Vous êtes sur la page 1sur 11

http://informahealthcare.

com/arp
ISSN: 1381-3455 (print), 1744-4160 (electronic)

Arch Physiol Biochem, 2014; 120(1): 1–11


! 2014 Informa UK Ltd. DOI: 10.3109/13813455.2013.838971

REVIEW ARTICLE

A vascular piece in the puzzle of adipose tissue dysfunction:


mechanisms and consequences
Tiago Rodrigues, Paulo Matafome, and Raquel Seiça
Archives of Physiology and Biochemistry Downloaded from informahealthcare.com by Cornell University on 12/28/14

Laboratory of Physiology, Faculty of Medicine, Institute of Biomedical Imaging and Life Sciences (IBILI), University of Coimbra, Portugal

Abstract Keywords
In the last years, several studies unravelled many aspects of adipose tissue pathophysiology in Adipocytokines, adipose tissue, angiogenesis,
metabolic diseases. Some studies suggested hypoxia as one of such aspects, despite the exact glycation, metabolism, microcirculation,
mechanisms and pathophysiological significance is still partially unknown. Adipose tissue was microvasculature, vasoactive factors
shown to be hypoxic in obesity, mainly resulting from adipocyte hypertrophy, leading to
increased activation of inflammatory pathways. In animal and cell models, hypoxia-induced History
inflammation was shown to lead to endocrine alterations and dysmetabolism. However, recent
evidences suggest that instead of a simple low oxygenation theory, adipose tissue Received 5 July 2013
microvasculature may be regulated by a series of factors, including vasoactive factors like Revised 21 August 2013
angiotensin II, angiogenesis and glycation, among others. This review summarizes the current Accepted 23 August 2013
knowledge about the role of these factors in the regulation of adipose tissue irrigation and the Published online 24 September 2013
functional consequences of adipose tissue microvascular dysfunction.
For personal use only.

Introduction decreased lipid esterification. As well, free fatty acids


inhibit insulin signalling through nuclear factor-kappa B
Obesity has reached epidemic proportions worldwide and
(NF-B)-dependent mechanisms (Guilherme et al., 2008;
abdominal obesity is a major risk factor for metabolic
Qatanani & Lazar, 2007). Adipose tissue inflammation leads
syndrome, cardiovascular diseases and ultimately to type 2
to increased circulating free fatty acids, which can accumulate
diabetes (Goossens, 2008; World Health Organization, 2000).
ectopically in muscle and liver. This may cause functional and
Besides the large number of cell types present in adipose
structural lesions in these tissues and possibly conduct to
tissue, it also has a large and dynamic vascular network,
systemic insulin resistance and ultimately to type 2 diabetes
which is in continuous re-modelling and is crucial for tissue
(Boden, 2011; Galic et al., 2010; Guilherme et al., 2008;
support during its expansion or regression (Lijnen, 2008;
Kaneto et al., 2010; Ozcan et al., 2004). Another consequence
Rajala & Scherer, 2003; Rutkowski et al., 2009). As a
of hypertrophy is that adipocyte’s volume may become higher
consequence of nutritional fluxes (higher in visceral adipose
than the oxygen diffusion distance, leading to local hypoxic
tissue), adipocytes continually accumulate lipids (triglycer-
regions (Hosogai et al., 2007; Trayhurn et al., 2008a). It has
ides) in lipid droplets, which may lead to hypertrophy and
metabolic dysregulation. On the other hand, hyperplasia may
been suggested that hypoxia development in adipose tissue 14
20
activates inflammatory pathways, causing insulin-resistance
be observed, resulting from increased pre-adipocyte differen-
and impaired lipid storage (Figures 1 and 2) (Galic et al.,
tiation. Hyperplasia is typically associated with better meta-
2010; Golay & Ybarra, 2005; Guilherme et al., 2008;
bolic regulation as it allows a better vascularization (Golay &
Rutkowski et al., 2009; Tilg & Moschen, 2006). The
Ybarra, 2005; Weyer et al., 2000).
activation of inflammatory mechanisms completely changes
Hypertrophy results in the accumulation of secondary
adipose tissue secretome. This compromises the expression of
products of lipid metabolism, causing progressive low-grade
anti-inflammatory adipokines and favours the secretion of
inflammation, insulin resistance and lipolysis. The regulation
pro-inflammatory, chemo-attractant and growth factors.
of substrate uptake by inflammatory mechanisms is a
Besides changing adipocyte metabolism, these products
physiological response aiming to decrease adipocyte
influence tissue angiogenesis and leukocyte recruitment,
volume. Inflammation strongly inhibits peroxisome prolifer-
causing an inflammatory feedback in the tissue (Guilherme
ating-activated receptor g (PPARg) activity, resulting in
et al., 2008; Trayhurn et al., 2008b; Vázquez-Vela et al.,
2008) (Figures 1 and 2). However, the in vivo regulation of
Correspondence: Paulo Matafome, Faculty of Medicine, Pole III of such mechanisms may be more complex than that previously
University of Coimbra, Subunit 1, 1st floor, Azinhaga de Santa Comba,
Celas, 3000-354 Coimbra, Portugal. Tel: +351239480014. Fax: demonstrated. Recently, Goossens and colleagues developed a
+351239480034. E-mail: paulomatafome@gmail.com method to measure in vivo oxygenation in adipose tissue.
2 T. Rodrigues et al. Arch Physiol Biochem, 2014; 120(1): 1–11
Archives of Physiology and Biochemistry Downloaded from informahealthcare.com by Cornell University on 12/28/14
For personal use only.

Figure 1. Events occurring in adipose tissue undergoing an inappropriate growth of the vascular network during tissue expansion. Adipocyte
hypertrophy and incorrect angiogenesis leads to hypoxia, activation of inflammatory pathways, adipocyte death and macrophage recruitment. Ang-2,
angiopoietin-2; CCR2, cytokines receptor; FFAs, free fatty acids; FFA-Sat, saturated free fatty acids; HIF-1, hypoxia inducible factor-1; ICAM,
intercellular adhesion molecule; IBa, NF-B inhibitor alpha; MCP-1, monocyte chemo-attractant protein-1; MMPs, matrix metalloproteinases;
NF-B, nuclear factor kappa b; PerA, Perilipin A; Tie-2, membrane receptor activated by angiopoietins; TLR4, toll-like receptor 4; TNF-a, Tumour
necrosis factor alpha; uPA, plasminogen activator urokinase-type; VEGF, vascular endothelial growth factor; VEGFR, VEGF receptor; VLDL,
very-low density lipoprotein.

They showed that decreased adipose tissue blood flow may Apparently, most of the mechanisms known to regulate
impair oxygen pressure in tissue. Nevertheless, the adipose systemic arterial tone also act on adipose tissue arterioles.
tissue of obese patients was hyperoxic, possibly resulting from These factors regulate blood flow in the tissue, although little
decreased metabolic activity (Goossens et al., 2011). attention has been devoted to this matter (Bagi et al., 2012).
Recent evidence supports the idea that instead of a simple More, microvascular dysfunction has been considered sec-
low oxygenation theory, adipose tissue microvasculature may ondary to adipose tissue dysfunction. Recent data suggest that
be regulated by a series of other factors (Scalia, 2013). microvascular dysfunction may in fact precede many of the
Archives of Physiology and Biochemistry Downloaded from informahealthcare.com by Cornell University on 12/28/14
For personal use only.

Figure 2. Mechanisms of inflammatory activation in adipocytes as a consequence of lipid accumulation and hypoxia. Activation of inflammatory
pathways leads to insulin resistance, lipolysis, PPARg down-regulation and expression of growth and chemo-attractant factors. AP1, adaptor protein-1;
cAMP, cyclic adenosine monophosphate; FATP, fatty acid binding protein; FFAs, free fatty acids; FFA-Sat, saturated free fatty acids; HDL, high
density lipoprotein; HIF-1, hypoxia inducible factor-1; HSL, hormone-sensitive lipase; IBa, NF-B inhibitor alpha; IRS-1, insulin receptor
substract-1; JNK, C-jun n-terminal kinase; LPL, lipoprotein lipase; MCP-1, monocyte chemoattractant protein-1; MMPs, matrix metalloproteinases;
NF-B, nuclear factor kappa b; PerA, perilipin A; PKC, protein kinase C; PO2, oxygen pressure; PPARg, peroxisome proliferating-activated receptor g;
Ser, serine; TLR4, toll-like receptor 4; TNF-a, Tumour necrosis factor alpha; uPA, plasminogen activator urokinase-type; VEGF, vascular endothelial
growth factor; VEGFR, VEGF receptor; VLDL, very-low density lipoprotein.
4 T. Rodrigues et al. Arch Physiol Biochem, 2014; 120(1): 1–11

alterations observed in adipose tissue in obesity. The impair- also a critic factor for its homeostasis. This is highly
ment of the mechanisms regulating blood flow in adipose regulated by the renin-angiotensin system, among other less-
tissue may influence its ability to expand correctly and thus studied vasoactive factors (Villela & Kramer-Aguiar, 2009).
may be associated with its dysfunction. The following Angiotensinogen is cleaved to Angiotensin I (AngI) by the
sections summarize the current knowledge about adipose enzyme renin, which is released from the kidney in
tissue microvascular lesions observed in obesity and how this consequence of low perfusion pressure. AngI is subsequently
can be implicated in altered tissue and systemic metabolism. cleaved to Angiotensin II (AngII) mainly by angiotensin
converting enzyme (ACE), but also by chymase and
Regulation of adipose tissue microvasculature cathepsin G (Negro, 2008; Weir, 2007). AngII receptors-1
During the embryonic development of adipose tissue, the (AT1) are the most expressed and are responsible for many
vascular network and perivascular extracellular matrix are of the pathological effects commonly associated to AngII
formed before the adipocytes. Angiogenesis and adipogenesis action, such as vasoconstriction, aldosterone production in
are mutually regulated, being each adipocyte surrounded by at the adrenal glands and salt and water retention in renal
least one blood vessel (Christiaens & Lijnen, 2010; Hausman tubules. On the other hand, angiotensin receptors-2 (AT2)
Archives of Physiology and Biochemistry Downloaded from informahealthcare.com by Cornell University on 12/28/14

& Richardson, 2004; Mannerås-Holm & Krook, 2012; seem to antagonize AT1 function (Steckelings et al., 2009,
Rutkowski et al., 2009; Zhang & Zhang, 2009). Expansion 2010; Weir, 2007).
or retraction of the microvasculature is permanently required. Angiotensinogen is mainly produced in the liver. However,
This plasticity is particularly important after adiposity its expression was also noticed in other tissues, namely
changes, in order to prevent the development of hypoxic adipose tissue. Although adipocytes produce low levels of
regions. In this context, adipocyte behaviour during tissue angiotensinogen, adipose tissue may become the second
growth depends on the type of vascular network alteration. higher source of angiotensinogen in obese individuals, due to
In case of adipocyte hyperplasia, neo-vascularization is the expansion of the fat mass. Angiotensinogen circulating
favoured, allowing adipocyte differentiation and regular levels are correlated with the body mass index (BMI) and
irrigation. However, if adipocytes are more hypertrophic, further increase in obese individuals after a sympathetic
existent capillaries suffer remodeling and dilation (Figures 1 stimulation (Kalupahana & Moustaid-Moussa, 2012a). In
and 2) (Christiaens & Lijnen, 2010; Mannerås-Holm & addition, angiotensinogen production in liver was observed to
Krook, 2012; Rutkowski et al., 2009). In such conditions, remain unchanged in models of obesity, suggesting a role of
For personal use only.

adipocyte diameter can reach 150–200 mm, although the adipose tissue in higher angiotensinogen production in
diffusion rate of oxygen is not higher than 100 mm (Gustafson, obesity (see a comprehensive review at Kalupahana &
2010; Trayhurn et al., 2008a; Wood et al., 2009). Moustaid-Moussa, 2012a). An animal model with angioten-
Lean individuals show a post-prandial increase of adipose sinogen expression only in adipocytes showed detectable
tissue irrigation and adipose arterioles are responsive to circulating levels, demonstrating that angiotensinogen pro-
angiotensin II (AngII, vasoconstrictor) and isoprenaline duced in adipose tissue have the ability to circulate and
(vasodilator). However, such events are not observed in change the arterial function (Kalupahana & Moustaid-
obese and type 2 diabetic patients (Chen et al., 2006; Moussa, 2012b). The fact that angiotensinogen is highly
Goossens et al., 2011; Tobin et al., 2010; Trayhurn et al., produced in adipose tissue suggests that renin-angiotensin
2008a; Wang et al., 2007; Wood et al., 2009). In fact, obese system (RAS) may strongly influence adipose tissue circula-
patients with regional hypoxia in adipose tissue do not show tion and physiology. This may have direct effects on
altered systemic oxygen pressure or haemoglobin concentra- endothelial cells and influence blood pressure in tissue
tion and saturation, suggesting that hypoxia derives from capillaries, what may inhibit the mechanical stimulus to
impaired adipose tissue irrigation (Goossens, 2008; Spencer angiogenesis.
et al., 2011; Trayhurn et al., 2008a). Previous studies AT1 and AT2 receptors, ACE and renin receptor were all
demonstrated a 30–40% reduction of adipose tissue irrigation shown to be present in the adipose tissue and AngII is known
in obese patients and animal models, in part due to the to have direct effects on adipocytes regulating adipogenesis
regression of the vascular network (Goossens, 2008; Ye, and lipolysis/lipogenesis (Kalupahana & Moustaid-Moussa,
2009). One of the factors probably involved in decreased 2012b; Steckelings et al., 2009; Weir, 2007). Activation
tissue perfusion is the impairment of endothelium-dependent of adipose RAS system leads to increased adiposity.
relaxation in adipose tissue arterioles (Farb et al., 2012; AngII was shown to increase lipogenesis but to decrease
Georgescu et al., 2011). We recently showed that decreased adipogenesis through the down-regulation of PPARg, what
irrigation in adipose tissue may trigger important systemic may cause adipocyte hypertrophy. RAS blockade with ACE
metabolic alterations. Using normal rats, we observed that a inhibitors leads to decreased adipocyte size and increased
partial reduction of blood supply in one single fat depot adipocyte number in models of obesity (Kalupahana &
causes hyperinsulinemia 48 hours after the surgery Moustaid-Moussa, 2012a). The involvement of each of the
(Rodrigues et al., in press). AngII receptors in these actions is still not completely
understood. Apparently, both AngII receptors are involved.
AT1 was shown to inhibit lipolysis and its specific inhibition
Vasoactive factors – renin-angiotensin system
increased adipocyte differentiation and decreased cell size
Despite the vascular system of the adipose tissue being (Iwai et al., 2007; Tomono et al., 2008). As well, AT2
highly regulated by several angiogenic factors and extracel- receptors increase lipogenesis and inhibit adipogenesis and
lular matrix regulators, blood pressure inside the tissue is thus mediate AngII-induced adipocyte hypertrophy
DOI: 10.3109/13813455.2013.838971 A vascular piece in the puzzle of adipose tissue dysfunction 5

(Iwai et al., 2009; Kalupahana & Moustaid-Moussa, 2012a; (Christiaens & Lijnen, 2010). VEGF is produced mainly by
Tsuchiya et al., 2006). stromal cells and mature adipocytes since the first embryonic
The mechanisms of AngII action in adipose tissue may stages of adipose tissue, acting in the adjacent capillaries
also include the regulation of the inflammatory cascade. (Christiaens & Lijnen, 2010; Neels et al., 2004). During
RAS system is known to strongly influence the inflammatory adipose tissue expansion, VEGF was also shown to be
mechanisms observed in the atherosclerotic plaques. produced by the inflammatory cells recruited to the tissue.
Despite that its influence in adipose tissue vasculature is The blockade of its receptor (VEGFR2) was shown to inhibit
unknown, AngII was shown to increase monocyte chemo- angiogenesis and pre-adipocyte differentiation, showing that
attractant protein 1 (MCP-1) expression in adipocytes and angiogenesis and adipogenesis are mutually regulated (Cao,
pre-adipocytes a NF-B- dependent manner (Kalupahana & 2007; Hausman & Richardson, 2004; Tam et al., 2009).
Moustaid-Moussa, 2012a; Tsuchiya et al., 2006). MCP-1 However, VEGF involvement in adipose tissue dysfunction is
stimulates angiogenesis through direct induction of endothe- still controversial. Transgenic mice with VEGF over-
lial cell proliferation and increased macrophage recruitment expression brown and white adipose tissue were recently
(Niu et al., 2008; Suganami & Ogawa, 2010). Together shown to be protected from high-fat diet-induced hypoxia and
Archives of Physiology and Biochemistry Downloaded from informahealthcare.com by Cornell University on 12/28/14

with the fact that AngII is known to stimulate matrix inflammation in adipose tissue (Elias et al., 2012). However,
metalloproteinases (MMPs) in other tissues, these mechan- animal models of obesity and obese patients were shown to
isms may contribute to the regulation of adipose tissue have higher VEGF circulating levels, which are decreased
microvasculature. after weight loss (Gómez-Ambrosi et al., 2010).
Contradictory reports may also be influenced by the different
characteristics of genetic models compared with diet-induced
Angiogenesis
and the stage of adipose tissue dysfunction. According to
Although angiogenesis was initially faced as a mechanism recent data, VEGF over-expression may be useful during
that should be inhibited in order to achieve decreased adipose adipose tissue expansion, allowing sustained angiogenesis. On
tissue expansion, recent studies suggested that it is essential the other hand, VEGF inhibition may be useful in the context
for tissue homeostasis and to prevent insulin resistance in of pre-existing adipose tissue dysfunction, inhibiting inflam-
already expanded adipose tissue (Gealekman et al., 2012; mation and improving metabolism and insulin signalling (Sun
Lemoine et al., 2012; Lijnen, 2008; Mannerås-Holm & et al., 2012). Although insulin and AMP-activated protein
For personal use only.

Krook, 2012). In fact, most of the known pro- and anti- kinase (AMPK) may induce VEGF expression, its main
angiogenic factors are produced by many of the cells present regulator is HIF-1a (Treins et al., 2002; Vona-Davis & Rose,
in adipose tissue. Inflammation is highly responsible for 2009; Ye, 2009). Recent reports from our colleagues show
angiogenesis as well, leading to remodelling of the extracel- that HIF-1a stabilization in hypoxic conditions can be
lular matrix and endothelial cell proliferation. The study of compromised by hyperglycaemia, which may contribute to
adipose tissue’s leukocytes demonstrated their pro-angiogenic different observations regarding VEGF expression in models
actions due to the broad range of cytokines and growth factors of obesity and type 2 diabetes (Bento et al., 2010a, 2010b).
produced by these cells. Even in lean models, vascularization Ang-1 and Ang-2 have opposite effects on vessel stabil-
of adipose tissue was reduced after macrophage removal ization. Ang-1 increases vessel stability and interaction
(Ye, 2009). Angiogenic factors may increase vascular stability between endothelial cells and pericytes and extracellular
and integrity or induce loss of stability and increase matrix. Ang-1 also leads to the expression of transforming
cellular migration and proliferation when vascular growth is growth factor b (TGFb) and structural proteins, such as
required (Christiaens & Lijnen, 2010; Lijnen, 2008; Zhang & integrins, fibrin and vitronectin (Hausman & Richardson,
Zhang, 2009). 2004; Hemmeryckx et al., 2010; Lijnen, 2008). On the other
The main stimulus to vascular growth is hypoxia, mainly due hand, Ang-2 expression is stimulated in hypoxic conditions
to the stabilization of the hypoxia inducible factor-1 (HIF-1) and its receptors commonly co-localize with VEGF receptors
(Hosogai et al., 2007; Regazzetti et al., 2009; Rutkowski et al., (Figure 1) (Bento et al., 2010a; Hausman & Richardson,
2009). In the presence of oxygen, HIF-1a factor is hydroxylated 2004). Ang-2 is essential for vessel growth as it turns cell
in proline residues and consequently ubiquitinated and interactions weaker, allowing VEGF-induced proliferation
degraded in the proteasome. On the other hand, during hypoxia and migration. However, VEGF inhibition after Ang-2-
proline-hydroxylases are inhibited, leading to stabilization of induced cell destabilization leads to vessel dysfunction
HIF-1a and activation of gene expression (Semenza, 2004; (Bento et al., 2010a; Hausman & Richardson, 2004). Our
Wood et al., 2009). Among these genes are the vascular group showed that VEGF/Ang-2 ratio is highly regulated.
endothelial growth factor (VEGF), angiopoietins (Ang-1 and Decreased hypoxia-induced HIF-1a stabilization in hypergly-
Ang-2), MMPs, leptin and plasminogen activators (Cao, 2007; caemia may imbalance this ratio, leading to the formation of
Christiaens & Lijnen, 2010; Goossens et al., 2011; Hosogai aberrant capillaries (Figure 1). More, these effects are thought
et al., 2007; Yamakawa et al., 2003; Ye, 2009). to be mediated by glucose-derived methylglyoxal (MG)
VEGF is crucial for endothelial cell proliferation, migra- (Bento et al., 2010a).
tion and survival, ultimately leading to angiogenesis Transforming growth factor (TGF) b was shown to be
(Christiaens & Lijnen, 2010; Hausman & Richardson, 2004; essential for adipose tissue development, as its inhibition
Lijnen, 2008; Tam et al., 2009). Most of VEGF actions in results in uncompleted vessel formation. TGFb is produced by
adipose tissue are mediated by VEGF-A, although other adipocytes and stromal cells in hypoxic conditions, regulating
VEGF forms are also involved in the angiogenic process the deposition of extracellular matrix and leading to vessel
6 T. Rodrigues et al. Arch Physiol Biochem, 2014; 120(1): 1–11

stabilization. However, in conditions of prolonged hypoxia, concentrations above the physiological range. Regarding this
TGFb is conducive to fibrosis, as observed in the adipose matter, we developed a rat model with oral MG administra-
tissue of obese individuals (Figure 1) (Christiaens & Lijnen, tion that had tissue and plasma MG concentrations similar to
2010; Hausman & Richardson, 2004; Ye, 2009). type 2 diabetic Goto-Kakizaki rats. This model showed
Angiogenesis may also be regulated by adipocytokines. several structural and functional alterations in adipose tissue,
Leptin was long shown to have receptors in endothelial cells but not insulin resistance (Matafome et al., 2012).
and a strong angiogenic role (Cao, 2007; Christiaens & MG-induced AGE accumulation is long known to cause
Lijnen, 2010; Hausman & Richardson, 2004; Lijnen, 2008). vascular dysfunction in several models (Berner et al., 2012;
Leptin was shown to promote in vitro proliferation, migration Cantero et al., 2007; Kim et al., 2012; Sena et al., 2012).
and survival and to inhibit the apoptosis of endothelial cells. However, until the last years, adipose tissue microvascular
In addition, leptin was shown to increase VEGF and MMP dysfunction was seen as a consequence of hyperglycaemia, as
production, further increasing vessel growth (Cao, 2007; observed for common diabetic complications, such as retin-
Christiaens & Lijnen, 2010; Hausman & Richardson, 2004; opathy or nephropathy. However, two major ideas should be
Lijnen, 2008; Ye, 2009). On the other hand, adiponectin was retained. First, adipose tissue dysfunction is not a simple
Archives of Physiology and Biochemistry Downloaded from informahealthcare.com by Cornell University on 12/28/14

suggested to balance the pro-angiogenic effects of leptin. consequence of hyperglycaemia. Given its important meta-
Adiponectin was shown to have anti-angiogenic and pro- bolic and endocrine actions, adipose tissue dysfunction due to
apoptotic actions on endothelial cells (Vona-Davis & Rose, microvascular lesions may decisively influence metabolic
2009). Unlike adiponectin, leptin is produced proportionally control and thus the progression of type 2 diabetes. Second,
to fat mass and its angiogenic effects may be a physiological glycation may occur from the early stages of type 2 diabetes
mechanism to compensate for adipose tissue expansion. development, even before open hyperglycaemia is observed.
This suggests that AGE accumulation may lead to a time-
dependent dysfunction of adipose tissue microvasculature,
Glycation
leading to an accelerated adipose tissue dysfunction and
Non-enzymatic glycosylation, also named glycation, was systemic dysmetabolism. In our model of MG-induced AGE
described for the first time by Louis-Camille Maillard. This accumulation, we observed increased fibrosis and impaired
reaction occurs in the presence of reducing sugars (glucose, apoptotic markers and VEGF/Ang2 ratio after a chronic
fructose and others) which are able to react with amine groups administration. This was associated with decreased irrigation
For personal use only.

of several biologic molecules, such as proteins, lipids and and more hypoxic regions. In addition, macrophage recruit-
nucleic acids (Brownlee, 2005; Goldin et al., 2006; Negre- ment to the tissue was increased (Matafome et al., 2012).
Salvayre et al., 2009). Glycation changes physical and More recently, we showed that these effects were time-
chemical properties of the molecules, as well as their dependent and were reverted by the dicarbonyl scavenger
biological functions, in a process which occurs through drug pyridoxamine (Rodrigues et al., 2013). Altogether,
several complex reactions and molecular rearrangements that these results suggest that progressive AGE accumulation in
originate Schiff bases, Amadori products and finally adipose tissue leads to microvascular dysfunction, what may
advanced glycation end-products (AGE) (Goldin et al., hamper a correct expansion of the tissue (Rutkowski et al.,
2006; Negre-Salvayre et al., 2009). Alternatively, AGE may 2009). During the process of expansion, the formation of
form from intermediary products of glucose metabolism such hypoxic regions is the trigger to induce angiogenesis. If
as gliceraldehyde-3-phosphate, dihydroxyacetone-phosphate angiogenesis does not occur in order to compensate for
and MG (see reviews at Negre-Salvayre et al., 2009; Desai hypoxia, the tissue may become dysfunctional. Using a short
et al., 2010; Matafome et al., 2013). period of MG administration in order to avoid its long-
AGE contribute to macro and microvascular diabetic term effects, we recently showed that MG decreases the
complications, mainly by two pathways. First, by formation ability of adipose tissue to adapt to a surgery-induced
of permanent cross-links with proteins of the extracellular reduction of blood supply (Rodrigues et al., in press). In
matrix and plasma, which changes their structure and function rats with MG-induced glycation, a partial decrease of blood
and, second, by interaction with AGE receptor (RAGE) in the supply caused a higher activation of inflammatory pathways
cell surface, leading to intracellular propagation of inflam- and Perilipin A degradation, as well as decreased PPARg
matory signals. Such signals include the activation of tissue levels and adiponectinemia, suggesting adipocyte
the TGF-b, leading to the synthesis of structural proteins dysfunction (Rodrigues et al., in press). Our results suggest
and fibrosis (Brownlee, 2005; Goldin et al., 2006; Matafome that glycation not only impairs adipose tissue microvascu-
et al., 2012; Matafome et al., 2013; Negre-Salvayre et al., lature, but also compromises its ability to adapt to such
2009). conditions, leading to metabolic alterations commonly
The involvement of AGE in adipose tissue dysfunction is observed in type 2 diabetes. However, the physiological
an almost unstudied subject. Until now, most of the studies relevance of such mechanisms in the context of type 2
focused on insulin resistance, showing that fructose supple- diabetes development has still to be proved.
mentation or direct MG administration lead to insulin
resistance in adipose tissue. Additionally, MG administration
Consequences of hypoxia
to 3T3-L1 adipocytes resulted in decreased activation of the
insulin pathway (Dhar et al., 2011; Jia and Wu, 2007; Genetic and diet-induced obese animal models commonly
Riboulet-Chavey, 2006). However, the physiological signifi- have decreased endothelial cell markers, lower partial oxygen
cance of these studies is questionable, as authors used MG pressure, higher accumulation of a hypoxia probe
DOI: 10.3109/13813455.2013.838971 A vascular piece in the puzzle of adipose tissue dysfunction 7

(pimonidazole) and lactate production (endogenous marker of Krishnan et al., 2012; Rutkowski et al., 2009; Tilg &
hypoxia). When hypoxia in adipose tissue becomes perman- Moschen, 2006; Trayhurn et al., 2008b).
ent, stress-response pathways are activated, leading to There is still no consensus about the role of adipose cell
inflammation, decreased glucose and lipid uptake and types in tissue’s response to hypoxia, as it is not possible yet
changes of the secretome (Hosogai et al., 2007; Pang et al., to completely isolate the different cell fractions in vivo and
2008; Spencer et al., 2011; Trayhurn et al., 2008a; Wood the use of cell lines does not entirely mimic in vivo
et al., 2009; Ye et al., 2007). conditions. However, higher NF-B and HIF-1 activity in
hypoxic 3T3 adipocytes was observed to lead to increased
expression of interleukine-6 (IL-6), MCP-1, MMPs, TGFb
Activation of cellular inflammation and stress
and leptin (Halberg et al., 2009; Trayhurn et al., 2008b; Wood
pathways
et al., 2009; Ye, 2009). As well, pre-adipocytes cultured in
Following hypertrophy, excessive intracellular accumulation hypoxia show increased MCP-1, TNFa, GLUT1 and VEGF
of free fatty acids and their metabolites such as diacylclycerol expression and acquire the ability to secrete leptin.
and ceramides, as well as the activation of toll-like receptors Interestingly, the differentiation of hypoxic pre-adipocytes
Archives of Physiology and Biochemistry Downloaded from informahealthcare.com by Cornell University on 12/28/14

(TLR) by circulating free fatty acids, are known to activate to adipocytes was observed to decrease, favouring their
stress and inflammatory pathways. Such pathways include transformation to macrophages (Goossens, 2008; Goossens
several protein kinase C (PKC) isoforms, c-jun N-terminal et al., 2011; Olefsky & Glass, 2010; Tilg & Moschen, 2006).
kinase/stress-activated protein kinase (JNK/SAPK) and kinase Using conditioned medium from hypoxic adipocytes and pre-
of the inhibitor of NF-B (IKK)b, all ending in the adipocytes, Mack and colleagues showed that pre-adipocytes
degradation of the NF-B inhibitor (IBa) in the proteasome exert a stronger influence on the expression of adhesion
and consequent NF-B activation (Figure 2) (Boden, 2011; molecules and activation of proliferation pathways in endo-
Brownlee, 2005; Guilherme et al., 2008; Moeschel et al., thelial cells (Mack et al., 2009).
2004; Qatanani & Lazar, 2007; Ueki & Kondo, 2004; Wellen Regarding the molecular mechanisms activated in hypoxia,
& Hotamisligil, 2005). Hypoxia was also shown to directly it is important to mention that many of the previous studies
activate similar mechanisms (Figure 2). NF-B activation in exploring inflammatory mechanisms used cultured hypoxic
response to hypoxia is well documented, namely in tumors. adipocytes (1% oxygen) and control adipocytes under ambient
HIF-1a was suggested to mediate the inflammatory activity, air (21% oxygen), which is not observed in vivo. Recently,
For personal use only.

as nitric oxide sintetase (iNOS)-mediated nitric oxide (NO) Famulla and colleagues suggested that adipocytes cultured
production was observed to be decreased in cells lacking under 21% oxygen may become hyperoxic, as the physio-
HIF-1a (Zarember & Malech, 2005). In addition, NF-kB was logical oxygen concentration was shown to be 3–11%
also shown to regulate HIF-1 activity. IKKa and IKKb (Famulla et al., 2012; Goossens et al., 2011). Authors
presence was shown to be necessary for HIF-1a stabilization reported that 21% oxygen may impair adipocyte metabolism,
even in hypoxia. Interestingly, NF-B activation in response differentiation and secretory function, in relation to adipo-
to cytokines also increases HIF-1a expression (Uden et al., cytes cultured below 10% oxygen (Famulla et al., 2012). This
2008; Ye, 2009). These data show that HIF-1 and NF-B raises new important questions for future studies.
strongly co-operate to activate cellular mechanisms of
response to hypoxia.
Inhibition of substrate uptake
The formation of hypoxic regions in adipose tissue causes
HIF-1-mediated expression of angiogenic factors and the The impact of the vascular dysfunction in adipocyte glucose
glucose transporter-1 (GLUT-1) (He et al., 2011; Rausch uptake is observable from the fact that the ability of insulin to
et al., 2008; Wang et al., 2007; Ye et al., 2007). The suppress lipolysis is correlated with the vessel density in the
expression of such genes in hypoxic adipocytes was shown to adipose tissue of obese patients (Pasarica et al., 2010). More,
depend on decreased PPARg activity. PPARg inhibition Jun and colleagues recently showed that mice submitted to
resulted in increased HIF-1-dependent expression of inflam- acute hypoxia showed decreased adipocyte ability to uptake
matory genes (Pino et al., 2012). Such genes include pro- triglycerides, in part due to decreased lipoprotein lipase
inflammatory cytokines and chemokines such as MCP-1, activity, causing hypertriglyceridemia (Jun & Shin, 2012).
interleukins and tumour necrosis factor-a (TNF-a), as well as In cultured adipocytes, hypoxia was shown to directly inhibit
adhesion molecules as intercellular adhesion molecule-1 insulin signalling, decreasing glucose uptake. More, this was
(ICAM-1), possibly resulting from higher HIF-1a interaction dependent on HIF-1 activation, despite the precise mechan-
with NF-B (Figure 1) (Donath & Shoelson, 2011; Pino et al., isms not being described (Bugianesi et al., 2005; Chen et al.,
2012; Uden et al., 2008). HIF-1 inhibition in vivo results in 2006; Gentil et al., 2006; Regazzetti et al., 2009; Wood et al.,
decreased blood vessel density, insulin resistance and adipos- 2009; Ye, 2009). However, HIF-1 over-expression in the
ity in high-fat fed mice (Jiang et al., 2011; Park et al., 2012). adipose tissue of mice fed a high-fat diet prevented the
Apparently, the regulation of HIF-1 interaction with NF-B development of insulin resistance (Zhang et al., 2011). These
alternatively to PPARg influences the genes that are results suggest that acute HIF-1 activation inhibits insulin
expressed in hypoxia. This may be of particular relevance signalling but it is crucial for angiogenesis during adipose
in chronic hypoxia where the permanent activation of such tissue expansion, allowing the correct development of the
mechanisms may lead to severe implications on adipose microvascular network.
tissue metabolic and endocrine function, which may The down-regulation of insulin signalling is a direct
have systemic repercussions (Donath & Shoelson, 2011; consequence of the activation of PKC, JNK and IKKb, as
8 T. Rodrigues et al. Arch Physiol Biochem, 2014; 120(1): 1–11

these proteins are serine kinases that phosphorylate insulin Most of the products secreted by the adipose tissue
receptor and its substrate (IRS-1), inhibiting downstream regulate simultaneously metabolic and inflammatory adapta-
signalling (Figure 2) (reviewed by Wellen & Hotamisligil, tions of adipose tissue to hypoxia. Adiponectin has anti-
2005; Guilherme et al., 2008). In particular, JNK is activated inflammatory insulin sensitizer effects and its expression was
by lipid-induced PKC activation, free fatty acids-induced observed to be decreased in the hypoxic adipose tissue of
TLR activation and inflammatory cytokines such as TNF-a, obese mice and in hypoxic cultured human adipocytes, in part
leading to IKKb and suppressor of cytokine signalling through PPARg down-regulation (Hosogai et al., 2007; Wang
(SOCS) activation (Figure 2). SOCS was shown to signalize et al., 2007). On the other hand, the expression of other
IRS-1 to proteasomal degradation, reinforcing the inhibition factors like leptin, resistin, visfatin, MCP-1, migration
of insulin signalling (Ueki and Kondo, 2004). However, inhibitory factor (MIF), TNF-a and VEGF is increased,
recent data is insufficient to understand if these pathways are contributing to inflammation, angiogenesis and insulin
directly activated in hypoxia or if they are secondarily resistance (Figure 2) (Golay & Ybarra, 2005; Gustafson,
activated by cytokines and free fatty acids released in 2010; Hosogai et al., 2007; Juge-Aubry et al., 2005; Maury &
consequence of hypoxia. The mechanisms of HIF-1-mediated Brichard, 2010; Rajala & Scherer, 2003; Segawa et al., 2006;
Archives of Physiology and Biochemistry Downloaded from informahealthcare.com by Cornell University on 12/28/14

down-regulation of insulin signalling in adipocytes should Tilg & Moschen, 2006; Wang et al., 2007). As well, some
retain future attention. adipocytokines have the ability to influence blood flow.
A major function of insulin in adipocytes is to inhibit While adiponectin is known to stimulate vasodilation through
lipolysis and consequent fatty acid release to the circulation. the activation of AMP-activated protein kinase (AMPK) and
Lipolysis is highly activated by the increase of intracellular the opening of voltage-gated potassium channels, leptin and
levels of cyclic adenosine monophosphate (cAMP), as in resistin lead to vasoconstriction (Zhang & Zhang, 2009).
fasting, leading to the activation of hormone-sensitive lipase However, these results were obtained in non-adipose arteri-
(HSL). On the other hand, insulin is a direct repressor of oles and may not be directly transposed to adipose tissue
AMPc production by adenylate cyclase (Guilherme et al., perfusion.
2008; Juge-Aubry et al., 2005; Ye, 2008). Cytokines like MCP-1 is a potent chemo-attractant factor, which induces
TNF-a activate adenylate cyclase and thus increase lipolysis. monocyte recruitment to adipose tissue, as observed in several
More, these mechanisms include Perilipin A degradation, animal models of obesity and vascular dysfunction in adipose
allowing the binding of HSL to triglycerides stored in lipid tissue (Maury & Brichard, 2010; Matafome et al., 2012;
For personal use only.

droplets (Figure 2) (Guilherme et al., 2008; Juge-Aubry et al., Olefsky & Glass, 2010; Rausch et al., 2008; Wang et al.,
2005). Thus, hypoxia-induced insulin resistance and TNF-a 2007; Ye et al., 2007). The MCP-1 secreted by adipocytes and
production lead to increased lipolysis and increased circulat- infiltrating macrophages leads to a progressive monocyte
ing free fatty acids (FFA). chemo-attraction, perpetuating the inflammatory loop
Along with lipolysis, decreased fatty acid storage is also (Guilherme et al., 2008; Olefsky & Glass, 2010; Ye, 2009).
caused by decreased PPARg activity. In cultured adipocytes, Furthermore, these signals also promote pre-adipocyte differ-
hypoxia was observed to reduce PPARg activity and entiation into macrophages (Goossens, 2008; Olefsky &
adiponectin secretion (Regazzetti et al., 2009; Wood et al., Glass, 2010; Tilg & Moschen, 2006). In addition, macrophage
2009; Ye, 2009). Decreased PPARg activity in hypoxic exit from adipose tissue is strongly inhibited by MIF
adipocytes leads to decreased expression of enzymes involved (Ye, 2009). Macrophage accumulation in adipose tissue and
in fatty acids uptake and esterification, decreasing adipocyte alteration of their sub-populations (M1 and M2) completely
ability to store lipids (Figure 2) (Guilherme et al., 2008; Juge- change tissue’s secretome. Interestingly, M1 polarity was
Aubry et al., 2005; Wellen & Hotamisligil, 2003). Besides its shown to be increased in hypoxia, at least in part through HIF-
relevant metabolic activities, PPARg apparently has pro- 1-dependent mechanisms (Fujisaka et al., 2013). Besides
angiogenic properties when activated in endothelial cells. having the ability to remove death adipocytes and to secrete
PPARg agonists troglitazone and rosiglitazone were shown to inflammatory factors (also mediate the angiogenic stimulus in
induce in vitro endothelial cell proliferation (Kakiuchi-Kiyota endothelial cells), macrophages are usually located in hypoxic
et al., 2009; Kakiuchi-Kiyota & Arnold, 2011). Similar regions and secrete a broad range of angiogenic and matrix
results were observed with rosiglitazone in cultured adipose remodelling factors (VEGF, MMPs, resistin, among others),
tissue explants from obese patients (Gealekman et al., 2012). favouring angiogenesis (Olefsky & Glass, 2010; Ye, 2009).

Alterations of the secretome Conclusions


Hypoxia-induced inflammation causes important alterations of Despite the exact mechanisms being partially unknown,
the secretory profile of adipose tissue. Besides stimulating hypoxia has been shown to activate inflammatory pathways
angiogenesis, these changes strongly contribute to the propa- in adipose tissue, leading to insulin resistance, lipolysis,
gation of the inflammatory feedback and to continually inhibit PPARg inhibition and macrophage recruitment. This com-
substrate uptake. Importantly, these mechanisms may be pletely changes adipose tissue secretome, which may con-
essential for tissue homeostasis during its expansion. tribute to accelerated systemic dysmetabolism and thus to
However, their chronic activation in obesity causes permanent type 2 diabetes.
low-grade inflammation, insulin resistance, lipolysis and In the recent years, many studies have been conducted in
macrophage recruitment (Kimura et al., 2011; Olefsky & order to determine the causes of adipose tissue hypoxia. From
Glass, 2010; Qatanani & Lazar, 2007; Tilg & Moschen, 2006). excessive adipocyte volume to impaired angiogenesis and
DOI: 10.3109/13813455.2013.838971 A vascular piece in the puzzle of adipose tissue dysfunction 9

blood flow, many hypotheses have been proposed. Even if the Farb M, Ganley-Leal L, Mott M, et al. (2012). Arteriolar function in
visceral adipose tissue is impaired in human obesity. Arteriosclerosis,
involvement of such mechanisms in adipose tissue vascular Thromb, Vasc Biol, 32:467–73.
dysfunction has been observed in several animal and human Fujisaka S, Usui I, Ikutani M, et al. (2013). Adipose tissue hypoxia
models, the physiological relevance of each one of them is induces inflammatory M1 polarity of macrophages in an HIF-1a-
still poorly understood. The real importance of the RAS dependent and HIF-1a-independent manner in obese mice.
Diabetologia, 56:1403–12.
system in the dysfunction of adipose tissue microvasculature Galic S, Oakhill S, Steinberg R. (2010). Adipose tissue as an endocrine
is still unknown. The role of glycation in angiogenesis organ. Molec Cell Endocrin, 316:129–39.
disturbance and its pathophysiological significance should Gealekman O, Guseva N, Gurav K, et al. (2012). Effect of rosiglitazone
continue to be focused on in the future. Furthermore, other on capillary density and angiogenesis in adipose tissue of normogly-
caemic humans in a randomised controlled trial. Diabetologia, 55:
unstudied mechanisms may interplay in the regulation of 2794–9.
adipose tissue irrigation. Such mechanisms may include, for Gentil C, Le Jan S, Philippe J, et al. (2006). Is oxygen a key factor in the
example, gastrointestinal hormones, catecholamines, gluco- lipodystrophy phenotype? Lipids Health Disease, 5:1–11.
corticoids or growth hormone, among others. Georgescu A, Popov D, Constantin A, et al. (2011). Dysfunction of
human subcutaneous fat arterioles in obesity alone or obesity
associated with Type 2 diabetes. Clin Sci, 120:463–72.
Archives of Physiology and Biochemistry Downloaded from informahealthcare.com by Cornell University on 12/28/14

Golay A, Ybarra J. (2005). Link between obesity and type 2 diabetes.


Declaration of interest Best practice and research. Clin Endocrin Metab, 19:649–63.
Goldin A, Beckman J, Schmidt M, Creager M. (2006). Advanced
The authors report no conflicts of interest. The authors alone glycation end products: Sparking the development of diabetic vascular
are responsible for the content and writing of this article. injury. Circulation, 114:597–605.
Gómez-Ambrosi J, Catalán V, Rodrı́guez A, et al. (2010). Involvement of
serum vascular endothelial growth factor family members in the
References development of obesity in mice and humans. J Nutri Biochem, 21:
774–80.
Bagi Z, Feher A, Cassuto J. (2012). Microvascular responsiveness in Goossens H. (2008). The role of adipose tissue dysfunction in the
obesity: Implications for therapeutic intervention. Brit J Pharm, 165: pathogenesis of obesity-related insulin resistance. Physiol Behav, 94:
544–60. 206–18.
Bento C, Fernandes R, Matafome P, et al. (2010a). Methylglyoxal- Goossens H, Bizzarri A, Venteclef N, et al. (2011). Increased adipose
induced imbalance in the ratio of vascular endothelial growth factor to tissue oxygen tension in obese compared with lean men is
angiopoietin 2 secreted by retinal pigment epithelial cells leads to accompanied by insulin resistance, impaired adipose tissue capillar-
endothelial dysfunction. Exp Phys, 95:955–70. ization, and inflammation. Circulation, 124:67–76.
For personal use only.

Bento C, Fernandes R, Ramalho J, et al. (2010b). The chaperone- Guilherme A, Virbasius V, Puri V, Czech P. (2008). Adipocyte
dependent ubiquitin ligase CHIP targets HIF-1a for degradation in the dysfunctions linking obesity to insulin resistance and type 2 diabetes.
presence of methylglyoxal. PloS One, 5:e15062. Nature reviews. Molec Cell Biol, 9:367–77.
Berner A, Brouwers O, Pringle R, et al. (2012). Protection against Gustafson B. (2010). Adipose tissue, inflammation and atherosclerosis.
methylglyoxal-derived AGEs by regulation of glyoxalase 1 prevents J Athero Thromb, 17:332–41.
retinal neuroglial and vasodegenerative pathology. Diabetologia, 55: Halberg N, Khan T, Trujillo E, et al. (2009). Hypoxia-inducible factor
845–54. 1alpha induces fibrosis and insulin resistance in white adipose tissue.
Boden G. (2011). Obesity, insulin resistance and free fatty acids. Curr Molec Cell Biol, 29:4467–83.
Opin Endocrinol Diabetes Obes, 18:1–11. Hausman J, Richardson L. (2004). Adipose tissue angiogenesis. J Animal
Brownlee, M. (2005). The pathobiology of diabetic complications: Sci, 82:925–34.
A unifying mechanism. Diabetes, 54:1615–25. He Q, Gao Z, Yin J, et al. (2011). Regulation of HIF-1{alpha} activity in
Bugianesi E, McCullough A, Marchesini G. (2005). Insulin resistance: A adipose tissue by obesity-associated factors: adipogenesis, insulin, and
metabolic pathway to chronic liver disease. Hepatology, 42:987–1000. hypoxia. Am J Physiol Endocrin Metab, 300:877–85.
Cantero A, Portero-Otı́n M, Ayala V, et al. (2007). Methylglyoxal Hemmeryckx B, Loeckx D, Dresselaers T, et al. (2010). Age-associated
induces advanced glycation end product (AGEs) formation and adaptations in murine adipose tissues. Endocrine J, 57:925–30.
dysfunction of PDGF receptor-beta: Implications for diabetic athero- Hosogai N, Fukuhara A, Oshima K, et al. (2007). Adipose tissue hypoxia
sclerosis. FASEB J (Official publication of the Federation of American in obesity and its impact on adipocytokine dysregulation. Diabetes,
Societies for Experimental Biology), 21:3096–106. 56:901–11.
Cao Y. (2007). Angiogenesis modulates adipogenesis and obesity. J Clin Iwai M, Chen R, Imura Y, Horiuchi M. (2007). Tak-536, a new AT1
Invest, 117:2362–8. receptor blocker, improves glucose intolerance and adipocyte differ-
Chen B, Lam L, Wang Y, et al. (2006). Hypoxia dysregulates the entiation. Am J Hyper, 20:579–86.
production of adiponectin and plasminogen activator inhibitor-1 Iwai M, Tomono Y, Inaba S, et al. (2009). AT2 receptor deficiency
independent of reactive oxygen species in adipocytes. Biochem attenuates adipocyte differentiation and decreases adipocyte number
Biophys Research Comm, 341:549–56. in atherosclerotic mice. Am J Hyper, 22:784–91.
Christiaens V, Lijnen R. (2010). Angiogenesis and development of Jia X, Wu L. (2007). Accumulation of endogenous methylglyoxal
adipose tissue. Molec Cell Endocrin, 318:2–9. impaired insulin signaling in adipose tissue of fructose-fed rats. Molec
Desai M, Chang T, Wang H, et al. (2010). Oxidative stress and aging: Is Cell Biochem, 306:133–9.
methylglyoxal the hidden enemy? Canadian J Phys Pharm, 88: Jiang C, Qu A, Matsubara T, et al. (2011). Disruption of hypoxia-
273–84. inducible factor 1 in adipocytes improves insulin sensitivity
Dhar A, Dhar I, Jiang B, et al. (2011). Chronic methylglyoxal infusion by and decreases adiposity in high-fat diet-fed mice. Diabetes, 60:
minipump causes pancreatic beta-cell dysfunction and induces type 2 2484–95.
diabetes in Sprague-Dawley rats. Diabetes, 60:899–908. Juge-Aubry E, Henrichot E, Meier C. (2005). Adipose tissue:
Donath Y, Shoelson E. (2011). Type 2 diabetes as an inflammatory A regulator of inflammation. Best Practice Res Clin Endocrin
disease. Nature Rev Immunol, 11:98–107. Metab, 19:547–66.
Elias I, Franckhauser S, Ferré T, et al. (2012). Adipose tissue Jun J, Shin M. (2012). Acute hypoxia induces hypertriglyceridemia by
overexpression of vascular endothelial growth factor protects against decreasing plasma triglyceride clearance in mice. Am J Physiol
diet-induced obesity and insulin resistance. Diabetes, 61:1801–13. Endocrin Metab, 303:377–88.
Famulla S, Schlich R, Sell H, Eckel J. (2012). Differentiation of human Kakiuchi-Kiyota S, Vetro J, Suzuki S, et al. (2009). Effects of the
adipocytes at physiological oxygen levels results in increased PPARgamma agonist troglitazone on endothelial cells in vivo and
adiponectin secretion and isoproterenol-stimulated lipolysis. in vitro: Differences between human and mouse. Toxicol Appl
Adipocyte, 1:132–41. Pharmacol, 237:83–90.
10 T. Rodrigues et al. Arch Physiol Biochem, 2014; 120(1): 1–11

Kakiuchi-Kiyota S, Arnold L. (2011). Evaluation of direct and indirect Qatanani M, Lazar M. (2007). Mechanisms of obesity-associated
effects of the PPARÎ3 agonist troglitazone on mouse endothelial cell insulin resistance: Many choices on the menu. Genes Develop, 21:
proliferation. Toxicol Pathol, 39:1032–45. 1443–55.
Kalupahana S, Moustaid-Moussa N. (2012a). The renin-angiotensin Rajala W, Scherer E. (2003). Minireview: The adipocyte – At the
system: a link between obesity, inflammation and insulin resistance. crossroads of energy homeostasis, inflammation, and atherosclerosis.
Obesity Rev (Official Journal of the International Association for the Endocrinology, 144:3765–73.
Study of Obesity), 13:136–49. Rausch E, Weisberg S, Vardhana P, Tortoriello V. (2008). Obesity in
Kalupahana S, Massiera F, Quignard-Boulange A, et al. (2012b). C57BL/6J mice is characterized by adipose tissue hypoxia and
Overproduction of angiotensinogen from adipose tissue induces cytotoxic T-cell infiltration. Inter J Obesity, 32:451–63.
adipose inflammation, glucose intolerance, and insulin resistance. Regazzetti C, Peraldi P, Grémeaux T, et al. (2009). Hypoxia
Obesity, 20:48–56. decreases insulin signaling pathways in adipocytes. Diabetes, 58:
Kaneto H, Katakami N, Matsuhisa M, Matsuoka T. (2010). Role of 95–103.
reactive oxygen species in the progression of type 2 diabetes and Riboulet-Chavey A, Pierron A, Durand I, et al. (2006). Methylglyoxal
atherosclerosis. Mediators Inflam, 2010:1–11. impairs the insulin signaling pathways independently of the formation
Kim J, Kim S, Kim S, et al. (2012). Accumulation of argpyrimidine, a of intracellular reactive oxygen species. Diabetes, 55:1289–99.
methylglyoxal-derived advanced glycation end product, increases Rodrigues T, Paulo M, Santos-Silva D, et al. (2013). Reduction of
apoptosis of lens epithelial cells both in vitro and in vivo. Exp Molec methylglyoxal-induced glycation by pyridoxamine improves adipose
Med, 44:167–75. tissue microvascular lesions. J Diab Res, 2013:1–9.
Archives of Physiology and Biochemistry Downloaded from informahealthcare.com by Cornell University on 12/28/14

Kimura R, Takahashi N, Murota K, et al. (2011). Activation of Rodrigues T, Matafome P, Seiça R. (in press). Methylglyoxal further
peroxisome proliferator-activated receptor-a (PPARa) suppresses impairs adipose tissue metabolism after partial decrease of blood
postprandial lipidemia through fatty acid oxidation in enterocytes. supply. Arch Physiol Biochem.
Biochem Biophys Res Comm, 410:1–6. Rutkowski M, Davis E, Scherer E. (2009). Mechanisms of obesity and
Krishnan J, Danzer C, Simka T, et al. (2012). Dietary obesity-associated related pathologies: The macro- and microcirculation of adipose
Hif1a activation in adipocytes restricts fatty acid oxidation and energy tissue. FEBS J, 276:5738–46.
expenditure via suppression of the Sirt2-NADþ system. Genes Scalia R. (2013). The microcirculation in adipose tissue inflammation.
Develop, 26:259–70. Rev Endo Metab Disorders, 14:69–76.
Lemoine Y, Ledoux S, Quéguiner I, et al. (2012). Link between adipose Segawa K, Fukuhara A, Hosogai N, et al. (2006). Visfatin in adipocytes
tissue angiogenesis and fat accumulation in severely obese subjects. is upregulated by hypoxia through HIF1alpha-dependent mechanism.
J Clin Endocrin Metab, 97:775–80. Biochem Biophys Research Comm, 349:875–82.
Lijnen R. (2008). Angiogenesis and obesity. Cardio Res, 78:286–93. Semenza L. (2004). Hydroxylation of HIF-1: Oxygen sensing at the
Mack I, Belaiba S, Djordjevic T, et al. (2009). Functional analyses reveal molecular level. Physiology, 19:176–82.
the greater potency of preadipocytes compared with adipocytes as Sena C, Matafome P, Crisóstomo J, et al. (2012). Methylglyoxal
endothelial cell activator under normoxia, hypoxia, and TNFalpha promotes oxidative stress and endothelial dysfunction. Pharm Res
exposure. Am J Physiol Endocrin Metab, 297:735–48. (Official Journal of the Italian Pharmacological Society), 65:
For personal use only.

Mannerås-Holm L, Krook A. (2012). Targeting adipose tissue angio- 497–506.


genesis to enhance insulin sensitivity. Diabetologia, 55:2562–4. Spencer M, Unal R, Zhu B, et al. (2011). Adipose tissue extracellular
Matafome P, Santos-Silva D, Crisóstomo J, et al. (2012). Methylglyoxal matrix and vascular abnormalities in obesity and insulin resistance.
causes structural and functional alterations in adipose tissue inde- J Clin Endocrin Metab, 96:1990–8.
pendently of obesity. Arch Physiol Biochem, 118:58–68. Steckelings M, Rompe F, Kaschina E, et al. (2010). The past, present and
Matafome P, Sena C, Seiça R. (2013). Methylglyoxal, obesity, and future of angiotensin II type 2 receptor stimulation. J Renin-Angio-
diabetes. Endocrine, 43:1–37. Aldo Syst (JRAAS), 11:67–73.
Maury E, Brichard M. (2010). Adipokine dysregulation, adipose tissue Steckelings M, Rompe F, Kaschina E, Unger T. (2009). The evolving
inflammation and metabolic syndrome. Molec Cell Endocrin, 314: story of the RAAS in hypertension, diabetes and CV disease: Moving
1–16. from macrovascular to microvascular targets. Fund Clinical Pharm,
Moeschel K, Beck A, Weigert C, et al. (2004). Protein kinase C-zeta- 23:693–703.
induced phosphorylation of Ser318 in insulin receptor substrate-1 Suganami T, Ogawa Y. (2010). Adipose tissue macrophages: their role in
(IRS-1) attenuates the interaction with the insulin receptor and the adipose tissue remodeling. J Leukocyte Biol, 88:33–9.
tyrosine phosphorylation of IRS-1. J Biol Chem, 279:25157–63. Sun K, Wernstedt I, Kusminski M, et al. (2012). Dichotomous effects of
Neels G, Thinnes T, Loskutoff J. (2004). Angiogenesis in an in vivo VEGF-A on adipose tissue dysfunction. Proc National Acad Sci USA,
model of adipose tissue development. FASEB J, 19:1–19. 109:5874–9.
Negre-Salvayre A, Salvayre R, Augé N, et al. (2009). Hyperglycemia and Tam J, Duda G, Perentes Y, et al. (2009). Blockade of VEGFR2 and not
glycation in diabetic complications. Antiox Redox Sig, 11:3071–109. VEGFR1 can limit diet-induced fat tissue expansion: Role of local
Negro R. (2008). Endothelial effects of antihypertensive treatment: versus bone marrow-derived endothelial cells. PloS One, 4:e4974.
Focus on irbesartan. Vasc Health Risk Manage, 4:89–101. Tilg H, Moschen R. (2006). Adipocytokines: mediators linking adipose
Niu J, Azfer A, Zhelyabovska O, et al. (2008). Monocyte chemotactic tissue, inflammation and immunity. Nature Rev Immunology, 6:
protein (MCP)-1 promotes angiogenesis via a novel transcription 772–83.
factor, MCP-1-induced protein (MCPIP). J Biol Chem, 283:14542–51. Tobin L, Simonsen L, Bülow J. (2010). Real-time contrast-enhanced
Olefsky M, Glass K. (2010). Macrophages, inflammation, and insulin ultrasound determination of microvascular blood volume in abdom-
resistance. Ann Rev Physiol, 72:219–46. inal subcutaneous adipose tissue in man. Evidence for adipose tissue
Ozcan U, Cao Q, Yilmaz E, et al. (2004). Endoplasmic reticulum stress capillary recruitment. Clin Physiol Func Imag, 30:447–52.
links obesity, insulin action, and type 2 diabetes. Science, 306:457–61. Tomono Y, Iwai M, Inaba S, et al. (2008). Blockade of AT1 receptor
Pang C, Gao Z, Yin J, et al. (2008). Macrophage infiltration into adipose improves adipocyte differentiation in atherosclerotic and diabetic
tissue may promote angiogenesis for adipose tissue remodeling in models. Am J Hyper, 21:206–12.
obesity. Am J Physiol Endocrin Metab, 295:313–22. Trayhurn P, Wang B, Wood S. (2008a). Hypoxia in adipose tissue: a
Park S, David E, Huang Park B, et al. (2012). In vivo delivery of cell- basis for the dysregulation of tissue function in obesity? Brit J
permeable antisense hypoxia-inducible factor 1a oligonucleotide to Nutrition, 100:227–35.
adipose tissue reduces adiposity in obese mice. J Cont Release Trayhurn P, Wang B, Wood S. (2008b). Hypoxia and the endocrine and
(Official Journal of the Controlled Release Society), 161:1–9. signalling role of white adipose tissue. Arch Physiol Biochem, 114:
Pasarica M, Rood J, Ravussin E, et al. (2010). Reduced oxygenation in 267–76.
human obese adipose tissue is associated with impaired insulin Treins C, Giorgetti-Peraldi S, Murdaca J, et al. (2002). Insulin stimulates
suppression of lipolysis. J Clin Endocrin Metab, 95:4052–5. hypoxia-inducible factor 1 through a phosphatidylinositol 3-kinase/
Pino E, Wang H, McDonald M. (2012). Roles for peroxisome target of rapamycin-dependent signaling pathway. J Bio Chem, 277:
proliferator-activated receptor g (PPARg) and PPARg coactivators 27975–81.
1a and b in regulating response of white and brown adipocytes to Tsuchiya K, Yoshimoto T, Hirono Y, et al. (2006). Angiotensin II
hypoxia. J Bio Chem, 287:18351–8. induces monocyte chemoattractant protein-1 expression via a nuclear
DOI: 10.3109/13813455.2013.838971 A vascular piece in the puzzle of adipose tissue dysfunction 11
factor-kappaB-dependent pathway in rat preadipocytes. Am J Physiol diabetes independent of insulin resistance. Diabetologia, 43:
Endocrin Metab, 291:E771–8. 1498–506.
Uden V, Kenneth S, Rocha S. (2008). Regulation of hypoxia inducible Wood S, De Heredia F, Wang B, Trayhurn P. (2009). Cellular hypoxia
factor-1a by NF-kB. Biochem J, 412:477–84. and adipose tissue dysfunction in obesity. Proc Nutrition Soc, 68:
Ueki K, Kondo T. (2004). Suppressor of cytokine signaling 1 (SOCS-1) 370–7.
and SOCS-3 cause insulin resistance through inhibition of tyrosine World Health Organization. (2000). Obesity: preventing and managing
phosphorylation of insulin receptor substrate proteins. Mol Cell Biol, the global epidemic. Report of a WHO consultation. WHO Tech Rep
24:5434–46. Series, 894:1–253.
Vázquez-Vela M, Torres N, Tovar R. (2008). White adipose tis- Yamakawa M, Liu X, Date T, et al. (2003). Hypoxia-inducible factor-1
sue as endocrine organ and its role in obesity. Arch Med Res, 39: mediates activation of cultured vascular endothelial cells by inducing
715–28. multiple angiogenic factors. Circ Res, 93:664–73.
Villela N, Kramer-Aguiar L. (2009). Metabolic disturbances linked to Ye J. (2008). Emerging role of adipose tissue hypoxia in obesity and
obesity: The role of impaired tissue perfusion. Arquivos brasileiros de insulin resistance. Int J Obesity, 33:22–25.
endocrinologia e metabolismo, 53:238–45. Ye J. (2009). Emerging role of adipose tissue hypoxia in obesity and
Vona-Davis L, Rose P. (2009). Angiogenesis, adipokines and breast insulin resistance. Int J Obesity, 33:54–66.
cancer. Cyto Growth Factor Rev, 20:193–201. Ye J, Gao Z, Yin J, He Q. (2007). Hypoxia is a potential risk factor for
Wang B, Wood S, Trayhurn P. (2007). Dysregulation of the expres- chronic inflammation and adiponectin reduction in adipose tissue of
sion and secretion of inflammation-related adipokines by hypoxia in ob/ob and dietary obese mice. Am J Physiol Endocrin Metab, 293:
Archives of Physiology and Biochemistry Downloaded from informahealthcare.com by Cornell University on 12/28/14

human adipocytes. Pflügers Archiv: Euro J Physiol, 455:479–92. 1118–28.


Weir R. (2007). Targeting mechanisms of hypertensive vascular disease Zarember K, Malech L. (2005). HIF-1a: A master regulator of innate
with dual calcium channel and renin-angiotensin system blockade. host defenses? J Clin Invest, 115:2003–5.
J Human Hyper, 21:770–79. Zhang H, Zhang C. (2009). Regulation of microvascular function by
Wellen K, Hotamisligil G. (2003). Obesity-induced inflammatory adipose tissue in obesity and type 2 diabetes: Evidence of an adipose-
changes in adipose tissue. J Clin Invest, 112:1785–8. vascular loop. Am J Biomed Sci, 1:1–13.
Wellen K, Hotamisligil G. (2005). Inflammation, stress, and diabetes. Zhang X, Lam K, Ye H, et al. (2011). Adipose tissue-specific inhibition
J Clin Invest, 115:1111–19. of hypoxia-inducible factor 1a induces obesity and glucose intoler-
Weyer C, Foley E, Bogardus C, et al. (2000). Enlarged subcutaneous ance by impeding energy expenditure in mice. Am Soc Biochem Molec
abdominal adipocyte size, but not obesity itself, predicts type II Biol, 285:32869–77.
For personal use only.

Vous aimerez peut-être aussi