Hostname: page-component-7c8c6479df-r7xzm Total loading time: 0 Render date: 2024-03-27T03:38:23.884Z Has data issue: false hasContentIssue false

Hypoxia in adipose tissue: a basis for the dysregulation of tissue function in obesity?

Published online by Cambridge University Press:  01 August 2008

Paul Trayhurn*
Affiliation:
Obesity Biology Unit, School of Clinical Sciences, University of Liverpool, University Clinical Departments, Duncan Building, LiverpoolL69 3GA, UK
Bohan Wang
Affiliation:
Obesity Biology Unit, School of Clinical Sciences, University of Liverpool, University Clinical Departments, Duncan Building, LiverpoolL69 3GA, UK
I. Stuart Wood
Affiliation:
Obesity Biology Unit, School of Clinical Sciences, University of Liverpool, University Clinical Departments, Duncan Building, LiverpoolL69 3GA, UK
*
*Corresponding author: Professor Paul Trayhurn, fax +44 151 706 5802, email p.trayhurn@liverpool.ac.uk
Rights & Permissions [Opens in a new window]

Abstract

White adipose tissue is a key endocrine and secretory organ, releasing multiple adipokines, many of which are linked to inflammation and immunity. During the expansion of adipose tissue mass in obesity there is a major inflammatory response in the tissue with increased expression and release of inflammation-related adipokines, including IL-6, leptin, monocyte chemoattractant protein-1 and TNF-α, together with decreased adiponectin production. We proposed in 2004 (Trayhurn & Wood, Br J Nutr92, 347–355) that inflammation in adipose tissue in obesity is a response to hypoxia in enlarged adipocytes distant from the vasculature. Hypoxia has now been directly demonstrated in adipose tissue of several obese mouse models (ob/ob, KKAy, diet-induced) and molecular studies indicate that the level of the hypoxia-inducible transcription factor, hypoxia-inducible factor-1α, is increased, as is expression of the hypoxia-sensitive marker gene, GLUT1. Cell- culture studies on murine and human adipocytes show that hypoxia (induced by low O2 or chemically) leads to stimulation of the expression and secretion of a number of inflammation-related adipokines, including angiopoietin-like protein 4, IL-6, leptin, macrophage migration inhibitory factor and vascular endothelial growth factor. Hypoxia also stimulates the inflammatory response of macrophages and inhibits adipocyte differentiation from preadipocytes. GLUT1 gene expression, protein level and glucose transport by human adipocytes are markedly increased by hypoxia, indicating that low O2 tension stimulates glucose utilisation. It is suggested that hypoxia has a pervasive effect on adipocyte metabolism and on overall adipose tissue function, underpinning the inflammatory response in the tissue in obesity and the subsequent development of obesity-associated diseases, particularly type 2 diabetes and the metabolic syndrome.

Type
Horizons in Nutritional Science
Copyright
Copyright © The Authors 2008

White adipose tissue – a major secretory organ

Our understanding of the physiological role of white adipose tissue (WAT) has changed radically over the past 15 years. What was viewed primarily as a vehicle for the highly efficient storage of fuel in the form of TAG has emerged as a major endocrine and secretory organ(Reference Trayhurn and Beattie1Reference Rosen and Spiegelman5). Indeed, for many individuals, and certainly the obese, adipose tissue is the largest endocrine organ. This is because white adipocytes, the predominant cell type within the tissue, release a multiplicity of factors in addition to fatty acids – which are quantitatively much the largest secretory product. The ‘secretome’ of the adipocyte also encompasses other lipid moieties, including cholesterol and retinol (neither of which is synthesised de novo by adipocytes), and steroid hormones and prostaglandins.

The paradigm shift in our understanding of the physiological role of WAT came in 1994 with the discovery of the protein hormone leptin, the product of the Lep (or Ob) gene(Reference Zhang, Proenca, Maffei, Barone, Leopold and Friedman6). This resulted in the immediate recognition of WAT as a major endocrine organ. Leptin was not, however, the first protein signal to be identified as being secreted from fat cells. The complement-related factor adipsin had been shown in the mid-1980s to be released from adipocytes(Reference Cook, Min, Johnson, Chaplinsky, Flier, Hunt and Spiegelman7, Reference Flier, Cook, Usher and Spiegelman8), while several years later the synthesis and secretion of the pro-inflammatory cytokine, TNF-α, was demonstrated(Reference Hotamisligil, Shargill and Spiegelman9).

Following the discovery of leptin, which was originally shown to be a satiety factor but is in practice a pleiotropic hormone, a rapidly expanding range of protein signals and factors termed adipokines (originally referred to as adipocytokines) have been found to be released from white adipocytes(Reference Trayhurn and Beattie1Reference Rosen and Spiegelman5). A number of these adipokines are linked to immunity and the inflammatory response, and their production is generally increased in obesity such that the tissue is in a state of inflammation(Reference Trayhurn and Wood4, Reference Hotamisligil10, Reference Wellen and Hotamisligil11). A key exception is adiponectin, with its anti-inflammatory and insulin-sensitising actions, the production and release of which fall in the obese(Reference Arita, Kihara and Ouchi12). Obesity itself is characterised by a state of chronic mild inflammation, the circulating levels of several inflammatory markers (such as C-reactive protein, haptoglobin, IL-6, monocyte chemoattractant protein-1, plasminogen activator inhibitor-1 (PAI-1), TNF-α) being increased(Reference Hotamisligil10, Reference Wellen and Hotamisligil11). This state of inflammation, particularly in adipose tissue through changes in the release of inflammation-related adipokines and the infiltration of macrophages(Reference Xu, Barnes and Yang13, Reference Weisberg, McCann, Desai, Rosenbaum, Leibel and Ferrante14), is viewed as central to the development of the major obesity-associated diseases – the metabolic syndrome, type 2 diabetes, CVD and certain cancers (such as breast and colon)(Reference Rosen and Spiegelman5, Reference Hotamisligil10, Reference Yudkin15).

Limited attention has been paid, however, as to why adipose tissue should mount an inflammatory response in obesity. The propositions include oxidative stress and endoplasmic reticulum stress(Reference Ozcan, Cao, Yilmaz, Lee, Iwakoshi, Ozdelen, Tuncman, Görgün, Glimcher and Hotamisligil16Reference Houstis, Rosen and Lander18). An alternative proposal, which we put forward in the British Journal of Nutrition in 2004, is that it is a response to hypoxia(Reference Trayhurn and Wood4). In the present article, we expand on the hypoxia concept and review the developments that have occurred in the past 2–3 years on the relationship between low O2 tension and adipose tissue function. It is emphasised that the different proposals for the basis for the inflammatory response in adipose tissue in obesity are not necessarily mutually exclusive; for example, hypoxia has been reported to induce endoplasmic reticulum stress and the generation of reactive oxygen species(Reference Koumenis, Naczki, Koritzinsky, Rastani, Diehl, Sonenberg, Koromilas and Wouters19, Reference Carriere, Carmona, Fernandez, Rigoulet, Wenger, Penicaud and Casteilla20).

The ‘hypoxia hypothesis’

In the original proposition it was suggested that the dysregulation of the production of inflammation-related adipokines in obesity, linked to the development of the metabolic syndrome and other obesity-associated disorders, is a specific response to relative hypoxia in clusters of adipocytes that become distant from the vasculature as adipose tissue mass expands(Reference Trayhurn and Wood4). The increased production of inflammation-related adipokines, including angiogenic signals, would be expected to lead acutely to increased blood flow and to the long-term stimulation of angiogenesis. The proposal built on the earlier suggestion that hypoxia may stimulate the expression of angiogenic factors and neovascularisation within WAT(Reference Lolmède, Durand de Saint Front, Galitzky, Lafontan and Bouloumié21).

There are several circumstantial arguments in favour of the hypoxia hypothesis. These are, in particular: (i) the proportion of the cardiac output and blood flow that goes to WAT is not increased in the obese, despite the expansion of the tissue mass(Reference Virtanen, Lonnroth and Parkkola22, Reference Kabon, Nagele, Reddy, Eagon, Fleshman, Sessler and Kurz23); (ii) obese subjects do not exhibit the postprandial increase in blood flow to adipose tissue that occurs in the lean(Reference Karpe, Fielding, Ilic, Macdonald, Summers and Frayn24); (iii) hypertrophied adipocytes are larger than the normal diffusion distance of O2 within tissues(Reference Lolmède, Durand de Saint Front, Galitzky, Lafontan and Bouloumié21). With respect to the last point, large adipocytes may be up to 150 (or even 200) μm in diameter(Reference Skurk, Alberti-Huber, Herder and Hauner25) with the normal diffusion distance of O2 being 100–200 μm(Reference Brahimi-Horn and Pouysségur26). Indeed, in some situations and in some tissues, a partial pressure of O2 of close to zero has been reported at only 100 μm from the vasculature(Reference Brahimi-Horn and Pouysségur26Reference Gatenby and Gillies28).

Hypoxia is well recognised to be characteristic of pathological situations such as wound healing, ischaemic disorders and solid tumours(Reference Brahimi-Horn and Pouysségur26, Reference Semenza29). Tissues in which O2 tension is normally low include the retina, where values of 2–25 mmHg are apparent, while parts of the rat brain may be even more hypoxic at just 0·8–8 mmHg(Reference Brahimi-Horn and Pouysségur26, Reference Erecinska and Silver30). These levels of O2 should be compared with a partial pressure of O2 of approximately 104 mmHg in the blood from the alveolar capillaries of the lungs and the arteries(Reference Brahimi-Horn and Pouysségur26), as summarised in Table 1.

Table 1 Oxygen levels in different tissues, including tissues where hypoxia is evident, as well as recent data on white adipose tissue from lean and obese mice

It is widely suggested that WAT is poorly oxygenated in the obese(Reference Virtanen, Lonnroth and Parkkola22, Reference Kabon, Nagele, Reddy, Eagon, Fleshman, Sessler and Kurz23, Reference Fleischmann, Kurz, Niedermayr, Schebesta, Kimberger, Sessler, Kabon and Prager31). Indeed, early data indicate an inverse relationship between fat cell size and the resting blood flow to adipose tissue in dogs(Reference Di Girolamo, Skinner, Hanley and Sachs32). Furthermore, one of us in a study in 1980 on brown adipose tissue thermogenesis found that the proportion of the cardiac output, measured using radioactively labelled microspheres, and the total blood flow to WAT (plus skin) was not increased in obese (ob/ob) mice relative to their lean siblings(Reference Thurlby and Trayhurn33). Similar studies have shown that this is also the case for the Zucker fa/fa rat(Reference West, Prinz, Francendese and Greenwood34).

Molecular signalling of hypoxia

The molecular mechanisms by which cells sense and respond to hypoxia have been extensively investigated and a number of excellent reviews have recently been published on this topic(Reference Brahimi-Horn and Pouysségur26, Reference Semenza35Reference Rocha39). Here, we outline the key components of the cellular hypoxia signalling pathway.

There are several transcription factors which are implicated in the molecular response to hypoxia, including NFκB and cAMP response element binding protein(Reference Cummins and Taylor38). Hypoxia-induced activation of NFκB, which has recently been demonstrated also in adipocytes(Reference Ye, Gao, Yin and He40), modulates the transcription of target genes such as cyclo-oxygenase 2 and TNF-α(Reference Cummins and Taylor38). However, a pivotal role in the response to hypoxia is played by hypoxia-inducible factor (HIF)-1 which is regarded as the ‘master regulator of O2 homeostasis’. This is a heterodimer composed of α- and β-subunits, one of which, HIF-1β, is constitutively expressed and not regulated directly by O2(Reference Brahimi-Horn and Pouysségur26, Reference Semenza35, Reference Rocha39).

There are at least three α-subunits of HIF-1, namely HIF-1α, HIF-2α and HIF-3α, and the combination of any of these with HIF-1β forms the functional transcription factor. HIF-1α is the most extensively investigated subunit, and appears to be the most important; little is known about HIF-3α(Reference Rocha39). In the presence of O2, HIF-1α (and HIF-2α) is constantly synthesised and degraded via the 26S proteosomal system. This process is initiated by the presence of active prolyl hydroxylation domain proteins which hydroxylate HIF-1α at specific proline residues, thereby presenting binding sites for the von Hippel–Lindau protein(Reference Ivan, Kondo, Yang, Kim, Valiando, Ohh, Salic, Asara, Lane and Kaelin41, Reference Jaakkola, Mole and Tian42). This von Hippel–Lindau protein forms an E3 ligase complex resulting in the polyubiquitylation of HIF-1α, tagging it for destruction. Simultaneously, transcriptional activity is prevented by the action of FIH (factor inhibiting HIF-1) which hydroxylates a specific arginine residue and prevents binding of the cAMP response element binding protein binding protein–p300 complex. Under hypoxic conditions these hydroxylases are rendered inactive. Consequently, HIF-1α is stabilised and HIF-1 moves into the nucleus where it binds to hypoxia response elements within target genes and initiates their transcription (see Fig. 1).

Fig. 1 Diagrammatic view of the molecular signalling response to hypoxia through the hypoxia-inducible factor (HIF) system. PHD, prolyl hydroxylase; FIH, factor inhibiting HIF-1; vHL, von Hippel–Lindau tumour suppressor protein; p300–CBP, p300–cAMP response element binding protein binding protein (transcriptional coactivator); HRE, hypoxia-response element; Ub, ubiquitin.

A substantial number of genes are recognised to be hypoxia sensitive through the hypoxia response elements for HIF-1, and these currently number about seventy. The target genes include those encoding proteins involved in angiogenesis, cell proliferation and survival, apoptosis, vascular tone, and glucose and energy metabolism(Reference Semenza29, Reference Rocha39). Specific hypoxia-sensitive genes include the facilitative glucose transporter, GLUT1, responsible for basal glucose uptake in many cells, leptin, the major angiogenic factor vascular endothelial growth factor (VEGF) and several enzymes involved in glucose metabolism such as phosphofructokinase(Reference Semenza29, Reference Rocha39). GLUT1 in particular, the first of the fourteen-member facilitative GLUT family (see later section ‘Beyond adipokines’) to be identified(Reference Mueckler, Caruso, Baldwin, Panico, Blench, Morris, Allard, Lienhard and Lodish43), is commonly employed as a marker gene of responsiveness to low O2 tension by cells, or a given tissue.

The tools used for investigating the effects of hypoxia and the role of HIF-1 in cell function encompass ‘hypoxia mimetics’ which induce a hypoxic response chemically. These agents include CoCl2, desferrioxamine and dimethyloxallyl glycine, which act by stabilising HIF-1α through inhibition of the prolyl hydroxylases which normally, in the presence of O2, instigate the degradation of the transcription factor subunit. This approach is extensively used in cell-culture studies in addition to, or instead of, incubation under low O2 tension, and helps to define whether signalling through HIF-1α occurs in the case of a particular hypoxia-sensitive gene.

Hypoxia in adipose tissue of obese animals

There are several techniques for measuring whether hypoxia is occurring in specific tissues in vivo, and these include the use of O2 microelectrodes and staining with the chemical marker pimonidazole hydrochloride. The latter is employed primarily in immunohistochemical analyses, forming an adduct with proteins when there is tissue hypoxia. The critical question of whether WAT is hypoxic in obesity has now been directly addressed with pimonidazole in obese mouse models. Using this technique, evidence for hypoxia in WAT has been clearly demonstrated in genetically obese KKAy and ob/ob mice, and in mice made obese through the provision of a high-fat diet(Reference Ye, Gao, Yin and He40, Reference Hosogai, Fukuhara and Oshima44, Reference Rausch, Weisberg, Vardhana and Tortoriello45). Hypoxia in WAT has also been demonstrated in both ob/ob mice and in diet-induced obesity using O2 sensors(Reference Ye, Gao, Yin and He40, Reference Rausch, Weisberg, Vardhana and Tortoriello45). In addition, reduced perfusion of WAT is evident in diet-induced and KKAy obese mice from studies using radiolabelled microspheres(Reference Hosogai, Fukuhara and Oshima44). These reductions in blood perfusion are selective for adipose tissue, there being no decrease in skeletal muscle, the kidney, heart or lungs.

Several molecular indices of hypoxia have also been examined in obese models. Increased HIF-1α protein levels have been found in WAT of both ob/ob and diet-induced obese mice, indicative of hypoxia(Reference Ye, Gao, Yin and He40, Reference Rausch, Weisberg, Vardhana and Tortoriello45). GLUT1 mRNA levels are also increased(Reference Ye, Gao, Yin and He40, Reference Hosogai, Fukuhara and Oshima44, Reference Rausch, Weisberg, Vardhana and Tortoriello45), as is GLUT1 protein in ob/ob mice though not in the diet-induced obese model(Reference Rausch, Weisberg, Vardhana and Tortoriello45). Thus the expression and protein content of a key hypoxia marker are generally increased in WAT in obese mice. Importantly, increased GLUT1 gene expression in ob/ob mice appears selective to WAT, there being no change in GLUT1 mRNA level in skeletal muscle of these obese mutants, consistent with local rather than generalised hypoxia(Reference Ye, Gao, Yin and He40). In parallel with the increased GLUT1 protein in WAT of diet-induced obese and KKAy mice is an increased lactate concentration in the tissue, which again was not evident in skeletal muscle(Reference Hosogai, Fukuhara and Oshima44).

The expression of a number of candidate genes, particularly those related to the inflammatory response, have been screened in WAT of the obese models described above with a view to determining whether hypoxia may underpin any such changes. Increased levels of the mRNA encoding IL-1β, IL-6, leptin, macrophage migration inhibitory factor (MIF), matrix metalloproteinase 2, PAI-1, transforming growth factor-β and TNF-α were each found to be increased, while adiponectin mRNA level is decreased(Reference Ye, Gao, Yin and He40, Reference Hosogai, Fukuhara and Oshima44). Many of these differences have, of course, been widely reported previously, especially the increased expression of leptin, IL-6 and TNF-α, and the reduced adiponectin expression. They have not, however, been linked to hypoxia per se. Interestingly, the expression of the angiogenic factor VEGF, which is well recognised as a hypoxia-sensitive gene, was not elevated in ob/ob mice and only slightly raised in diet-induced obesity(Reference Ye, Gao, Yin and He40).

Changes in the expression of key inflammation-related adipokines in obesity in association with hypoxia do not indicate, of course, a causal link. Indeed, in the case of adipokines such as leptin and adiponectin a number of factors which can modulate expression and which may be altered in obesity are well recognised. These include insulin levels and sympathetic activity, since insulin and β-adrenoceptor agonists can influence the expression of these key adipokines(Reference Leroy, Dessolin, Villageois, Moon, Friedman, Ailhaud and Dani46Reference Trayhurn, Duncan, Hoggard and Rayner48).

Cell-culture studies: hypoxia-sensitive genes in adipocytes

The effect of hypoxia on the expression and secretion of inflammation-related adipokine genes has been directly examined in adipocytes in culture, both murine and human. The presence of immunoreactive HIF-1α and its induction by hypoxia, or hypoxia mimetics, has been demonstrated in several cell-culture studies employing either the 3T3-F442A or 3T3-L1 adipocyte clonal cell lines(Reference Lolmède, Durand de Saint Front, Galitzky, Lafontan and Bouloumié21, Reference Chen, Lam, Wang, Wu, Lam, Shen, Wong, Hoo, Zhang and Xu49, Reference Segawa, Fukuhara and Hosogai50). Lolmède et al. (Reference Lolmède, Durand de Saint Front, Galitzky, Lafontan and Bouloumié21) in a pioneering report first showed that the production of pro-angiogenic factors such as VEGF, leptin and matrix metalloproteinases is increased in 3T3-F442A adipocytes in response to low O2 tension (5 % O2) or chemical hypoxia. Hypoxia-induced expression of VEGF had also been shown earlier in isolated rat omental adipocytes(Reference Zhang, Magovern, Mack, Budenbender, Ko and Rosengart51).

These observations were followed in 2006 by a report with 3T3-L1 cells that PAI-1 expression and release are augmented by hypoxia and hypoxia mimetics(Reference Chen, Lam, Wang, Wu, Lam, Shen, Wong, Hoo, Zhang and Xu49). Importantly, this study also demonstrated that adiponectin expression and release were inhibited in parallel with the increase in PAI-1, and that the changes were independent of reactive oxygen species. This down-regulation of adiponectin gene expression by hypoxia in 3T3-L1 adipocytes has subsequently been confirmed(Reference Hosogai, Fukuhara and Oshima44), and occurs also in human adipocytes(Reference Wang, Wood and Trayhurn52). Further screening studies on murine adipocytes, both primary and 3T3-L1, have indicated that the expression of a number of genes is stimulated by hypoxia (1 % O2), including GLUT1, MIF, matrix metalloproteinase 9, PAI-1, TNF-α and VEGF(Reference Ye, Gao, Yin and He40, Reference Hosogai, Fukuhara and Oshima44), as illustrated in Fig. 2. However, the changes observed in mRNA levels are rather small, at approximately 1·5–4-fold compared with the controls, in normoxia, and protein secretion (amount in the case of GLUT1) was not measured in these studies.

Fig. 2 Hypoxia-sensitive gene expression in adipocytes. The diagram shows those genes whose expression have been reported to be up-regulated ( ↑ ) or down-regulated ( ↓ ) in adipocytes (murine or human) in cell culture by hypoxia. Genes where changes in the protein, or its activity, have also been documented are shown in red. Where there are conflicting results, as with TNF-α, the gene has not been included. Names and abbreviations are those that are commonly used, rather than the formal gene name. Angptl4, angiopoietin-like protein 4; Hemox, haeme oxygenase 1; MIF, macrophage migration inhibitory factor; MMP, matrix metalloproteinase; MT-3, metallothionein-3; PAI-1, plasminogen activator inhibitor-1; PDK, pyruvate dehydrogenase kinase-1; VEGF, vascular endothelial growth factor.

Visfatin (or pre-B-cell colony-enhancing factor) gene expression, which was originally proposed as an insulin mimetic and to be pro-angiogenic, has additionally been shown to be induced by hypoxia in 3T3-L1 cells; however, the effects recorded are again modest(Reference Segawa, Fukuhara and Hosogai50). Studies on MCF7 breast cancer cells have indicated that visfatin gene expression is directly mediated by HIF-1α(Reference Bae, Kim, Kim, Kim, Koo, Jang, Yun, Yoo and Bae53). Apelin, which has cardioprotective properties, has also been shown to be hypoxia sensitive in 3T3-L1 adipocytes, as in cardiomyocytes(Reference Ronkainen, Ronkainen, Hanninen, Leskinen, Ruas, Pereira, Poellinger, Vuolteenaho and Tavi54), through the mediation of HIF-1α, both cellular mRNA levels and protein in the medium being increased by low O2 tension and hypoxia mimetics(Reference Glassford, Yue, Sheikh, Chun, Zarafshar, Chan, Reaven, Quertermous and Tsao55).

In the case of humans, expression of the HIF-1α gene has been reported in WAT, the level of the mRNA being increased in obese subjects(Reference Cancello, Henegar and Viguerie56). HIF-1α mRNA is evident in both isolated adipocytes and macrophages, the relative level being much greater in the latter(Reference Cancello, Henegar and Viguerie56). Increased HIF-1α mRNA levels in WAT in obese humans is not, however, necessarily evidence that there is hypoxia in the tissue in human obesity; in contrast to the majority of situations where mRNA and protein levels change in parallel, in our experience while the amount of HIF-1α protein increases in adipocytes in cell culture in response to hypoxia, mRNA levels may actually fall.

Our own studies on the effects of hypoxia on adipokine production have focused directly on human adipocytes differentiated from preadipocytes in primary culture. Immunoreactive HIF-1α protein is present in cultured human adipocytes, the level increasing rapidly and substantially (up to 8-fold in 4 h) with exposure to either 1 % O2 or to CoCl2(Reference Wang, Wood and Trayhurn52). The level of 1 % O2, which is a level of oxygenation widely used in in vitro studies of hypoxia, is equivalent to a partial pressure of O2 of 7·6 mmHg, and this is close to that observed in WAT in ob/ob mice(Reference Ye, Gao, Yin and He40). GLUT1 gene expression also increases, indicating that the cells respond to the hypoxia-induced rise in HIF-1α.

A candidate gene approach has been used to examine the effects of hypoxia on the expression of key inflammation-related adipokines in human adipocytes(Reference Wang, Wood and Trayhurn52). Hypoxia induces the expression of several inflammation-related adipokines; these include angiopoietin-like protein 4 (also known as fasting-induced adipose factor), IL-6, leptin, MIF, PAI-1 and VEGF(Reference Wang, Wood and Trayhurn52). The secretion of IL-6, leptin, MIF and VEGF into the medium is also increased by hypoxia (the other adipokines were not measured). In contrast, the expression and secretion of adiponectin, on the other hand, were inhibited by low O2 tension in human adipocytes, as in 3T3-L1 cells, but the effect was modest(Reference Wang, Wood and Trayhurn52). The expression of the acute-phase protein haptoglobin was also decreased under hypoxic conditions.

In array studies on human adipocytes, using a hypoxia signalling pathway PCR array (SuperArray), the expression of one particular gene – metallothionein (MT)-3 – was found to be very highly induced by hypoxia(Reference Wang, Wood and Trayhurn57). MT-3 mRNA levels increased > 600-fold in 24 h, with a > 100-fold increase by just 60 min after exposure of the adipocytes to 1 % O2. This response is not general to the MTs, since expression of the MT-2A gene, which we have previously shown occurs in human adipocytes(Reference Do, Nam, Hong, Kim, Duncan, Beattie and Trayhurn58), is essentially unaltered by hypoxia(Reference Wang, Wood and Trayhurn57). The functional significance of MT-3 expression by hypoxia in adipocytes is unclear, but previous work has shown that MT-3 is highly hypoxia inducible in human astrocytes where it is suggested to be protective against hypoxic damage(Reference Tanji, Irie, Uchida, Mori, Satoh, Mizushima and Wakabayashi59).

Overall, the cell-culture observations are generally consistent with hypoxia leading to an increase in the production of inflammation-related adipokines, both in murine and human adipocytes, this being mediated through HIF-1α as indicated by the studies with the hypoxia mimetics. The hypoxia-induced increase in the expression of the major angiogenic factor VEGF is consistent with a stimulation of angiogenesis. Other adipokines have also been implicated in angiogenesis, including angiopoietin-like protein 4 and leptin(Reference Sierra-Honigmann, Nath and Murakami60Reference Le Jan, Amy and Cazes63). The inhibition of adiponectin production and release by hypoxia demonstrated by the cell-culture studies is of particular significance since this adipokine has important anti-inflammatory and insulin-sensitising actions(Reference Ouchi, Kihara and Arita64Reference Yamauchi, Kamon and Waki67). It has been suggested that hypoxia may lead to a decrease in adiponectin production in adipocytes via the induction of endoplasmic reticulum stress(Reference Hosogai, Fukuhara and Oshima44).

Beyond adipokines: hypoxia and other adipocyte cellular processes

Hypoxia has been considered so far in terms of adipokine synthesis, reflecting the importance of these factors in WAT physiology. However, low O2 tension would be expected to impact on adipocyte function in multiple ways and two particular areas have been explored – cell differentiation and glucose utilisation. Several studies have examined the effects of hypoxia on adipocyte differentiation using various cell systems, including 3T3-L1 preadipocytes and bone marrow stromal cells. In all but one case, hypoxia or the CoCl2 mimetic inhibited differentiation(Reference Carriere, Carmona, Fernandez, Rigoulet, Wenger, Penicaud and Casteilla20, Reference Yun, Maecker, Johnson and Giaccia68Reference Lin, Lee and Yun71). However, a study employing mouse bone marrow stromal cells suggested that hypoxia accelerates differentiation into adipocytes(Reference Ren, Cao and Zhao72). Whether this different response is a reflection of the specific cell system, or the precise culture conditions, is unclear. Our own exploratory studies on human preadipocytes suggest that adipocyte differentiation in these cells is indeed inhibited by hypoxia (B Wang, IS Wood and P Trayhurn, unpublished results).

As described above, hypoxia leads to an increase in GLUT1 gene expression in white adipocytes, and this appears to be HIF-1 dependent in that incubation with CoCl2 under normoxic conditions leads to an increased GLUT1 mRNA level(Reference Wang, Wood and Trayhurn52). White adipocytes express several of the fourteen members of the facilitative GLUT gene family (GLUT1–GLUT12, HMIT, GLUT14; gene name SLC2A), including the insulin-sensitive transporter, GLUT4(Reference Wood and Trayhurn73, Reference Wood, Hunter and Trayhurn74). Exposure of human adipocytes to hypoxia for up to 24 h has been shown to lead to increased expression of the GLUT3 and GLUT5 genes, as well as of GLUT1, while GLUT4, GLUT10 and GLUT12 expression remains unchanged(Reference Wood, Wang, Lorente-Cebrián and Trayhurn75). The increase in GLUT1 mRNA level in hypoxia is paralleled by a substantial (10-fold) increase in the cellular level of GLUT1 protein(Reference Wood, Wang, Lorente-Cebrián and Trayhurn75). However, despite the increase in GLUT5 mRNA, GLUT5 protein level is unchanged, while GLUT3 protein could not be detected.

Importantly, functional studies with 2-deoxy-d-glucose indicate that glucose uptake by human adipocytes is strongly stimulated by hypoxia, presumably as a consequence of the increased amount of GLUT1(Reference Wood, Wang, Lorente-Cebrián and Trayhurn75). Increased glucose utilisation by adipocytes in response to hypoxia is, of course, a reflection of the Pasteur effect. It seems probable from these results that the metabolic activity of adipocytes is widely influenced by hypoxia. Certainly, glycolysis would be expected to be greatly stimulated and the utilisation of other nutrients may well also change.

Of particular significance is the possibility that insulin sensitivity may alter, either acutely or chronically, in response to hypoxia. This has been demonstrated in in vivo studies under conditions such as high altitude (particularly with acute exposure) and with obstructive sleep apnoea syndrome(Reference Larsen, Hansen, Olsen, Galbo and Dela76, Reference Oltmanns, Gehring, Rudolf, Schultes, Rook, Schweiger, Born, Fehm and Peters77). Intermittent hypoxia is commonly used to simulate obstructive sleep apnoea syndrome in experimental models and has been shown to induce insulin resistance(Reference Polotsky, Li, Punjabi, Rubin, Smith, Schwartz and O'Donnell78, Reference Iiyori, Alonso, Li, Sanders, Garcia-Ocana, O'Doherty, Polotsky and O'Donnell79). Although there is some evidence for insulin resistance following hypoxia in certain tissues, such as skeletal muscle, the overall picture is unclear(Reference Iiyori, Alonso, Li, Sanders, Garcia-Ocana, O'Doherty, Polotsky and O'Donnell79, Reference Gulve, Ren, Marshall, Gao, Hansen, Holloszy and Mueckler80). Specific studies are needed on whether local hypoxia leads to a reduction in insulin sensitivity in adipocytes, perhaps through the translocation of GLUT4 between the intracellular storage sites and the plasma membrane or by alterations in the insulin signalling pathway.

Hypoxia and the other cells in adipose tissue

The effects of hypoxia on WAT function have been discussed in terms of adipocytes, reflecting the fact that these are the cells that are characteristic of adipose tissue and which greatly enlarge in obesity. Adipocytes generally account, however, for no more than 50 % of the total cell content of WAT, the exact proportion varying between depots. The other cells include preadipocytes, endothelial cells, mast cells and macrophages, and it is argued from in vitro studies that adipocytes are only minor contributors compared with the non-adipocyte fraction in the production and release of many inflammatory cytokines and chemokines from WAT(Reference Fain, Madan, Hiler, Cheema and Bahouth81, Reference Fain82). However, the precise contribution of adipocytes, macrophages and other cells to the production of specific adipokines (other than leptin and adiponectin which essentially are produced in WAT only by mature adipocytes) is difficult to assess quantitatively. For example, is not certain whether the process of preparing and subsequent incubation of the different cell fractions alters their relative rate of production and release of various adipokines. In addition, large adipocytes show the highest rates of production of inflammatory adipokines(Reference Skurk, Alberti-Huber, Herder and Hauner25), but because of their greater fragility they are likely to be under-represented, or absent, from most cell fractionations.

Macrophages have attracted considerable recent attention in adipose tissue biology following the discovery that they infiltrate the tissue in obesity and appear to play a substantial role in the inflammatory process(Reference Xu, Barnes and Yang13, Reference Weisberg, McCann, Desai, Rosenbaum, Leibel and Ferrante14). These immune cells are recognised to respond to hypoxia with changes in the production of key inflammatory mediators(Reference Lewis, Lee, Underwood, Harris and Lewis83Reference Oda, Hirota and Nishi85). This has been directly examined recently in the context of macrophage involvement in adipose tissue function, with the induction of IL-1β, IL-6, MIF, TNF-α and VEGF gene expression under hypoxia being evident in peritoneal macrophages(Reference Ye, Gao, Yin and He40). Given the importance attributed to macrophages in the inflammatory response in WAT in obesity, it is of particular interest that immunohistochemical evidence suggests that F4/80+ macrophages are located primarily in the areas of the tissue exhibiting hypoxia(Reference Rausch, Weisberg, Vardhana and Tortoriello45). This suggests that there is a link between pockets of inflammation within WAT and the recruitment of macrophages into these same areas of the tissue through the hypoxia-induced production of inflammation-related adipokines.

The preadipocytes within WAT also exhibit major inflammatory responses, expressing and secreting a range of inflammation-related adipokines, including IL-1β, IL-6, IL-8, monocyte chemoattractant protein-1, MIF and TNF-α(Reference Cousin, Munoz, Andre, Fontanilles, Dani, Cousin, Laharrague, Casteilla and Penicaud86Reference Chung, LaPoint, Martinez, Kennedy, Boysen Sandberg and McIntosh91). Indeed, preadipocytes (human) are highly responsive to lipopolysaccharide challenge and it is suggested that they may play a pivotal role in the inflammatory response within WAT(Reference Chung, LaPoint, Martinez, Kennedy, Boysen Sandberg and McIntosh91). Studies on the effects of hypoxia on preadipocytes have focused on adipogenesis, as described above, rather than on the inflammatory response or general metabolic function. We have, however, recently found some similar responses to hypoxia in human preadipocytes as in adipocytes – together with some differences (B Wang, IS Wood and P Trayhurn, unpublished results). The similarities include increases in GLUT1 gene expression, and in VEGF expression and secretion. In contrast to mature adipocytes, however, hypoxia did not lead to major changes in preadipocytes in the expression of either IL-6 or angiopoietin-like protein 4.

Conclusions and implications

It is evident that hypoxia can influence the function of both adipocytes and adipose tissue. In particular, the expression and secretion of several key adipokines linked to inflammation are altered by hypoxia; indeed, the increase in leptin and reduction in adiponectin in obesity may be partly a direct result of hypoxia within WAT. While the changes in the production of inflammation-related adipokines are generally consistent with the original hypoxia hypothesis(Reference Trayhurn and Wood4), some major inflammatory adipokines, particularly TNF-α, may not be up-regulated under hypoxic conditions, at least in human adipocytes(Reference Wang, Wood and Trayhurn52). However, even if low O2 tension proves not to be a critical factor in initiating the inflammatory response in WAT, it is nonetheless likely to impact broadly on adipocytes and on adipose tissue.

We suggest that the hypoxic environment in which current evidence suggests enlarged adipocytes exist needs to be taken into account in studies on WAT. Thus the possibility that most metabolic systems, or the response to particular agents or factors, might be modified (directly or indirectly) by hypoxia should be considered. Hypoxia as a factor in adipose tissue biology has been viewed exclusively in terms of obesity, but in principle it could underlie other situations in which there is a marked increase in adipose tissue mass. For example, hypoxia may well occur in WAT during the physiological fattening that can take place in late pregnancy, and during the pre-hibernatory and pre-migratory period in those species which hibernate or migrate, respectively. Hypoxia, at least initially, could also occur in WAT on acute cold exposure as blood is channelled to brown adipose tissue and skeletal muscle for the generation of thermoregulatory heat by non-shivering and shivering mechanisms.

Finally, targeting the hypoxia signalling pathway, whether through HIF-1α itself or another key intermediate, may provide a novel route for the treatment of obesity-associated diseases, as noted previously(Reference Hosogai, Fukuhara and Oshima44, Reference Chen, Lam, Wang, Wu, Lam, Shen, Wong, Hoo, Zhang and Xu49). Directing any potential such treatment specifically to adipose tissue, and more particularly to the adipocyte, may alleviate many of these disorders.

Acknowledgements

We are grateful to the Biotechnology and Biological Sciences Research Council (BBSRC UK) for funding our studies on hypoxia in adipose tissue.

The authors declare that they have no conflicts of interest.

P. T. wrote the draft of the article, with subsequent additions and revisions from B. W. and I. S. W.; P. T. prepared the final version.

References

1Trayhurn, 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
2Rajala, MW & Scherer, PE (2003) The adipocyte – at the crossroads of energy homeostasis, inflammation, and atherosclerosis. Endocrinology 144, 37653773.CrossRefGoogle ScholarPubMed
3Kershaw, EE & Flier, JS (2004) Adipose tissue as an endocrine organ. J Clin Endocrinol Metab 89, 25482556.CrossRefGoogle ScholarPubMed
4Trayhurn, P & Wood, IS (2004) Adipokines: inflammation and the pleiotropic role of white adipose tissue. Br J Nutr 92, 347355.CrossRefGoogle ScholarPubMed
5Rosen, ED & Spiegelman, BM (2006) Adipocytes as regulators of energy balance and glucose homeostasis. Nature 444, 847853.CrossRefGoogle ScholarPubMed
6Zhang, YY, Proenca, R, Maffei, M, Barone, M, Leopold, L & Friedman, JM (1994) Positional cloning of the mouse obese gene and its human homolog. Nature 372, 425432.CrossRefGoogle Scholar
7Cook, KS, Min, HY, Johnson, D, Chaplinsky, RJ, Flier, JS, Hunt, CR & Spiegelman, BM (1987) Adipsin: a circulating serine protease homolog secreted by adipose tissue and sciatic nerve. Science 237, 402405.CrossRefGoogle ScholarPubMed
8Flier, JS, Cook, KS, Usher, P & Spiegelman, BM (1987) Severely impaired adipsin expression in genetic and acquired obesity. Science 237, 405408.CrossRefGoogle ScholarPubMed
9Hotamisligil, GS, Shargill, NS & Spiegelman, BM (1993) Adipose expression of tumor necrosis factor-α – direct role in obesity-linked insulin resistance. Science 259, 8791.CrossRefGoogle ScholarPubMed
10Hotamisligil, GS (2006) Inflammation and metabolic disorders. Nature 444, 860867.CrossRefGoogle ScholarPubMed
11Wellen, KE & Hotamisligil, GS (2005) Inflammation, stress, and diabetes. J Clin Invest 115, 11111119.CrossRefGoogle Scholar
12Arita, 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
13Xu, 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
14Weisberg, SP, McCann, D, Desai, M, Rosenbaum, M, Leibel, RL & Ferrante, AW Jr (2003) Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest 112, 17961808.CrossRefGoogle ScholarPubMed
15Yudkin, JS (2003) Adipose tissue, insulin action and vascular disease: inflammatory signals. Int J Obes 27, Suppl. 3, S25S28.CrossRefGoogle ScholarPubMed
16Ozcan, U, Cao, Q, Yilmaz, E, Lee, AH, Iwakoshi, NN, Ozdelen, E, Tuncman, G, Görgün, C, Glimcher, LH & Hotamisligil, GS (2004) Endoplasmic reticulum stress links obesity, insulin action, and type 2 diabetes. Science 306, 457461.CrossRefGoogle ScholarPubMed
17Gregor, MF & Hotamisligil, GS (2007) Adipocyte biology. Adipocyte stress: the endoplasmic reticulum and metabolic disease. J Lipid Res 48, 19051914.CrossRefGoogle ScholarPubMed
18Houstis, N, Rosen, ED & Lander, ES (2006) Reactive oxygen species have a causal role in multiple forms of insulin resistance. Nature 440, 944948.CrossRefGoogle ScholarPubMed
19Koumenis, C, Naczki, C, Koritzinsky, M, Rastani, S, Diehl, A, Sonenberg, N, Koromilas, A & Wouters, BG (2002) Regulation of protein synthesis by hypoxia via activation of the endoplasmic reticulum kinase PERK and phosphorylation of the translation initiation factor eIF2α. Mol Cell Biol 22, 74057416.CrossRefGoogle ScholarPubMed
20Carriere, A, Carmona, M-C, Fernandez, Y, Rigoulet, M, Wenger, RH, Penicaud, L & Casteilla, L (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
21Lolmède, K, Durand de Saint Front, V, Galitzky, J, Lafontan, M & Bouloumié, A (2003) Effects of hypoxia on the expression of proangiogenic factors in differentiated 3T3-F442A adipocytes. Int J Obes 27, 11871195.CrossRefGoogle ScholarPubMed
22Virtanen, KA, Lonnroth, P, Parkkola, R, et al. (2002) Glucose uptake and perfusion in subcutaneous and visceral adipose tissue during insulin stimulation in nonobese and obese humans. J Clin Endocrinol Metab 87, 39023910.CrossRefGoogle ScholarPubMed
23Kabon, B, Nagele, A, Reddy, D, Eagon, C, Fleshman, JW, Sessler, DI & Kurz, A (2004) Obesity decreases perioperative tissue oxygenation. Anesthesiology 100, 274280.CrossRefGoogle ScholarPubMed
24Karpe, F, Fielding, BA, Ilic, V, Macdonald, IA, Summers, LKM & Frayn, KN (2002) Impaired postprandial adipose tissue blood flow response is related to aspects of insulin sensitivity. Diabetes 51, 24672473.CrossRefGoogle ScholarPubMed
25Skurk, T, Alberti-Huber, C, Herder, C & Hauner, H (2007) Relationship between adipocyte size and adipokine expression and secretion. J Clin Endocrinol Metab 92, 10231033.CrossRefGoogle ScholarPubMed
26Brahimi-Horn, MC & Pouysségur, J (2007) Oxygen, a source of life and stress. FEBS Lett 581, 35823591.CrossRefGoogle ScholarPubMed
27Folkman, J, Hahnfeldt, P & Hlatky, L (2000) Cancer: looking outside the genome. Nat Rev Mol Cell Biol 1, 7679.CrossRefGoogle ScholarPubMed
28Gatenby, RA & Gillies, RJ (2004) Why do cancers have high aerobic glycolysis? Nat Rev Cancer 4, 891899.CrossRefGoogle ScholarPubMed
29Semenza, GL (2003) Targeting HIF-1 for cancer therapy. Nat Rev Cancer 3, 721732.CrossRefGoogle ScholarPubMed
30Erecinska, M & Silver, IA (2001) Tissue oxygen tension and brain sensitivity to hypoxia. Respir Physiol 128, 263276.CrossRefGoogle ScholarPubMed
31Fleischmann, E, Kurz, A, Niedermayr, M, Schebesta, K, Kimberger, O, Sessler, DI, Kabon, B & Prager, G (2005) Tissue oxygenation in obese and non-obese patients during laparoscopy. Obes Surg 15, 813819.CrossRefGoogle ScholarPubMed
32Di Girolamo, M, Skinner, NS Jr, Hanley, HG & Sachs, RG (1971) Relationship of adipose tissue blood flow to fat cell size and number. Am J Physiol 220, 932937.CrossRefGoogle ScholarPubMed
33Thurlby, PL & Trayhurn, P (1980) Regional blood flow in genetically obese (ob/ob) mice: the importance of brown adipose tissue to the reduced energy expenditure on non-shivering thermogenesis. Pflügers Arch 385, 193201.CrossRefGoogle Scholar
34West, DB, Prinz, WA, Francendese, AA & Greenwood, MRC (1987) Adipocyte blood flow is decreased in obese Zucker rats. Am J Physiol Reg Integr Comp Physiol 253, R228R233.CrossRefGoogle ScholarPubMed
35Semenza, GL (2001) HIF-1 and mechanisms of hypoxia sensing. Curr Opin Cell Biol 13, 167171.CrossRefGoogle ScholarPubMed
36Höpfl, G, Ogunshola, O & Gassmann, M (2004) HIFs and tumors – causes and consequences. Am J Physiol Reg Integr Comp Physiol 286, R608R623.CrossRefGoogle ScholarPubMed
37Lopez-Barneo, J, del Toro, R, Levitsky, KL, Chiara, MD & Ortega-Saenz, P (2004) Regulation of oxygen sensing by ion channels. J Appl Physiol 96, 11871195.CrossRefGoogle ScholarPubMed
38Cummins, EP & Taylor, CT (2005) Hypoxia-responsive transcription factors. Pflügers Arch 450, 363371.CrossRefGoogle ScholarPubMed
39Rocha, S (2007) Gene regulation under low oxygen: holding your breath for transcription. Trends Biochem Sci 32, 389397.CrossRefGoogle ScholarPubMed
40Ye, J, Gao, Z, Yin, J & He, Q (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
41Ivan, M, Kondo, K, Yang, H, Kim, W, Valiando, J, Ohh, M, Salic, A, Asara, JM, Lane, WS & Kaelin, WG Jr (2001) HIFα targeted for VHL-mediated destruction by proline hydroxylation: implications for O2 sensing. Science 292, 464468.CrossRefGoogle ScholarPubMed
42Jaakkola, P, Mole, DR, Tian, YM, et al. (2001) Targeting of HIF-α to the von Hippel–Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation. Science 292, 468472.CrossRefGoogle Scholar
43Mueckler, M, Caruso, C, Baldwin, SA, Panico, M, Blench, I, Morris, HR, Allard, WJ, Lienhard, GE & Lodish, HF (1985) Sequence and structure of a human glucose transporter. Science 229, 941945.CrossRefGoogle ScholarPubMed
44Hosogai, N, Fukuhara, A, Oshima, K, et al. (2007) Adipose tissue hypoxia in obesity and its impact on adipocytokine dysregulation. Diabetes 56, 901911.CrossRefGoogle ScholarPubMed
45Rausch, ME, Weisberg, S, Vardhana, P & Tortoriello, DV (2007) Obesity in C57BL/6J mice is characterized by adipose tissue hypoxia and cytotoxic T-cell infiltration. Int J Obes (Lond) (Epublication ahead of print version 25 September 2007).Google ScholarPubMed
46Leroy, P, Dessolin, S, Villageois, P, Moon, BC, Friedman, JM, Ailhaud, G & Dani, C (1996) Expression of ob gene in adipose-cells – regulation by insulin. J Biol Chem 271, 23652368.CrossRefGoogle Scholar
47Fasshauer, M, Klein, J, Neumann, S, Eszlinger, M & Paschke, R (2001) Adiponectin gene expression is inhibited by β-adrenergic stimulation via protein kinase A in 3T3-L1 adipocytes. FEBS Lett 507, 142146.CrossRefGoogle ScholarPubMed
48Trayhurn, P, Duncan, JS, Hoggard, N & Rayner, DV (1998) Regulation of leptin production: a dominant role for the sympathetic nervous system? Proc Nutr Soc 57, 413419.CrossRefGoogle ScholarPubMed
49Chen, B, Lam, KSL, Wang, Y, Wu, D, Lam, MC, Shen, J, Wong, L, Hoo, RL, Zhang, J & Xu, A (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
50Segawa, K, Fukuhara, A, Hosogai, N, et al. (2006) Visfatin in adipocytes is upregulated by hypoxia through HIF1α-dependent mechanism. Biochem Biophys Res Commun 349, 875882.CrossRefGoogle ScholarPubMed
51Zhang, QX, Magovern, CJ, Mack, CA, Budenbender, KT, Ko, W & Rosengart, TK (1997) Vascular endothelial growth factor is the major angiogenic factor in omentum: mechanism of the omentum-mediated angiogenesis. J Surg Res 67, 147154.CrossRefGoogle ScholarPubMed
52Wang, B, Wood, IS & Trayhurn, P (2007) Dysregulation of the expression and secretion of inflammation-related adipokines by hypoxia in human adipocytes. Pflügers Arch 455, 479492.CrossRefGoogle ScholarPubMed
53Bae, S-K, Kim, S-R, Kim, JG, Kim, JY, Koo, TH, Jang, H-O, Yun, I, Yoo, M-A & Bae, M-K (2006) Hypoxic induction of human visfatin gene is directly mediated by hypoxia-inducible factor-1. FEBS Lett 580, 41054113.CrossRefGoogle ScholarPubMed
54Ronkainen, VP, Ronkainen, JJ, Hanninen, SL, Leskinen, H, Ruas, JL, Pereira, T, Poellinger, L, Vuolteenaho, O & Tavi, P (2007) Hypoxia inducible factor regulates the cardiac expression and secretion of apelin. FASEB J 21, 18211830.CrossRefGoogle ScholarPubMed
55Glassford, AJ, Yue, P, Sheikh, AY, Chun, HJ, Zarafshar, S, Chan, DA, Reaven, GM, Quertermous, T & Tsao, PS (2007) HIF-1 regulates hypoxia- and insulin-induced expression of apelin in adipocytes. Am J Physiol Endocrinol Metab 293, E1590E1596.CrossRefGoogle ScholarPubMed
56Cancello, R, Henegar, C, Viguerie, N, et al. (2005) Reduction of macrophage infiltration and chemoattractant gene expression changes in white adipose tissue of morbidly obese subjects after surgery-induced weight loss. Diabetes 54, 22772286.CrossRefGoogle ScholarPubMed
57Wang, 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
58Do, MS, Nam, SY, Hong, SE, Kim, KW, Duncan, JS, Beattie, JH & Trayhurn, P (2002) Metallothionein gene expression in human adipose tissue from lean and obese subjects. Horm Metab Res 34, 348351.CrossRefGoogle ScholarPubMed
59Tanji, K, Irie, Y, Uchida, Y, Mori, F, Satoh, K, Mizushima, Y & Wakabayashi, K (2003) Expression of metallothionein-III induced by hypoxia attenuates hypoxia-induced cell death in vitro. Brain Res 976, 125129.CrossRefGoogle ScholarPubMed
60Sierra-Honigmann, MR, Nath, AK, Murakami, C, et al. (1998) Biological action of leptin as an angiogenic factor. Science 281, 16831686.CrossRefGoogle ScholarPubMed
61Bouloumié, A, Drexler, HCA, Lafontan, M & Busse, R (1998) Leptin, the product of Ob gene, promotes angiogenesis. Circ Res 83, 10591066.CrossRefGoogle Scholar
62Zhu, H, Li, J, Qin, W, Yang, Y, He, X, Wan, D & Gu, J (2002) Cloning of a novel gene, ANGPTL4 and the functional study in angiogenesis. Zhonghua Yi Xue Za Zhi 82, 9499.Google ScholarPubMed
63Le Jan, S, Amy, C, Cazes, A, et al. (2003) Angiopoietin-like 4 is a proangiogenic factor produced during ischemia and in conventional renal cell carcinoma. Am J Pathol 162, 15211528.CrossRefGoogle ScholarPubMed
64Ouchi, N, Kihara, S, Arita, Y, et al. (1999) Novel modulator for endothelial adhesion molecules: adipocyte-derived plasma protein adiponectin. Circulation 100, 24732476.CrossRefGoogle ScholarPubMed
65Ouchi, N, Kihara, S, Arita, Y, et al. (2000) Adiponectin, an adipocyte-derived plasma protein, inhibits endothelial NF-κB signaling through a cAMP-dependent pathway. Circulation 102, 12961301.CrossRefGoogle ScholarPubMed
66Berg, AH, Combs, TP, Du, X, Brownlee, M & Scherer, PE (2001) The adipocyte-secreted protein Acrp30 enhances hepatic insulin action. Nat Med 7, 947953.CrossRefGoogle ScholarPubMed
67Yamauchi, T, Kamon, J, Waki, H, et al. (2001) The fat-derived hormone adiponectin reverses insulin resistance associated with both lipoatrophy and obesity. Nat Med 7, 941946.CrossRefGoogle ScholarPubMed
68Yun, Z, Maecker, HL, Johnson, RS & Giaccia, AJ (2002) Inhibition of PPARγ 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
69Zhou, S, Lechpammer, S, Greenberger, JS & Glowacki, J (2005) Hypoxia inhibition of adipocytogenesis in human bone marrow stromal cells requires transforming growth factor-β/Smad3 signaling. J Biol Chem 280, 2268822696.CrossRefGoogle ScholarPubMed
70Kim, KH, Song, MJ, Chung, J, Park, H & Kim, JB (2005) Hypoxia inhibits adipocyte differentiation in a HDAC-independent manner. Biochem Biophys Res Commun 333, 11781184.CrossRefGoogle Scholar
71Lin, Q, Lee, Y-J & Yun, Z (2006) Differentiation arrest by hypoxia. J Biol Chem 281, 3067830683.CrossRefGoogle ScholarPubMed
72Ren, H, Cao, Y, Zhao, Q, et al. (2006) Proliferation and differentiation of bone marrow stromal cells under hypoxic conditions. Biochem Biophys Res Commun 347, 1221.CrossRefGoogle ScholarPubMed
73Wood, IS & Trayhurn, P (2003) Glucose transporters (GLUT and SGLT): expanded families of sugar transport proteins. Br J Nutr 89, 39.CrossRefGoogle ScholarPubMed
74Wood, 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
75Wood, IS, Wang, B, Lorente-Cebrián, S & Trayhurn, P (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
76Larsen, JJ, Hansen, JM, Olsen, NV, Galbo, H & Dela, F (1997) The effect of altitude hypoxia on glucose homeostasis in men. J Physiol 504, 241249.CrossRefGoogle ScholarPubMed
77Oltmanns, KM, Gehring, H, Rudolf, S, Schultes, B, Rook, S, Schweiger, U, Born, J, Fehm, HL & Peters, A (2004) Hypoxia causes glucose intolerance in humans. Am J Respir Crit Care Med 169, 12311237.CrossRefGoogle ScholarPubMed
78Polotsky, VY, Li, J, Punjabi, NM, Rubin, AE, Smith, PL, Schwartz, AR & O'Donnell, CP (2003) Intermittent hypoxia increases insulin resistance in genetically obese mice. J Physiol 552, 253264.CrossRefGoogle ScholarPubMed
79Iiyori, N, Alonso, LC, Li, J, Sanders, MH, Garcia-Ocana, A, O'Doherty, RM, Polotsky, VY & O'Donnell, CP (2007) Intermittent hypoxia causes insulin resistance in lean mice independent of autonomic activity. Am J Respir Crit Care Med 175, 851857.CrossRefGoogle ScholarPubMed
80Gulve, EA, Ren, JM, Marshall, BA, Gao, J, Hansen, PA, Holloszy, JO & Mueckler, M (1994) Glucose transport activity in skeletal muscles from transgenic mice overexpressing GLUT1. Increased basal transport is associated with a defective response to diverse stimuli that activate GLUT4. J Biol Chem 269, 1836618370.CrossRefGoogle ScholarPubMed
81Fain, JN, Madan, AK, Hiler, ML, Cheema, P & Bahouth, SW (2004) Comparison of the release of adipokines by adipose tissue, adipose tissue matrix, and adipocytes from visceral and subcutaneous abdominal adipose tissues of obese humans. Endocrinology 145, 22732282.CrossRefGoogle ScholarPubMed
82Fain, 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
83Lewis, JS, Lee, JA, Underwood, JC, Harris, AL & Lewis, CE (1999) Macrophage responses to hypoxia: relevance to disease mechanisms. J Leukoc Biol 66, 889900.CrossRefGoogle ScholarPubMed
84Bosco, MC, Puppo, M, Pastorino, S, Mi, Z, Melillo, G, Massazza, S, Rapisarda, A & Varesio, L (2004) Hypoxia selectively inhibits monocyte chemoattractant protein-1 production by macrophages. J Immunol 172, 16811690.CrossRefGoogle ScholarPubMed
85Oda, 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
86Cousin, B, Munoz, O, Andre, M, Fontanilles, AM, Dani, C, Cousin, JL, Laharrague, P, Casteilla, L & Penicaud, L (1999) A role for preadipocytes as macrophage-like cells. FASEB J 13, 305312.CrossRefGoogle ScholarPubMed
87Gerhardt, CC, Romero, IA, Cancello, R, Camoin, L & Strosberg, AD (2001) Chemokines control fat accumulation and leptin secretion by cultured human adipocytes. Mol Cell Endocrinol 175, 8192.CrossRefGoogle ScholarPubMed
88Harkins, JM, Moustaid-Moussa, N, Chung, Y-J, Penner, KM, Pestka, JJ, North, CM & Claycombe, KJ (2004) Expression of interleukin-6 is greater in preadipocytes than in adipocytes of 3T3-L1 cells and C57BL/6J and ob/ob mice. J Nutr 134, 26732677.Google ScholarPubMed
89Skurk, T, Herder, C, Kraft, I, Muller-Scholze, S, Hauner, H & Kolb, H (2005) Production and release of macrophage migration inhibitory factor from human adipocytes. Endocrinology 146, 10061011.CrossRefGoogle ScholarPubMed
90Wang, B, Jenkins, JR & Trayhurn, P (2005) Expression and secretion of inflammation-related adipokines by human adipocytes differentiated in culture: integrated response to TNF-α. Am J Physiol Endocrinol Metab 288, E731E740.CrossRefGoogle ScholarPubMed
91Chung, S, LaPoint, K, Martinez, K, Kennedy, A, Boysen Sandberg, M & McIntosh, MK (2006) Preadipocytes mediate lipopolysaccharide-induced inflammation and insulin resistance in primary cultures of newly differentiated human adipocytes. Endocrinology 147, 53405351.CrossRefGoogle ScholarPubMed
92Braun, RD, Lanzen, JL, Snyder, SA & Dewhirst, MW (2001) Comparison of tumor and normal tissue oxygen tension measurements using OxyLite or microelectrodes in rodents. Am J Physiol Heart Circ Physiol 280, H2533H2544.CrossRefGoogle ScholarPubMed
93Yu, DY & Cringle, SJ (2005) Retinal degeneration and local oxygen metabolism. Exp Eye Res 80, 745751.CrossRefGoogle ScholarPubMed
Figure 0

Table 1 Oxygen levels in different tissues, including tissues where hypoxia is evident, as well as recent data on white adipose tissue from lean and obese mice

Figure 1

Fig. 1 Diagrammatic view of the molecular signalling response to hypoxia through the hypoxia-inducible factor (HIF) system. PHD, prolyl hydroxylase; FIH, factor inhibiting HIF-1; vHL, von Hippel–Lindau tumour suppressor protein; p300–CBP, p300–cAMP response element binding protein binding protein (transcriptional coactivator); HRE, hypoxia-response element; Ub, ubiquitin.

Figure 2

Fig. 2 Hypoxia-sensitive gene expression in adipocytes. The diagram shows those genes whose expression have been reported to be up-regulated ( ↑ ) or down-regulated ( ↓ ) in adipocytes (murine or human) in cell culture by hypoxia. Genes where changes in the protein, or its activity, have also been documented are shown in red. Where there are conflicting results, as with TNF-α, the gene has not been included. Names and abbreviations are those that are commonly used, rather than the formal gene name. Angptl4, angiopoietin-like protein 4; Hemox, haeme oxygenase 1; MIF, macrophage migration inhibitory factor; MMP, matrix metalloproteinase; MT-3, metallothionein-3; PAI-1, plasminogen activator inhibitor-1; PDK, pyruvate dehydrogenase kinase-1; VEGF, vascular endothelial growth factor.