disease. Cell Death Differ 15, 628634.CrossRefGoogle ScholarPubMed
37.Bardos, JI & Ashcroft, M (2005) Negative and positive regulation of HIF-1: a complex network. Biochim Biophys Acta 1755, 107120.Google ScholarPubMed
38.Leonard, MO, Howell, K, Madden, SF et al. (2008) Hypoxia selectively activates the CREB family of transcription factors in the in vivo lung. Am J Respir Crit Care Med 178, 977983.CrossRefGoogle ScholarPubMed
39.Taylor, CT (2008) Interdependent roles for hypoxia inducible factor and nuclear factor-kappaB in hypoxic inflammation. J Physiol 586, 40554059.CrossRefGoogle ScholarPubMed
40.Hosogai, N, Fukuhara, A, Oshima, K et al. (2007) Adipose tissue hypoxia in obesity and its impact on adipocytokine dysregulation. Diabetes 56, 901911.CrossRefGoogle ScholarPubMed
41.Ye, J, Gao, Z, Yin, J et al. (2007) Hypoxia is a potential risk factor for chronic inflammation and adiponectin reduction in adipose tissue of ob/ob and dietary obese mice. Am J Physiol Endocrinol Metab 293, E1118E1128.CrossRefGoogle Scholar
42.Pasarica, M, Sereda, OR, Redman, LM et al. (2009) Reduced adipose tissue oxygenation in human obesity: evidence for rarefaction, macrophage chemotaxis, and inflammation without an angiogenic response. Diabetes 58, 718725.CrossRefGoogle ScholarPubMed
43.Wang, B, Wood, IS & Trayhurn, P (2007) Dysregulation of the expression and secretion of inflammation-related adipokines by hypoxia in human adipocytes. Pflugers Arch 455, 479492.CrossRefGoogle ScholarPubMed
44.Wood, IS, Wang, B, Lorente-Cebrian, S et al. (2007) Hypoxia increases expression of selective facilitative glucose transporters (GLUT) and 2-deoxy-D-glucose uptake in human adipocytes. Biochem Biophys Res Commun 361, 468473.CrossRefGoogle ScholarPubMed
45.Chen, B, Lam, KS, Wang, Y et al. (2006) Hypoxia dysregulates the production of adiponectin and plasminogen activator inhibitor-1 independent of reactive oxygen species in adipocytes. Biochem Biophys Res Commun 341, 549556.CrossRefGoogle ScholarPubMed
46.Wang, B, Wood, IS & Trayhurn, P (2008) PCR arrays identify metallothionein-3 as a highly hypoxia-inducible gene in human adipocytes. Biochem Biophys Res Commun 368, 8893.CrossRefGoogle ScholarPubMed
47.Tanji, K, Irie, Y, Uchida, Y et al. (2003) Expression of metallothionein-III induced by hypoxia attenuates hypoxia-induced cell death in vitro. Brain Res 976, 125129.CrossRefGoogle ScholarPubMed
48.Semenza, GL (2007) Oxygen-dependent regulation of mitochondrial respiration by hypoxia-inducible factor 1. Biochem J 405, 19.CrossRefGoogle ScholarPubMed
49.Regazzetti, C, Peraldi, P, Gremeaux, T et al. (2009) Hypoxia decreases insulin signaling pathways in adipocytes. Diabetes 58, 95–103.CrossRefGoogle ScholarPubMed
50.Yin, J, Gao, Z, He, Q et al. (2009) Role of hypoxia in obesity-induced disorders of glucose and lipid metabolism in adipose tissue. Am J Physiol Endocrinol Metab 296, E333E342.CrossRefGoogle ScholarPubMed
51.Wood, IS, Hunter, L & Trayhurn, P (2003) Expression of Class III facilitative glucose transporter genes (GLUT-10 and GLUT-12) in mouse and human adipose tissues. Biochem Biophys Res Commun 308, 4349.CrossRefGoogle ScholarPubMed
52.Wood, IS & Trayhurn, P (2003) Glucose transporters (GLUT and SGLT): expanded families of sugar transport proteins. Br J Nutr 89, 39.CrossRefGoogle ScholarPubMed
53.Douen, A, Ramlal, T, Rastogi, S et al. (1990) Exercise induces recruitment of the ‘insulin-responsive glucose transporter’. Evidence for distinct intracellular insulin- and exercise-recruitable transporter pools in skeletal muscle. J Biol Chem 265, 1342713430.CrossRefGoogle ScholarPubMed
54.Zierath, JR, Tsao, TS, Stenbit, AE et al. (1998) Restoration of hypoxia-stimulated glucose uptake in GLUT4-deficient muscles by muscle-specific GLUT4 transgenic complementation. J Biol Chem 273, 2091020915.CrossRefGoogle ScholarPubMed
55.Meredith, D & Christian, HC (2008) The SLC16 monocaboxylate transporter family. Xenobiotica 38, 10721106.CrossRefGoogle ScholarPubMed
56.Ullah, MS, Davies, AJ & Halestrap, AP (2006) The plasma membrane lactate transporter MCT4, but not MCT1, is up-regulated by hypoxia through a HIF-1alpha-dependent mechanism. J Biol Chem 281, 90309037.CrossRefGoogle Scholar
57.de Luca, C & Olefsky, JM (2008) Inflammation and insulin resistance. FEBS Lett 582, 97–105.CrossRefGoogle ScholarPubMed
58.Shepherd, PR & Kahn, BB (1999) Glucose transporters and insulin action – implications for insulin resistance and diabetes mellitus. N Engl J Med 341, 248257.CrossRefGoogle ScholarPubMed
59.Abel, ED, Peroni, O, Kim, JK et al. (2001) Adipose-selective targeting of the GLUT4 gene impairs insulin action in muscle and liver. Nature 409, 729733.CrossRefGoogle ScholarPubMed
60.Pérez de Heredia, F, Wood, IS & Trayhurn, P (2009) Acute and chronic hypoxia selectively modulates the expression of glucose transporters (GLUTs) in human adipocytes. Obes Facts 2, Suppl. 2, 39.Google Scholar
61.Chung, S, Lapoint, K, Martinez, K et al. (2006) Preadipocytes mediate lipopolysaccharide-induced inflammation and insulin resistance in primary cultures of newly differentiated human adipocytes. Endocrinology 147, 53405351.CrossRefGoogle ScholarPubMed
62.Simons, PJ, van den Pangaart, PS, van Roomen, CP et al. (2005) Cytokine-mediated modulation of leptin and adiponectin secretion during in vitro adipogenesis: evidence that tumor necrosis factor-alpha- and interleukin-1beta-treated human preadipocytes are potent leptin producers. Cytokine 32, 94–103.CrossRefGoogle ScholarPubMed
63.Yun, Z, Maecker, HL, Johnson, RS et al. (2002) Inhibition of PPAR gamma 2 gene expression by the HIF-1-regulated gene DEC1/Stra13: a mechanism for regulation of adipogenesis by hypoxia. Dev Cell 2, 331341.CrossRefGoogle ScholarPubMed
64.Carriere, A, Carmona, MC, Fernandez, Y et al. (2004) Mitochondrial reactive oxygen species control the transcription factor CHOP-10/GADD153 and adipocyte differentiation: a mechanism for hypoxia-dependent effect. J Biol Chem 279, 4046240469.CrossRefGoogle ScholarPubMed
65.Kim, KH, Song, MJ, Chung, J et al. (2005) Hypoxia inhibits adipocyte differentiation in a HDAC-independent manner. Biochem Biophys Res Commun 333, 11781184.CrossRefGoogle Scholar
66.Zhou, S, Lechpammer, S, Greenberger, JS et al. (2005) Hypoxia inhibition of adipocytogenesis in human bone marrow stromal cells requires transforming growth factor-beta/Smad3 signaling. J Biol Chem 280, 2268822696.CrossRefGoogle ScholarPubMed
67.Wang, B, Wood, IS & Trayhurn, P (2008) Hypoxia induces leptin gene expression and secretion in human preadipocytes: differential effects of hypoxia on adipokine expression by preadipocytes. J Endocrinol 198, 127134.CrossRefGoogle ScholarPubMed
68.Sagawa, N, Yura, S, Itoh, H et al. (2002) Role of leptin in pregnancy-a review. Placenta 23, Suppl. A, S80S86.CrossRefGoogle ScholarPubMed
69.Lewis, JS, Lee, JA, Underwood, JC et al. (1999) Macrophage responses to hypoxia: relevance to disease mechanisms. J Leukoc Biol 66, 889900.CrossRefGoogle ScholarPubMed
70.Oda, T, Hirota, K, Nishi, K et al. (2006) Activation of hypoxia-inducible factor 1 during macrophage differentiation. Am J Physiol Cell Physiol 291, C104C113.CrossRefGoogle ScholarPubMed
71.Rausch, ME, Weisberg, S, Vardhana, P et al. (2008) Obesity in C57BL/6J mice is characterized by adipose tissue hypoxia and cytotoxic T-cell infiltration. Int J Obes (Lond) 32, 451463.CrossRefGoogle ScholarPubMed
72.Skurk, T, Herder, C, Kraft, I et al. (2005) Production and release of macrophage migration inhibitory factor from human adipocytes. Endocrinology 146, 10061011.CrossRefGoogle ScholarPubMed
73.Trayhurn, P, Wang, B & Wood, IS (2008) Hypoxia in adipose tissue: a basis for the dysregulation of tissue function in obesity? Br J Nutr 100, 227235.CrossRefGoogle ScholarPubMed
74.Gruen, ML, Hao, M, Piston, DW et al. (2007) Leptin requires canonical migratory signaling pathways for induction of monocyte and macrophage chemotaxis. Am J Physiol Cell Physiol 293, C1481C1488.CrossRefGoogle ScholarPubMed
75.Trayhurn, P & Wood, IS (2005) Signalling role of adipose tissue: adipokines and inflammation in obesity. Biochem Soc Trans 33, 10781081.CrossRefGoogle ScholarPubMed
76.Constant, VA, Gagnon, A, Landry, A et al. (2006) Macrophage-conditioned medium inhibits the differentiation of 3T3-L1 and human abdominal preadipocytes. Diabetologia 49, 14021411.CrossRefGoogle ScholarPubMed
77.Lacasa, D, Taleb, S, Keophiphath, M et al. (2007) Macrophage-secreted factors impair human adipogenesis: involvement of proinflammatory state in preadipocytes. Endocrinology 148, 868877.CrossRefGoogle Scholar
78.Permana, PA, Menge, C & Reaven, PD (2006) Macrophage-secreted factors induce adipocyte inflammation and insulin resistance. Biochem Biophys Res Commun 341, 507514.CrossRefGoogle ScholarPubMed
79.Permana, PA, Zhang, W, Wabitsch, M et al. (2009) Pioglitazone reduces inflammatory responses of human adipocytes to factors secreted by monocytes/macrophages. Am J Physiol Endocrinol Metab 296, E1076E1084.CrossRefGoogle ScholarPubMed
80.Houstis, N, Rosen, ED & Lander, ES (2006) Reactive oxygen species have a causal role in multiple forms of insulin resistance. Nature 440, 944948.CrossRefGoogle ScholarPubMed
81.Gregor, MF & Hotamisligil, GS (2007) Adipocyte stress: the endoplasmic reticulum and metabolic disease. J Lipid Res 48, 19051914.CrossRefGoogle ScholarPubMed
82.Ozcan, U, Cao, Q, Yilmaz, E et al. (2004) Endoplasmic reticulum stress links obesity, insulin action, and type 2 diabetes. Science 306, 457461.CrossRefGoogle ScholarPubMed
83.Williams, A & Scharf, SM (2007) Obstructive sleep apnea, cardiovascular disease, and inflammation – is NF-kappaB the key? Sleep Breath 11, 6976.CrossRefGoogle ScholarPubMed
84.Sheehan, AL, Warren, BF, Gear, MW et al. (1992) Fat-wrapping in Crohn's disease: pathological basis and relevance to surgical practice. Br J Surg 79, 955958.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1. Overview of the regulation of hypoxic inducible factor (HIF)-1α under normoxia and hypoxia conditions. PHD, propyl hydrogenase domain proteins; VHL, von Hippel-Lindau protein; Ub, ubiquitin; HIF-1β, HIF-1β subunit; CBP/p300, cAMP-binding protein-binding protein/p300 subunit. , Increase; , decrease.

Figure 1

Fig. 2. Cross talk between cell types within adipose tissue under hypoxia. Possible interactions occurring within adipose tissue that may be responsible for amplifying the inflammatory response observed during hypoxia. MIF-1, macrophage migration inhibitory factor-1; MMP9, matrix metallopeptidase-9; VEGF, vascular endothelial growth factor; PAI-1, plasminogen-activating factor-1; angptl4, angiopoietin-like protein-4; MT-3, metallothionein-3. , Increase; , decrease; , cross talk.