Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
1.
Br J Nutr ; 127(2): 161-164, 2022 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-35016740

RESUMO

I had been working on the endocrine and signalling role of white adipose tissue (WAT) since 1994 following the identification of the ob (Lep) gene(1), this after some 15 years investigating the physiological role of brown adipose tissue. The ob gene, a mutation in which it is responsible for the profound obesity of ob/ob (Lepob/Lepob) mice, is expressed primarily in white adipocytes and encodes the pleiotropic hormone leptin. The discovery of this adipocyte hormone had wide-ranging implications, including that white fat has multiple functions that far transcend the traditional picture of a simple lipid storage organ.


Assuntos
Adipocinas , Tecido Adiposo Branco , Adipócitos , Tecido Adiposo Marrom/fisiologia , Tecido Adiposo Branco/fisiologia , Animais , Inflamação , Leptina/genética , Camundongos
2.
Physiol Rev ; 93(1): 1-21, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23303904

RESUMO

The rise in the incidence of obesity has led to a major interest in the biology of white adipose tissue. The tissue is a major endocrine and signaling organ, with adipocytes, the characteristic cell type, secreting a multiplicity of protein factors, the adipokines. Increases in the secretion of a number of adipokines occur in obesity, underpinning inflammation in white adipose tissue and the development of obesity-associated diseases. There is substantial evidence, particularly from animal studies, that hypoxia develops in adipose tissue as the tissue mass expands, and the reduction in Po(2) is considered to underlie the inflammatory response. Exposure of white adipocytes to hypoxic conditions in culture induces changes in the expression of >1,000 genes. The secretion of a number of inflammation-related adipokines is upregulated by hypoxia, and there is a switch from oxidative metabolism to anaerobic glycolysis. Glucose utilization is increased in hypoxic adipocytes with corresponding increases in lactate production. Importantly, hypoxia induces insulin resistance in fat cells and leads to the development of adipose tissue fibrosis. Many of the responses of adipocytes to hypoxia are initiated at Po(2) levels above the normal physiological range for adipose tissue. The other cell types within the tissue also respond to hypoxia, with the differentiation of preadipocytes to adipocytes being inhibited and preadipocytes being transformed into leptin-secreting cells. Overall, hypoxia has pervasive effects on the function of adipocytes and appears to be a key factor in adipose tissue dysfunction in obesity.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo Branco/metabolismo , Hipóxia/metabolismo , Obesidade/metabolismo , Oxigênio/sangue , Adipócitos/patologia , Adipogenia , Adipocinas/genética , Adipocinas/metabolismo , Tecido Adiposo Branco/patologia , Tecido Adiposo Branco/fisiopatologia , Animais , Metabolismo Energético , Regulação da Expressão Gênica , Humanos , Hipóxia/genética , Hipóxia/patologia , Hipóxia/fisiopatologia , Mediadores da Inflamação/metabolismo , Obesidade/genética , Obesidade/patologia , Obesidade/fisiopatologia , Transdução de Sinais
3.
Cytokine ; 110: 189-193, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29775920

RESUMO

Expression of GPCR fatty acid sensor/receptor genes in adipocytes is modulated by inflammatory mediators, particularly IL-1ß. In this study we examined whether the IL-1 gene superfamily member, IL-33, also regulates expression of the fatty acid receptor genes in adipocytes. Human fat cells, differentiated from preadipocytes, were incubated with IL-33 at three different dose levels for 3 or 24 h and mRNA measured by qPCR. Treatment with IL-33 induced a dose-dependent increase in GPR84 mRNA at 3 h, the level with the highest dose being 13.7-fold greater than in controls. Stimulation of GPR84 expression was transitory; the mRNA level was not elevated at 24 h. In contrast to GPR84, IL-33 had no effect on GPR120 expression. IL-33 markedly stimulated expression of the IL1B, CCL2, IL6, CXCL2 and CSF3 genes, but there was no effect on ADIPOQ expression. The largest effect was on CSF3, the mRNA level of which increased 183-fold over controls at 3 h with the highest dose of IL-33; there was a parallel increase in the secretion of G-CSF protein into the medium. It is concluded that in human adipocytes IL-33, which is synthesised in adipose tissue, has a strong stimulatory effect on the expression of cytokine and chemokine genes, particularly CSF3, and on the expression of GPR84, a pro-inflammatory fatty acid receptor.


Assuntos
Adipócitos/metabolismo , Quimiocinas/genética , Citocinas/genética , Interleucina-33/genética , Receptores de Superfície Celular/genética , Adiponectina/genética , Tecido Adiposo/metabolismo , Células Cultivadas , Ácidos Graxos/genética , Fator Estimulador de Colônias de Granulócitos/genética , Humanos , Interleucina-1beta/genética , Macrófagos/metabolismo , RNA Mensageiro/genética , Receptores Acoplados a Proteínas G , Fator de Necrose Tumoral alfa/genética
4.
Annu Rev Nutr ; 34: 207-36, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24819450

RESUMO

Hypoxia develops in white adipose tissue in obese mice, resulting in changes in adipocyte function that may underpin the dysregulation that leads to obesity-associated disorders. Whether hypoxia occurs in adipose tissue in human obesity is unclear, with recent studies contradicting earlier reports that this was the case. Adipocytes, both murine and human, exhibit extensive functional changes in culture in response to hypoxia, which alters the expression of up to 1,300 genes. These include genes encoding key adipokines such as leptin, interleukin (IL)-6, vascular endothelial growth factor (VEGF), and matrix metalloproteinase-2 (MMP-2), which are upregulated, and adiponectin, which is downregulated. Hypoxia also inhibits the expression of genes linked to oxidative metabolism while stimulating the expression of genes associated with glycolysis. Glucose uptake and lactate release by adipocytes are both stimulated by hypoxia, and insulin sensitivity falls. Preadipocytes and macrophages in adipose tissue also respond to hypoxia. The hypoxia-signaling pathway may provide a new target for the treatment of obesity-associated disorders.


Assuntos
Adipócitos Marrons/metabolismo , Adipócitos Brancos/metabolismo , Modelos Biológicos , Obesidade/metabolismo , Adipócitos Marrons/imunologia , Adipócitos Marrons/patologia , Adipócitos Brancos/imunologia , Adipócitos Brancos/patologia , Adipogenia , Animais , Hipóxia Celular , Metabolismo Energético , Humanos , Fator 1 Induzível por Hipóxia/metabolismo , Resistência à Insulina , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Obesidade/imunologia , Obesidade/patologia
5.
Br J Nutr ; 108(11): 1915-23, 2012 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-23046765

RESUMO

Vitamin D deficiency and the rapid increase in the prevalence of obesity are both considered important public health issues. The classical role of vitamin D is in Ca homoeostasis and bone metabolism. Growing evidence suggests that the vitamin D system has a range of physiological functions, with vitamin D deficiency contributing to the pathogenesis of several major diseases, including obesity and the metabolic syndrome. Clinical studies have shown that obese individuals tend to have a low vitamin D status, which may link to the dysregulation of white adipose tissue. Recent studies suggest that adipose tissue may be a direct target of vitamin D. The expression of both the vitamin D receptor and 25-hydroxyvitamin D 1α-hydroxylase (CYP27B1) genes has been shown in murine and human adipocytes. There is evidence that vitamin D affects body fat mass by inhibiting adipogenic transcription factors and lipid accumulation during adipocyte differentiation. Some recent studies demonstrate that vitamin D metabolites also influence adipokine production and the inflammatory response in adipose tissue. Therefore, vitamin D deficiency may compromise the normal metabolic functioning of adipose tissue. Given the importance of the tissue in energy balance, lipid metabolism and inflammation in obesity, understanding the mechanisms of vitamin D action in adipocytes may have a significant impact on the maintenance of metabolic health. In the present review, we focus on the signalling role of vitamin D in adipocytes, particularly the potential mechanisms through which vitamin D may influence adipose tissue development and function.


Assuntos
Adipogenia , Tecido Adiposo/metabolismo , Metabolismo dos Lipídeos , Receptores de Calcitriol/metabolismo , Transdução de Sinais , Vitamina D/metabolismo , Adipocinas/metabolismo , Tecido Adiposo/imunologia , Animais , Humanos , Resistência à Insulina , Obesidade/etiologia , Obesidade/imunologia , Obesidade/metabolismo , Deficiência de Vitamina D/imunologia , Deficiência de Vitamina D/metabolismo , Deficiência de Vitamina D/fisiopatologia
6.
Methods Mol Biol ; 2448: 1-18, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35167087

RESUMO

Brown adipose tissue (BAT) was first identified by Conrad Gessner in 1551, but it was only in 1961 that it was firmly identified as a thermogenic organ. Key developments in the subsequent two decades demonstrated that: (1) BAT is quantitatively important to non-shivering thermogenesis in rodents, (2) uncoupling of oxidative phosphorylation through a mitochondrial proton conductance pathway is the central mechanism by which heat is generated, (3) uncoupling protein-1 is the critical factor regulating proton leakage in BAT mitochondria. Following pivotal studies on cafeteria-fed rats and obese ob/ob mice, BAT was then shown to have a central role in the regulation of energy balance and the etiology of obesity. The application of fluorodeoxyglucose positron emission tomography in the late 2000s confirmed that BAT is present and active in adults, resulting in renewed interest in the tissue in human energetics and obesity. Subsequent studies have demonstrated a broad metabolic role for BAT, the tissue being an important site of glucose disposal and triglyceride clearance, as well as of insulin action. BAT continues to be a potential target for the treatment of obesity and related metabolic disorders.


Assuntos
Tecido Adiposo Marrom , Termogênese , Tecido Adiposo Marrom/metabolismo , Animais , Metabolismo Energético , Camundongos , Obesidade/metabolismo , Ratos , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
7.
Pflugers Arch ; 462(3): 469-77, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21698390

RESUMO

Adipose tissue becomes hypoxic in obesity, and cell culture studies have demonstrated that hypoxia leads to major changes in adipocyte function. Studies on the response of adipocytes to low O2 tension have employed marked hypoxia (1% O2). Here, we have examined the effects of modest hypoxia, utilising differing concentrations of O2 (1-21%), on adipokine production and glucose uptake by human adipocytes. Incubation with 10% O2 (24 h) increased expression of the leptin, vascular endothelial growth factor (VEGF) and Angptl4 genes, while leptin expression was elevated even at 15% O2 (compared to 'normoxia'-21% O2). Overall, there was a concentration-dependent increase in the expression of these genes as O2 fell, with the highest mRNA level evident at 1% O2. Parallel changes were observed in the secretion of leptin, VEGF and IL-6 into the medium, an increased release being evident at 10% O2 (15% O(2) for leptin). Adiponectin gene expression was reduced at 15% O2 and below, while adiponectin release was significantly reduced at 5% O2. Both 2-deoxy-D: -glucose uptake and lactate release showed progressive increases as O2 concentration fell, being significantly raised at 10% and 5% O2, respectively. The alterations in substrate transport were accompanied by parallel changes in transporter gene expression, GLUT1 and MCT1 mRNA level increasing from 15% and 10% O2, respectively. These results indicate that marked responses to reduced O2 concentration are exhibited by human adipocytes at O2 levels well above those associated with hypoxia and employed in cell culture studies. Adipocytes are sensitive to small changes in O2 tension.


Assuntos
Adipócitos/metabolismo , Adipocinas/biossíntese , Hipóxia Celular/fisiologia , Glucose/metabolismo , Ácido Láctico/metabolismo , Oxigênio/metabolismo , Adipócitos/citologia , Adulto , Proteína 4 Semelhante a Angiopoietina , Angiopoietinas/genética , Angiopoietinas/metabolismo , Animais , Células Cultivadas , Proteínas de Ligação a Ácido Graxo/genética , Proteínas de Ligação a Ácido Graxo/metabolismo , Feminino , Transportador de Glucose Tipo 1/genética , Transportador de Glucose Tipo 1/metabolismo , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Leptina/genética , Leptina/metabolismo , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Simportadores/genética , Simportadores/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
8.
Br J Nutr ; 106(9): 1310-6, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21902860

RESUMO

Lactate is increasingly recognised to be more than a simple end product of anaerobic glycolysis. Skeletal muscle and white adipose tissue are considered to be the main sites of lactate production and release. Recent studies have demonstrated that there is a specific G-protein coupled receptor for lactate, GPR81, which is expressed primarily in adipose tissue, and also in muscle. Lactate inhibits lipolysis in adipose tissue by mediating, through GPR81, the anti-lipolytic action of insulin. A high proportion (50 % or more) of the glucose utilised by white adipose tissue is converted to lactate and lactate production by the tissue increases markedly in obesity; this is likely to reflect a switch towards anaerobic metabolism with the development of hypoxia in the tissue. During exercise, there is a shift in fuel utilisation by muscle from lipid to carbohydrate, but this does not appear to be a result of the inhibition of lipolysis in the main adipose tissue depots by muscle-derived lactate. It is suggested instead that a putative autocrine lactate loop in myocytes may regulate fuel utilisation by muscle during exercise, operating via a muscle GPR81 receptor. In addition to being an important substrate, lactate is a key signal in metabolic regulation.


Assuntos
Tecido Adiposo Branco/metabolismo , Exercício Físico/fisiologia , Ácidos Graxos/metabolismo , Ácido Láctico/metabolismo , Músculo Esquelético/metabolismo , Obesidade/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Metabolismo dos Carboidratos , Metabolismo Energético , Glucose/metabolismo , Humanos , Hipóxia/metabolismo , Insulina/metabolismo , Metabolismo dos Lipídeos , Lipólise
11.
Proc Nutr Soc ; 80(1): 92-104, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32046799

RESUMO

The paper is based on the lecture that I gave on receiving the Nutrition Society's inaugural Gowland Hopkins Award for contributions to Cellular and Molecular Nutrition. It reviews studies on the adipose tissues, brown and white, conducted by the groups that I have led since entering nutrition research in 1975. The initial focus was on exploring metabolic factors that underpin the development of obesity using animal models. This resulted in an interest in non-shivering thermogenesis with brown adipose tissue being identified as the key effector of facultative heat production. Brown fat is less thermogenically active in various obese rodents, and major changes in activity are exhibited under physiological conditions such as lactation and fasting consistent with a general role for the tissue in nutritional energetics. My interests moved to white adipose tissue following the cloning of the Ob gene. Our initial contributions in this area included demonstrating nutritional regulation of Ob gene expression and circulating leptin levels, as well as a regulatory role for the sympathetic nervous system operating through ß3-adrenoceptors. My interests subsequently evolved to a wider concern with the endocrine/signalling role of adipose tissue. Inflammation is a characteristic of white fat in obesity with the release of inflammation-related adipokines, and we proposed that hypoxia underlies this inflammatory state. O2-deprivation was shown to have substantial effects on gene expression and cellular function in white adipocytes. The hypoxia studies led to the proposition that O2 should be considered as a critical macronutrient.


Assuntos
Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , Fenômenos Fisiológicos da Nutrição , Animais , Humanos , Obesidade/metabolismo , Termogênese
12.
Antioxid Redox Signal ; 35(8): 642-687, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34036800

RESUMO

Significance: In recent years, a number of studies have shown altered oxygen partial pressure at a tissue level in metabolic disorders, and some researchers have considered oxygen to be a (macro) nutrient. Oxygen availability may be compromised in obesity and several other metabolism-related pathological conditions, including sleep apnea-hypopnea syndrome, the metabolic syndrome (which is a set of conditions), type 2 diabetes, cardiovascular disease, and cancer. Recent Advances: Strategies designed to reduce adiposity and its accompanying disorders have been mainly centered on nutritional interventions and physical activity programs. However, novel therapies are needed since these approaches have not been sufficient to counteract the worldwide increasing rates of metabolic disorders. In this regard, intermittent hypoxia training and hyperoxia could be potential treatments through oxygen-related adaptations. Moreover, living at a high altitude may have a protective effect against the development of abnormal metabolic conditions. In addition, oxygen delivery systems may be of therapeutic value for supplying the tissue-specific oxygen requirements. Critical Issues: Precise in vivo methods to measure oxygenation are vital to disentangle some of the controversies related to this research area. Further, it is evident that there is a growing need for novel in vitro models to study the potential pathways involved in metabolic dysfunction to find appropriate therapeutic targets. Future Directions: Based on the existing evidence, it is suggested that oxygen availability has a key role in obesity and its related comorbidities. Oxygen should be considered in relation to potential therapeutic strategies in the treatment and prevention of metabolic disorders. Antioxid. Redox Signal. 35, 642-687.


Assuntos
Diabetes Mellitus Tipo 2 , Hiperóxia , Síndrome Metabólica , Humanos , Hiperóxia/metabolismo , Hipóxia , Oxigênio
13.
Pflugers Arch ; 459(3): 509-18, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19876643

RESUMO

Hypoxia modulates white adipose tissue function, and this includes stimulating glucose uptake and the expression of facilitative glucose transporters (particularly GLUT1) in adipocytes. This study has examined the effect of hypoxia on lactate release from adipocytes and whether the monocarboxylate transporters that mediate lactate transport (MCTs1-4) are expressed in human adipocytes and are induced by low O(2) tension. Exposure of human Simpson-Golabi-Behmel syndrome adipocytes to 1% O(2) for 24 h resulted in increased lactate release (2.3-fold) compared with cells in normoxia (21% O(2)). Screening by reverse transcription polymerase chain reaction indicated that the genes encoding MCT1, MCT2, and MCT4 are expressed in human adipose tissue, and in adipocytes and preadipocytes in culture. Hypoxia (48 h) increased MCT1 (8.5-fold) and MCT4 (14.3-fold) messenger RNA (mRNA) levels in human adipocytes, but decreased MCT2 mRNA (fourfold). MCT1 protein level was also increased (2.7-fold at 48 h) by hypoxia, but there was no change in MCT4 protein. The changes in MCT gene expression induced by hypoxia were reversed on return to normoxia. Treatment with the hypoxia mimetic CoCl(2) resulted in up-regulation of MCT1 (up to twofold) and MCT4 (fivefold) mRNA level, but there was no significant effect on MCT2 expression. It is concluded that hypoxia increases lactate release from adipocytes and modulates MCT expression in a type-specific manner, with MCT1 and MCT4 expression being hypoxia-inducible transcription factor-1 (HIF-1) dependent. Increased lactate production and monocarboxylate transporter expression are likely to be key components of the adaptive response of adipocytes to low O(2) tension as adipose tissue mass expands in obesity.


Assuntos
Adipócitos/metabolismo , Hipóxia/metabolismo , Ácido Láctico/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteínas Musculares/metabolismo , Simportadores/metabolismo , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Antimutagênicos/farmacologia , Células Cultivadas , Cobalto/farmacologia , Feminino , Regulação da Expressão Gênica , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Transportadores de Ácidos Monocarboxílicos/genética , Proteínas Musculares/genética , Oxigênio/metabolismo , Simportadores/genética , Síndrome
14.
Pflugers Arch ; 460(3): 603-16, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20473515

RESUMO

Adiposity and obesity are increasing in dogs. We have examined here the endocrine function of canine adipose tissue and the regulation of production of inflammation-related adipokines by dog adipocytes. Adiponectin, leptin, IL-6, MCP-1 and TNFalpha genes were expressed in the main adipose depots of dogs, but there were no major depot differences in mRNA levels. Each adipokine was expressed in canine adipocytes differentiated in culture and secreted into the medium (leptin undetected). IL-6, MCP-1 and TNFalpha were also expressed and secreted by preadipocytes; adiponectin and leptin were only expressed after adipocyte differentiation. The inflammatory mediators LPS and TNFalpha had major stimulatory effects on the expression and secretion of IL-6, MCP-1 and TNFalpha; there was a >5,000-fold increase in IL-6 mRNA level with LPS. IL-6 release into the medium was increased >50-fold over 24 h with LPS and TNFalpha, while MCP-1 release was increased 23- and 40-fold by TNFalpha and LPS, respectively. However, there was no effect, or small reductions, in adiponectin and leptin mRNA levels with the inflammatory mediators. Dexamethasone-stimulated leptin gene expression, had no effect on adiponectin expression, but decreased the expression and secretion of IL-6 and MCP-1. The PPARgamma agonist rosiglitazone stimulated both adiponectin and leptin expression and inhibited the expression of IL-6, MCP-1 and TNFalpha; MCP-1 secretion was reduced. These results demonstrate that canine adipocytes express and secrete key adipokines and show that adipocytes of this species are highly responsive to inflammatory mediators with the induction of major increases in the production of inflammation-related adipokines.


Assuntos
Adipócitos/metabolismo , Adiponectina/metabolismo , Citocinas/metabolismo , Leptina/metabolismo , Adipócitos/efeitos dos fármacos , Animais , Anti-Inflamatórios , Células Cultivadas , Dexametasona , Cães , Feminino , Hipoglicemiantes , Lipopolissacarídeos , Masculino , Rosiglitazona , Tiazolidinedionas , Fator de Necrose Tumoral alfa
15.
J Nutr Sci ; 9: e23, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32595967

RESUMO

Rodents with mutations in the leptin, or leptin receptor, genes have been extensively used to investigate the regulation of energy balance and the factors that underlie the development of obesity. The excess energy gain of these mutants has long been considered as being due in part to increased metabolic efficiency, consequent to reduced energy expenditure, but this view has recently been challenged. We argue, particularly though not exclusively, from data on ob/ob mice, that three lines of evidence support the proposition that reduced expenditure is important in the aetiology of obesity in leptin pathway mutants (irrespective of the genetic background): (i) milk intake is similar in suckling ob/ob and +/? mice; (ii) ob/ob mice deposit excess energy when pair-fed to the ad libitum food intake of lean siblings; (iii) in several studies mutant mice have been shown to exhibit a lower RMR 'per animal' at temperatures below thermoneutrality. When metabolic rate is expressed 'per unit body weight' (inappropriately, because of body composition differences), then it is invariably lower in the obese than the lean. It is important to differentiate the causes from the consequences of obesity. Hyperphagic, mature obese animals weighing 2-3 times their lean siblings may well have higher expenditure 'per animal', reflecting the costs of being larger and of enhanced obligatory diet-induced thermogenesis resulting from the increased food intake. This cannot, however, be used to inform the aetiology of their obesity.


Assuntos
Metabolismo Energético , Leptina/genética , Mutação , Obesidade/genética , Receptores para Leptina/genética , Animais , Composição Corporal , Ingestão de Alimentos , Hiperfagia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Leite , Termogênese
16.
Pflugers Arch ; 458(6): 1103-14, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19585142

RESUMO

White adipose tissue exhibits inflammation as tissue mass expands in obesity, involving macrophage infiltration and a direct inflammatory response by adipocytes. DNA microarrays and conditioned medium have been used to examine the effects of macrophages on global gene expression in human adipocytes. SGBS adipocytes, differentiated in culture, were treated with macrophage-conditioned medium (U937 cells) for 4 or 24 h; control cells received unconditioned medium. Agilent arrays comprising 44,000 probes were used to analyse gene expression. Microarray analysis identified 1,088 genes differentially expressed in response to the conditioned medium at both 4 and 24 h (754 up-regulated, 334 down-regulated at 24 h); these included genes associated with inflammation and macrophage infiltration. A cluster of matrix metalloproteinase genes were highly up-regulated at both time-points, including MMP1, MMP3, MMP9, MMP10, MMP12 and MMP19. At 4 and 24 h, MMP1 was the most highly up-regulated gene (>2,400-fold increase in mRNA at 24 h). ELISA measurements indicated that substantial quantities of MMP1 and MMP3 were released from adipocytes incubated with conditioned medium, with little release by control adipocytes. Treatment with TNFalpha induced substantial increases in MMP1 (>100-fold) and MMP3 (27-fold) mRNA level and MMP1 and MMP3 release in adipocytes, suggesting that this cytokine could contribute to the stimulation of MMP expression by macrophages. In conclusion, macrophage-secreted factors induce a major inflammatory response in human adipocytes, with expression of MMP family members being strongly up-regulated. The induction of MMP1 and other MMPs suggests that macrophages stimulate tissue remodelling during adipose tissue expansion in obesity.


Assuntos
Metaloproteinases da Matriz/genética , Adipócitos/metabolismo , Meios de Cultivo Condicionados/farmacologia , Humanos , Inflamação/etiologia , Macrófagos/fisiologia , Metaloproteinase 1 da Matriz/biossíntese , Metaloproteinase 3 da Matriz/biossíntese , Metaloproteinases da Matriz/biossíntese , Análise de Sequência com Séries de Oligonucleotídeos , Fator de Necrose Tumoral alfa/farmacologia , Células U937 , Regulação para Cima
17.
Biochem Biophys Res Commun ; 384(1): 105-9, 2009 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-19393621

RESUMO

Inflammation occurs in adipose tissue in obesity. We have examined whether IL-33, a recently identified IL-1 gene family member, and its associated receptors are expressed in human adipocytes. IL-33, IL-1RL1 and IL-1RAP gene expression was observed in human visceral white fat, in preadipocytes and in adipocytes (SGBS cells). Treatment with TNFalpha for 24h induced a 6-fold increase in IL-33 mRNA level in preadipocytes and adipocytes. Time-course studies with adipocytes showed that the increase in IL-33 mRNA with TNFalpha was maximal (>55-fold) at 12h. This response was markedly different to IL-1beta (peak mRNA increase at 2h; 5.4-fold) and 1L-18 (peak mRNA increase at 6h; >1500-fold). Exposure of adipocytes to hypoxia (1% O(2), 24h) did not alter IL-33 mRNA level; in preadipocytes, however, there was a 3-fold increase. Human adipocytes and preadipocytes express IL-33, but the various IL-1 family members exhibit major differences in responsiveness to TNFalpha.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo Branco/metabolismo , Expressão Gênica , Interleucina-1/genética , Interleucinas/genética , Células Cultivadas , Expressão Gênica/efeitos dos fármacos , Humanos , Interleucina-33 , Receptores de Interleucina-1/genética , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
18.
Front Nutr ; 6: 10, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30809528

RESUMO

Gaseous oxygen is essential for all aerobic animals, without which mitochondrial respiration and oxidative phosphorylation cannot take place. It is not, however, regarded as a "nutrient" by nutritionists and does not feature as such within the discipline of nutritional science. This is primarily a consequence of the route by which O2 enters the body, which is via the nose and lungs in terrestrial animals as opposed to the mouth and gastrointestinal tract for what are customarily considered as nutrients. It is argued that the route of entry should not be the critical factor in defining whether a substance is, or is not, a nutrient. Indeed, O2 unambiguously meets the standard dictionary definitions of a nutrient, such as "a substance that provides nourishment for the maintenance of life and for growth" (Oxford English Dictionary). O2 is generally available in abundance, but deficiency occurs at high altitude and during deep sea dives, as well as in lung diseases. These impact on the provision at a whole-body level, but a low pO2 is characteristic of specific tissues includings the retina and brain, while deficiency, or overt hypoxia, is evident in certain conditions such as ischaemic disease and in tumours - and in white adipose tissue in obesity. Hypoxia results in a switch from oxidative metabolism to increased glucose utilisation through anaerobic glycolysis, and there are extensive changes in the expression of multiple genes in O2-deficient cells. These changes are driven by hypoxia-sensitive transcription factors, particularly hypoxia-inducible factor-1 (HIF-1). O2 deficiency at a whole-body level can be treated by therapy or supplementation, but O2 is also toxic through the generation of reactive oxygen species. It is concluded that O2 is a critical, but overlooked, nutrient which should be considered as part of the landscape of nutritional science.

19.
Biochem Biophys Res Commun ; 368(1): 88-93, 2008 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-18206644

RESUMO

Hypoxia-signalling pathway PCR arrays were used to examine the integrated response of human adipocytes to low O(2) tension. Incubation of adipocytes in 1% O(2) for 24h resulted in no change in the expression of 63 of the 84 genes on the arrays, a reduction in expression of 9 genes (including uncoupling protein 2) and increased expression of 12 genes. Substantial increases (>10-fold) in leptin, angiopoietin-like protein 4, VEGF and GLUT-1 mRNA levels were observed. The expression of one gene, metallothionein-3 (MT-3), was dramatically (>600-fold) and rapidly (by 60 min) increased by hypoxia. MT-3 gene expression was also substantially induced by hypoxia mimetics (CoCl(2), desferrioxamine, dimethyloxalylglycine), indicating transcriptional regulation through HIF-1. Hypoxia additionally induced MT-3 expression in preadipocytes, and MT-3 mRNA was detected in human (obese) subcutaneous and omental adipose tissue. MT-3 is a highly hypoxia-inducible gene in human adipocytes; the protein may protect adipocytes from hypoxic damage.


Assuntos
Adipócitos/metabolismo , Regulação da Expressão Gênica , Fator 1 Induzível por Hipóxia/genética , Metalotioneína/genética , Células Cultivadas , Feminino , Humanos , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , RNA Mensageiro/genética , Transdução de Sinais , Técnicas de Cultura de Tecidos
20.
Br J Nutr ; 100(1): 18-26, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18081944

RESUMO

Angiopoietin-like protein 4 (Angptl4)/FIAF (fasting-induced adipose factor) was first identified as a target for PPAR and to be strongly induced in white adipose tissue (WAT) by fasting. Here we have examined the regulation of the expression and release of this adipokine in mouse WAT and in 3T3-L1 adipocytes. Angptl4/FIAF expression was measured by RT-PCR and real-time PCR; plasma Angptl4/FIAF and release of the protein in cell culture was determined by western blotting. The Angptl4/FIAF gene was expressed in each of the major WAT depots of mice, the mRNA level in WAT being similar to the liver and much higher (>50-fold) than skeletal muscle. Fasting mice (18 h) resulted in a substantial increase in Angptl4/FIAF mRNA in liver and muscle (9.5- and 21-fold, respectively); however, there was no effect of fasting on Angptl4/FIAF mRNA in WAT and the plasma level of Angptl4/FIAF was unchanged. The Angptl4/FIAF gene was expressed in 3T3-L1 adipocytes before and after differentiation, the level increasing post-differentiation; Angptl4/FIAF was released into the culture medium. Insulin, leptin, dexamethasone, noradrenaline, TNFalpha and several IL (IL-1beta, IL-6, IL-10, IL-18) had little effect on Angptl4/FIAF mRNA levels in 3T3-L1 adipocytes. However, a major stimulation of Angptl4/FIAF expression was observed with rosiglitazone and the inflammatory prostaglandins PGD2 and PGJ2. Angptl4/FIAF does not act as an adipose tissue signal of nutritional status, but is markedly induced by fasting in liver and skeletal muscle.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo Branco/metabolismo , Angiopoietinas/biossíntese , Regulação da Expressão Gênica , Adipócitos/citologia , Proteína 4 Semelhante a Angiopoietina , Angiopoietinas/genética , Animais , Diferenciação Celular , Células Cultivadas , Jejum/metabolismo , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos , Músculo Esquelético/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA