Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 221, 2024 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-38177096

RESUMEN

Lymphedema (LD) is characterized by the accumulation of interstitial fluid, lipids and inflammatory cell infiltrate in the limb. Here, we find that LD tissues from women who developed LD after breast cancer exhibit an inflamed gene expression profile. Lipidomic analysis reveals decrease in specialized pro-resolving mediators (SPM) generated by the 15-lipoxygenase (15-LO) in LD. In mice, the loss of SPM is associated with an increase in apoptotic regulatory T (Treg) cell number. In addition, the selective depletion of 15-LO in the lymphatic endothelium induces an aggravation of LD that can be rescued by Treg cell adoptive transfer or ALOX15-expressing lentivector injections. Mechanistically, exogenous injections of the pro-resolving cytokine IFN-ß restores both 15-LO expression and Treg cell number in a mouse model of LD. These results provide evidence that lymphatic 15-LO may represent a therapeutic target for LD by serving as a mediator of Treg cell populations to resolve inflammation.


Asunto(s)
Araquidonato 15-Lipooxigenasa , Linfedema , Humanos , Ratones , Femenino , Animales , Araquidonato 15-Lipooxigenasa/genética , Araquidonato 15-Lipooxigenasa/metabolismo , Inflamación/metabolismo , Citocinas/metabolismo , Linfocitos T Reguladores/metabolismo
2.
Nat Commun ; 10(1): 2549, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31186409

RESUMEN

Human adipose tissue (hAT) is constituted of structural units termed lobules, the organization of which remains to be defined. Here we report that lobules are composed of two extracellular matrix compartments, i.e., septa and stroma, delineating niches of CD45-/CD34+/CD31- progenitor subsets characterized by MSCA1 (ALPL) and CD271 (NGFR) expression. MSCA1+ adipogenic subset is enriched in stroma while septa contains mainly MSCA1-/CD271- and MSCA1-/CD271high progenitors. CD271 marks myofibroblast precursors and NGF ligand activation is a molecular relay of TGFß-induced myofibroblast conversion. In human subcutaneous (SC) and visceral (VS) AT, the progenitor subset repartition is different, modulated by obesity and in favor of adipocyte and myofibroblast fate, respectively. Lobules exhibit depot-specific architecture with marked fibrous septa containing mesothelial-like progenitor cells in VSAT. Thus, the human AT lobule organization in specific progenitor subset domains defines the fat depot intrinsic capacity to remodel and may contribute to obesity-associated cardiometabolic risks.


Asunto(s)
Tejido Adiposo/anatomía & histología , Tejido Adiposo/citología , Nicho de Células Madre , Células Madre/citología , Adipocitos/metabolismo , Adipogénesis , Fosfatasa Alcalina , Diferenciación Celular , Matriz Extracelular , Humanos , Grasa Intraabdominal/citología , Miofibroblastos/citología , Miofibroblastos/efectos de los fármacos , Proteínas del Tejido Nervioso/metabolismo , Obesidad , Receptores de Factor de Crecimiento Nervioso/metabolismo , Células Madre/metabolismo , Grasa Subcutánea/citología , Factor de Crecimiento Transformador beta1/farmacología
3.
Int J Obes (Lond) ; 39(5): 762-9, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25388403

RESUMEN

BACKGROUND/OBJECTIVES: The presence of T lymphocytes in human adipose tissue has only recently been demonstrated and relatively little is known of their potential relevance in the development of obesity-related diseases. We aimed to further characterise these cells and in particular to investigate how they interact with modestly increased levels of adiposity typical of common overweight and obesity. SUBJECTS/METHODS: Subcutaneous adipose tissue and fasting blood samples were obtained from healthy males aged 35-55 years with waist circumferences in lean (<94 cm), overweight (94-102 cm) and obese (>102 cm) categories. Adipose tissue-resident CD4+ and CD8+ T lymphocytes together with macrophages were identified by gene expression and flow cytometry. T lymphocytes were further characterised by their expression of activation markers CD25 and CD69. Adipose tissue inflammation was investigated using gene expression analysis and tissue culture. RESULTS: Participants reflected a range of adiposity from lean to class I obesity. Expression of CD4 (T-helper cells) and CD68 (macrophage), as well as FOXP3 RNA transcripts, was elevated in subcutaneous adipose tissue with increased levels of adiposity (P<0.001, P<0.001 and P=0.018, respectively). Flow cytometry revealed significant correlations between waist circumference and levels of CD25 and CD69 expression per cell on activated adipose tissue-resident CD4+ and CD8+ T lymphocytes (P-values ranging from 0.053 to <0.001). No such relationships were found with blood T lymphocytes. This increased T lymphocyte activation was related to increased expression and secretion of various pro- and anti-inflammatory cytokines from subcutaneous whole adipose tissue explants. CONCLUSIONS: This is the first study to demonstrate that even modest levels of overweight/obesity elicit modifications in adipose tissue immune function. Our results underscore the importance of T lymphocytes during adipose tissue expansion, and the presence of potential compensatory mechanisms that may work to counteract adipose tissue inflammation, possibly through an increased number of T-regulatory cells.


Asunto(s)
Tejido Adiposo/metabolismo , Adiposidad/inmunología , Inflamación/metabolismo , Activación de Linfocitos , Macrófagos/metabolismo , Sobrepeso/metabolismo , Linfocitos T Reguladores/metabolismo , Adiposidad/fisiología , Adulto , Composición Corporal , Humanos , Resistencia a la Insulina , Masculino , Persona de Mediana Edad , Sobrepeso/fisiopatología , Reacción en Cadena en Tiempo Real de la Polimerasa
4.
Int J Obes (Lond) ; 38(9): 1234-40, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24406482

RESUMEN

BACKGROUND: Prader-Willi syndrome (PWS) results from abnormalities in the genomic imprinting process leading to hypothalamic dysfunction with an alteration of growth hormone (GH) secretion. PWS is associated with early morbid obesity and short stature which can be efficiently improved with GH treatment. OBJECTIVES: Our aims were to highlight adipose tissue structural and functional impairments in children with PWS and to study the modifications of those parameters on GH treatment. SUBJECTS AND METHODS: Plasma samples and adipose tissue biopsies were obtained from 23 research centers in France coordinated by the reference center for PWS in Toulouse, France. Lean controls (n=33), non-syndromic obese (n=53), untreated (n=26) and GH-treated PWS (n=43) children were enrolled in the study. Adipose tissue biopsies were obtained during scheduled surgeries from 15 lean control, 7 untreated and 8 GH-treated PWS children. RESULTS: Children with PWS displayed higher insulin sensitivity as shown by reduced glycemia, insulinemia and HOMA-IR compared with non-syndromic obese children. In contrast, plasma inflammatory cytokines such as TNF-α, MCP-1 and IL-8 were increased in PWS. Analysis of biopsies compared with control children revealed decreased progenitor cell content in the stromal vascular fraction of adipose tissue and an impairment of lipolytic response to ß-adrenergic agonist in PWS adipocytes. Interestingly, both of these alterations in PWS seem to be ameliorated on GH treatment. CONCLUSION: Herein, we report adipose tissue dysfunctions in children with PWS which may be partially restored by GH treatment.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Estatura/efectos de los fármacos , Terapia de Reemplazo de Hormonas , Hormona de Crecimiento Humana/uso terapéutico , Obesidad Mórbida/tratamiento farmacológico , Obesidad Infantil/tratamiento farmacológico , Síndrome de Prader-Willi/tratamiento farmacológico , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Adolescente , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Composición Corporal , Niño , Preescolar , Femenino , Francia , Humanos , Lactante , Lipólisis , Masculino , Obesidad Mórbida/etiología , Obesidad Mórbida/metabolismo , Obesidad Infantil/etiología , Obesidad Infantil/metabolismo , Síndrome de Prader-Willi/complicaciones , Síndrome de Prader-Willi/metabolismo , Resultado del Tratamiento , Adulto Joven
5.
Diabetologia ; 56(8): 1792-801, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23666167

RESUMEN

AIMS/HYPOTHESIS: Alterations in white adipose tissue (WAT) function, including changes in protein (adipokine) secretion and extracellular matrix (ECM) composition, promote an insulin-resistant state. We set out to identify novel adipokines regulated by body fat mass in human subcutaneous WAT with potential roles in adipose function. METHODS: Adipose transcriptome data and secretome profiles from conditions with increased/decreased WAT mass were combined. WAT donors were predominantly women. In vitro effects were assessed using recombinant protein. Results were confirmed by quantitative PCR/ELISA, metabolic assays and immunochemistry in human WAT and adipocytes. RESULTS: We identified a hitherto uncharacterised adipokine, semaphorin 3C (SEMA3C), the expression of which correlated significantly with body weight, insulin resistance (HOMA of insulin resistance [HOMAIR], and the rate constant for the insulin tolerance test [KITT]) and adipose tissue morphology (hypertrophy vs hyperplasia). SEMA3C was primarily found in mature adipocytes and had no direct effect on human adipocyte differentiation, lipolysis, glucose transport or the expression of ß-oxidation genes. This could in part be explained by the significant downregulation of its cognate receptors during adipogenesis. In contrast, in pre-adipocytes, SEMA3C increased the production/secretion of several ECM components (fibronectin, elastin and collagen I) and matricellular factors (connective tissue growth factor, IL6 and transforming growth factor-ß1). Furthermore, the expression of SEMA3C in human WAT correlated positively with the degree of fibrosis in WAT. CONCLUSIONS/INTERPRETATION: SEMA3C is a novel adipokine regulated by weight changes. The correlation with WAT hypertrophy and fibrosis in vivo, as well as its effects on ECM production in human pre-adipocytes in vitro, together suggest that SEMA3C constitutes an adipocyte-derived paracrine signal that influences ECM composition and may play a pathophysiological role in human WAT.


Asunto(s)
Adipoquinas/metabolismo , Matriz Extracelular/metabolismo , Semaforinas/metabolismo , Adipoquinas/genética , Tejido Adiposo Blanco/metabolismo , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Microscopía Confocal , Semaforinas/genética
6.
Int J Obes (Lond) ; 36(9): 1256-8, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22083548

RESUMEN

Recent evidence indicates that adipose tissue macrophages and lymphocytes have a major role in the pathophysiology of obesity. The arterio-venous (A-V) difference technique has been used very effectively to understand adipose tissue metabolism in humans in vivo, and we set out to investigate whether it is possible to apply and use this technique to determine A-V differences for peripheral blood mononuclear cells (PBMCs) across human adipose tissue. We used flow-cytometric analysis of arterial blood and venous blood draining upper- (abdominal) and lower-body (femoral) adipose tissue depots in middle-aged volunteers (age 45±8 years, BMI 25.9±4.1 kg m(-2)). We determined A-V differences for various PBMCs. In basal conditions, there was evidence of modest retention of some PBMCs in adipose tissue, whereas the infusion of low-dose (physiological) adrenaline led to a marked release of many PBMCs (with little evidence of depot-specific differences). In addition to the demonstration that this approach is technically feasible, these results also indicate that physiological stimuli that change adrenaline concentrations and/or adipose tissue blood flow (such as physical activity) provoke the release of PBMCs from femoral and abdominal adipose depots.


Asunto(s)
Tejido Adiposo/metabolismo , Agonistas Adrenérgicos/farmacología , Arterias/metabolismo , Epinefrina/farmacología , Leucocitos Mononucleares/metabolismo , Obesidad/metabolismo , Venas/metabolismo , Tejido Adiposo/efectos de los fármacos , Agonistas Adrenérgicos/administración & dosificación , Arterias/efectos de los fármacos , Epinefrina/administración & dosificación , Femenino , Citometría de Flujo , Humanos , Infusiones Intravenosas , Leucocitos Mononucleares/efectos de los fármacos , Masculino , Persona de Mediana Edad , Obesidad/tratamiento farmacológico , Flujo Sanguíneo Regional , Venas/efectos de los fármacos
7.
Diabetes Metab ; 37(4): 283-90, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21507694

RESUMEN

Obesity, defined as the excess development of adipose tissue, is an important risk factor for metabolic and cardiovascular diseases such as type 2 diabetes, hypertension and atherosclerosis. Over the past few years, metabolic inflammation has emerged as a major process underlying the link between obesity and its associated pathologies. Adipose tissue appears to play a primary and crucial role as a source and site of inflammation. Accumulation of immune cells within adipose tissue occurs in obese conditions. The present review focuses on the relationship between adipose tissue and immune cells, including macrophages, dendritic cells, T and B lymphocytes, and natural killer cells, in both the physiological state and under obese conditions. The factors involved in the accumulation of both myeloid and lymphoid cells in adipose tissue are also described. In addition, the role of adipose-tissue immune cells on adipocyte metabolism and cells of the adipose tissue stromal-vascular fraction are discussed, with particular emphasis on the cross-talk between macrophages and adipocytes, together with recent reports of T lymphocytes in adipose tissue.


Asunto(s)
Tejido Adiposo/inmunología , Tejido Adiposo/patología , Enfermedades Metabólicas/inmunología , Enfermedades Metabólicas/patología , Adipoquinas/inmunología , Animales , Células Dendríticas/inmunología , Humanos , Inflamación/inmunología , Inflamación/patología , Linfocitos/inmunología , Ratones , Obesidad/inmunología , Obesidad/patología
8.
Int J Obes (Lond) ; 35(12): 1502-10, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21386798

RESUMEN

OBJECTIVE: Tartrate-resistant acid phosphatase (TRAP) expressed by adipose tissue macrophages (ATMs) induces mice obesity and human adipocyte differentiation in vitro. This study aimed to investigate whether TRAP was secreted differently from human obese versus lean adipose tissues and to identify the cellular source of adipose tissue TRAP. DESIGN: Subcutaneous adipose tissues obtained from healthy subjects. Enzyme-linked immunosorbent assays (ELISAs) for total (5a+5b) and cleaved TRAP (5b) were used. TRAP secretion was determined in adipose tissue biopsies, and mRNA expression was studied in cell types isolated from the same. SUBJECTS: Results of 24 lean and 24 obese women (in vitro) and 8 subjects (in vivo) were compared. The main outcome measurements were TRAP expression and secretion in vitro and in vivo. RESULTS: In-house total TRAP ELISA showed high sensitivity and a coefficient of variance of 11%. Adipose secretion of total TRAP was linear in vitro with time and was evident in vivo. Total TRAP secretion in vitro was similar in lean and obese women expressed per unit weight of the adipose tissue but correlated positively with the number/size of adipocytes (P ≤ 0.01) and with adipose secretion of tumor necrosis factor-α and interleukin-6 (P<0.01). TRAP 5b was not secreted from the adipose tissue. ATMs displayed highest cellular expression of TRAP mRNA in adipose tissue cells derived from lean or obese women. CONCLUSIONS: TRAP is a novel human adipokine produced by macrophages and secreted from the subcutaneous adipose tissue in vivo and in vitro. Secretion is linked to the size and number of adipocytes, as well as to concomitant secretion of inflammatory mediators, suggesting that TRAP is involved in fat accumulation and adipose inflammation.


Asunto(s)
Fosfatasa Ácida/metabolismo , Adipoquinas/metabolismo , Isoenzimas/metabolismo , Macrófagos/metabolismo , Obesidad/metabolismo , Grasa Subcutánea/metabolismo , Delgadez/metabolismo , Adulto , Anciano , Análisis de Varianza , Animales , Biomarcadores/metabolismo , Peso Corporal , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Resistencia a la Insulina , Interleucina-6/metabolismo , Macrófagos/citología , Masculino , Ratones , Ratones Obesos , Persona de Mediana Edad , ARN Mensajero , Fosfatasa Ácida Tartratorresistente , Factor de Necrosis Tumoral alfa/metabolismo
9.
Int J Obes (Lond) ; 35(9): 1141-53, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21266947

RESUMEN

OBJECTIVES: Beside having roles in energy homeostasis and endocrine modulation, adipose tissue (AT) is now considered a promising source of mesenchymal stromal cells (adipose-derived stromal cells or ASCs) for regenerative medicine. Despite numerous studies on cultured ASCs, native human ASCs are rarely investigated. Indeed, the phenotype of ASCs in their native state, their localization within AT and comparison with bone marrow-derived mesenchymal stromal cells (BM-MSCs) has been poorly investigated. DESIGN: To address these issues, the stroma vascular fraction (SVF) of human AT was extracted and native cell subtypes were isolated by immunoselection to study their clonogenic potential in culture. Immunohistology on samples of human AT in combination with reconstruction of confocal sections were performed in order to localize ASCs. RESULTS: Compared with BM-MNCs, all native ASCs were found in the CD34(+) cell fraction of the AT-SVF. Native ASCs expressed classical mesenchymal markers described for BM-MSCs. Interestingly, CD34 expression decreased during ASC cell culture and was negatively correlated with cell proliferation rate. Immunohistological analysis revealed that native ASCs exhibited specific morphological features with protrusions. They were found scattered in AT stroma and did not express in vivo pericytic markers such as NG2, CD140b or alpha-smooth muscle actin, which appeared during the culture process. Finally, ASCs spontaneous commitment to adipocytic lineage was enhanced in AT from obese humans. CONCLUSIONS: The use of complementary methodological approaches to study native human ASCs revealed their immunophenotype, their specific morphology, their location within AT and their stemness. Furthermore, our data strongly suggest that human ASCs participate in adipogenesis during AT development.


Asunto(s)
Adipogénesis , Tejido Adiposo/citología , Diferenciación Celular/fisiología , Células Madre Mesenquimatosas , Obesidad , Células del Estroma , Adipogénesis/genética , Adulto , Biomarcadores , Diferenciación Celular/genética , Células Cultivadas , Femenino , Citometría de Flujo , Humanos , Inmunofenotipificación , Células Madre Mesenquimatosas/citología , Obesidad/genética , Fenotipo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células del Estroma/citología
10.
Diabetologia ; 54(4): 876-87, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21267541

RESUMEN

AIMS/HYPOTHESIS: Our goal was to identify a set of human adipose tissue macrophage (ATM)-specific markers and investigate whether their gene expression in subcutaneous adipose tissue (SAT) as well as in visceral adipose tissue (VAT) is related to obesity and to the occurrence of the metabolic syndrome. METHODS: ATM-specific markers were identified by DNA microarray analysis of adipose tissue cell types isolated from SAT of lean and obese individuals. We then analysed gene expression of these markers by reverse transcription quantitative PCR in paired samples of SAT and VAT from 53 women stratified into four groups (lean, overweight, obese and obese with the metabolic syndrome). Anthropometric measurements, euglycaemic-hyperinsulinaemic clamp, blood analysis and computed tomography scans were performed. RESULTS: A panel of 24 genes was selected as ATM-specific markers based on overexpression in ATM compared with other adipose tissue cell types. In SAT and VAT, gene expression of ATM markers was lowest in lean and highest in the metabolic syndrome group. mRNA levels in the two fat depots were negatively correlated with glucose disposal rate and positively associated with indices of adiposity and the metabolic syndrome. CONCLUSIONS/INTERPRETATION: In humans, expression of ATM-specific genes increases with the degree of adiposity and correlates with markers of insulin resistance and the metabolic syndrome to a similar degree in SAT and in VAT.


Asunto(s)
Tejido Adiposo/citología , Grasa Intraabdominal/citología , Grasa Intraabdominal/metabolismo , Macrófagos/metabolismo , Síndrome Metabólico/metabolismo , Obesidad/metabolismo , Grasa Subcutánea/citología , Grasa Subcutánea/metabolismo , Tejido Adiposo/metabolismo , Adulto , Anciano , Células Cultivadas , Femenino , Humanos , Persona de Mediana Edad , Sobrepeso/metabolismo , Adulto Joven
11.
Int J Obes (Lond) ; 35(1): 91-8, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20531347

RESUMEN

OBJECTIVE: Accumulation of adipose tissue macrophages (ATMs) is observed in obesity and may participate in the development of insulin resistance and obesity-related complications. The aim of our study was to investigate the effect of long-term dietary intervention on ATM content in human adipose tissue. DESIGN: We performed a multi-phase longitudinal study. SUBJECTS AND MEASUREMENTS: A total of 27 obese pre-menopausal women (age 39 ± 2 years, body mass index 33.7 ± 0.5 kg m(-2)) underwent a 6-month dietary intervention consisting of two periods: 4 weeks of very low-calorie diet (VLCD) followed by weight stabilization composed of 2 months of low-calorie diet and 3 to 4 months of weight maintenance diet. At baseline and at the end of each dietary period, samples of subcutaneous adipose tissue (SAT) were obtained by needle biopsy and blood samples were drawn. ATMs were determined by flow cytometry using combinations of cell surface markers. Selected cytokine and chemokine plasma levels were measured using enzyme-linked immunosorbent assay. In addition, in a subgroup of 16 subjects, gene expression profiling of macrophage markers in SAT was performed using real-time PCR. RESULTS: Dietary intervention led to a significant decrease in body weight, plasma insulin and C-reactive protein levels. After VLCD, ATM content defined by CD45+/14+/206+ did not change, whereas it decreased at the end of the intervention. This decrease was associated with a downregulation of macrophage marker mRNA levels (CD14, CD163, CD68 and LYVE-1 (lymphatic vessel endothelial hyaluronan receptor-1)) and plasma levels of monocyte-chemoattractant protein-1 (MCP-1) and CXCL5 (chemokine (C-X-C motif) ligand 5). During the whole dietary intervention, the proportion of two ATM subpopulations distinguished by the CD16 marker was not changed. CONCLUSION: A 6-month weight-reducing dietary intervention, but not VLCD, promotes a decrease in the number of the whole ATM population with no change in the relative distribution of ATM subsets.


Asunto(s)
Tejido Adiposo Blanco/patología , Dieta Reductora , Macrófagos/patología , Obesidad/patología , Pérdida de Peso , Adulto , Índice de Masa Corporal , Peso Corporal , Proteína C-Reactiva/genética , Quimiocina CXCL5/genética , Regulación hacia Abajo , Femenino , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Estudios Longitudinales , Obesidad/dietoterapia , Obesidad/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas de Transporte Vesicular/genética , Pérdida de Peso/genética
12.
Diabetes Metab ; 35(4): 251-60, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19539513

RESUMEN

Obesity is associated with systemic chronic low-grade inflammation, a major contributor to the aetiology of insulin resistance (IR). An inflammatory response in the presence of obesity appears to be triggered by, and to reside predominantly in, adipose tissue (AT). The discovery that the AT in obese mice and humans is infiltrated with macrophages has provided a major advance in our understanding of how obesity propagates inflammation. Interestingly, AT-infiltrating macrophages exhibit a proinflammatory phenotype (classical activation) whereas macrophages residing in AT have a reparative phenotype (alternative activation). In this review, the processes involved in monocyte/macrophage recruitment into the AT, and the events underlying the activation of infiltrating and/or resident AT macrophages (ATM) are described. Also, the localized roles of ATM on AT growth, metabolism and remodelling, as well as their systemic effects in promoting IR, are revealed. Finally, the new therapeutic targets that have recently emerged, and which have the potential to modulate the recruitment and/or activation of ATM, are discussed.


Asunto(s)
Resistencia a la Insulina/fisiología , Macrófagos/fisiología , Obesidad/fisiopatología , Tejido Adiposo/fisiopatología , Animales , Humanos , Inflamación/fisiopatología
14.
Circulation ; 117(6): 806-15, 2008 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-18227385

RESUMEN

BACKGROUND: Adipose tissue macrophages (ATMs) have become a focus of attention recently because they have been shown to accumulate with an increase in fat mass and to be involved in the genesis of insulin resistance in obese mice. However, the phenotype and functions of human ATMs are still to be defined. METHODS AND RESULTS: The present study, performed on human subcutaneous AT, showed that ATMs from lean to overweight individuals are composed of distinct macrophage subsets based on the expression of several cell surface markers: CD45, CD14, CD31, CD44, HLA-DR, CD206, and CD16, as assessed by flow cytometry. ATMs isolated by an immunoselection protocol showed a mixed expression of proinflammatory (tumor necrosis factor-alpha, interleukin-6 [IL-6], IL-23, monocyte chemoattractant protein-1, IL-8, cyclooxygenase-2) and antiinflammatory (IL-10, transforming growth factor-beta, alternative macrophage activation-associated cc chemokine-1, cyclooxygenase-1) factors. Fat mass enlargement is associated with accumulation of the CD206+/CD16- macrophage subset that exhibits an M2 remodeling phenotype characterized by decreased expression of proinflammatory IL-8 and cyclooxygenase-2 and increased expression of lymphatic vessel endothelial hyaluronan receptor-1. ATMs specifically produced and released matrix metalloproteinase-9 compared with adipocytes and capillary endothelial cells, and secretion of matrix metalloproteinase-9 from human AT in vivo, assessed by arteriovenous difference measurement, was correlated with body mass index. Finally, ATMs exerted a marked proangiogenic effect on AT-derived endothelial and progenitor cells. CONCLUSIONS: The present results showed that the ATMs that accumulate with fat mass development exhibit a particular M2 remodeling phenotype. ATMs may be active players in the process of AT development through the extension of the capillary network and in the genesis of obesity-associated cardiovascular pathologies.


Asunto(s)
Macrófagos/inmunología , Grasa Subcutánea/citología , Antígenos CD , Índice de Masa Corporal , Células Cultivadas , Femenino , Citometría de Flujo , Humanos , Macrófagos/enzimología , Metaloproteinasa 9 de la Matriz/biosíntesis , Fenotipo
15.
J Intern Med ; 262(4): 415-21, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17875177

RESUMEN

In recent years, the general concept has emerged that chronic low-grade inflammation can be the condition linking excessive development of adipose tissue (AT) and obesity-associated pathologies such as type II diabetes and atherosclerosis. Moreover, the evidence that the growth of the fat mass was associated with an accumulation of adipose tissue macrophages (ATM) has raised the hypothesis that the development of an inflammatory process within the growing fat mass is a primary event involved in the genesis of systemic metabolic and vascular alterations. As ATM originate from the bone marrow/blood compartment, enhanced macrophage recruitment to growing AT is suspected. However, the mechanisms responsible for attracting the blood cells and their entry into the fat mass remain to be clearly defined. The present review highlights the key role of endothelial cells in the control of the inflammatory process and describes the potential involvement of AT-endothelial cells as well as the factors involved in the regulation of their phenotype in the 'inflamed fat tissue'.


Asunto(s)
Diabetes Mellitus Tipo 2/patología , Células Endoteliales/metabolismo , Obesidad/patología , Paniculitis/patología , Adipocitos/patología , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Aterosclerosis/patología , Moléculas de Adhesión Celular/metabolismo , Comunicación Celular/fisiología , Quimiocinas/metabolismo , Angiopatías Diabéticas/patología , Humanos , Resistencia a la Insulina/fisiología , Macrófagos/patología , Obesidad/complicaciones , Obesidad/metabolismo
16.
Diabetologia ; 50(5): 1038-47, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17318625

RESUMEN

AIMS/HYPOTHESIS: Increased adipose tissue secretion of adipokines and cytokines has been implicated in the chronic low-grade inflammation state and insulin resistance associated with obesity. We tested here whether the cardiovascular and metabolic hormone atrial natriuretic peptide (ANP) was able to modulate adipose tissue secretion of several adipokines (derived from adipocytes) and cytokines (derived from adipose tissue macrophages). SUBJECTS AND METHODS: We used protein array to measure the secretion of adipokines and cytokines after a 24-h culture of human subcutaneous adipose tissue pieces treated or not with a physiological concentration of ANP. The effect of ANP on protein secretion was also directly studied on isolated adipocytes and macrophages. Gene expression was measured by real-time RT-quantitative PCR. RESULTS: ANP decreased the secretion of the pro-inflammatory cytokines IL-6 and TNF-alpha, of several chemokines, and of the adipokines leptin and retinol-binding protein-4 (RBP-4). The secretion of the anti-inflammatory molecules IL-10 and adiponectin remained unaffected. The cytokines were mainly expressed in macrophages that expressed all components of the ANP-dependent signalling pathway. The adipokines, leptin, adiponectin and RBP-4 were specifically expressed in mature adipocytes. ANP directly inhibited the secretion of IL-6 and monocyte chemoattractant protein-1 by macrophages. The inhibitory effects of ANP on leptin and growth-related oncogene-alpha secretions were not seen under selective hormone-sensitive lipase inhibition. CONCLUSIONS/INTERPRETATION: We suggest that ANP, either by direct action on adipocytes and macrophages or through activation of adipocyte hormone-sensitive lipase, inhibits the secretion of factors involved in inflammation and insulin resistance.


Asunto(s)
Adiponectina/biosíntesis , Tejido Adiposo/fisiología , Factor Natriurético Atrial/farmacología , Citocinas/biosíntesis , Inflamación/fisiopatología , Resistencia a la Insulina , Abdomen , Adipocitos/efectos de los fármacos , Adipocitos/fisiología , Adiponectina/antagonistas & inhibidores , Tejido Adiposo/efectos de los fármacos , Adulto , Células Cultivadas , Citocinas/antagonistas & inhibidores , Femenino , Humanos , Macrófagos/fisiología , Persona de Mediana Edad , Sobrepeso , Reacción en Cadena de la Polimerasa , ARN/genética , ARN/aislamiento & purificación
17.
Diabetologia ; 49(4): 744-7, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16496121

RESUMEN

AIMS/HYPOTHESIS: Increased visceral white adipose tissue (WAT) is linked to the risk of developing diabetes. METHODS/RESULTS: We showed by fluorescence activated cell sorting analysis that human visceral WAT contains macrophages, the proportion of which increased with obesity. Selective isolation of mature adipocytes and macrophages from human visceral WAT by CD14 immunoselection revealed that macrophages expressed higher levels of chemokines (monocyte chemotactic protein 1, macrophage inflammatory protein 1alpha, IL-8) and the adipokines resistin and visfatin than did mature adipocytes, as assessed by real-time PCR analysis. Moreover, resistin and visfatin proteins were found to be released predominantly by visceral WAT macrophages. Macrophage-derived secretory products stimulated phosphorylation of protein kinase B in human hepatocytes. CONCLUSIONS/INTERPRETATION: Resistin and visfatin might be considered to be proinflammatory markers. The increased macrophage population in obese human visceral WAT might be responsible for the enhanced production of chemokines as well as resistin and visfatin.


Asunto(s)
Grasa Abdominal/metabolismo , Citocinas/metabolismo , Macrófagos/metabolismo , Obesidad/metabolismo , Resistina/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Movimiento Celular , Femenino , Hepatocitos/metabolismo , Humanos , Receptores de Lipopolisacáridos/metabolismo , Macrófagos/citología , Masculino , Persona de Mediana Edad , Nicotinamida Fosforribosiltransferasa
18.
Int J Obes (Lond) ; 30(3): 447-52, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16261187

RESUMEN

OBJECTIVE: Inflammation in adipose tissue may link obesity to insulin resistance and atherosclerosis. Arachidonate 5-lipoxygenase activating protein (ALOX5AP) gene is involved in the pathogenesis of atherosclerotic cardiovascular disease (CVD). We investigated ALOX5AP expression in adipose tissue, and association of gene polymorphisms with obesity and insulin resistance. DESIGN: For gene expression analysis in adipose tissue, we studied 12 lean and 36 obese women, eight lean and 13 obese men, and nine women before and 2-4 years after gastric banding surgery. For genetic analysis, we studied 231 nonobese and 350 obese men. RESULTS: The ALOX5AP protein, 5-lipoxygenase activating protein (FLAP), as well as 5-lipoxygenase (5LO) itself, were detected in adipocytes. The mRNA expression of ALOX5AP in subcutaneous adipose tissue was increased in obesity and normalized following weight reduction. High adipose tissue mRNA expression of ALOX5AP is associated with insulin resistance as measured by homeostasis model assessment (HOMA(IR)). ALOX5AP haplotypes that associate with CVD are not associated with obesity or insulin resistance. CONCLUSION: ALOX5AP is present in adipose tissue, where its expression is associated with body weight and HOMA(IR), and may provide a link between adipose tissue, inflammation and insulin resistance. Investigated ALOX5AP haplotypes are not major primary risk factors for obesity and insulin resistance.


Asunto(s)
Proteínas Portadoras/metabolismo , Resistencia a la Insulina , Proteínas de la Membrana/metabolismo , Obesidad/metabolismo , Proteínas Activadoras de la 5-Lipooxigenasa , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Anciano , Alelos , Proteínas Portadoras/genética , Expresión Génica , Predisposición Genética a la Enfermedad , Haplotipos , Homeostasis , Humanos , Resistencia a la Insulina/genética , Masculino , Proteínas de la Membrana/genética , Persona de Mediana Edad , Obesidad/genética , Fenotipo , Reacción en Cadena de la Polimerasa/métodos , ARN Mensajero/genética
19.
Circulation ; 110(3): 349-55, 2004 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-15238461

RESUMEN

BACKGROUND: Several studies have suggested that stem cells are present in the stroma-vascular fraction (SVF) of adipose tissue (AT). METHODS AND RESULTS: To characterize the cell populations that compose the SVF of human AT originating from subcutaneous and visceral depots, fluorescence-activated cell sorter analysis was performed by use of fluorescent antibodies directed against the endothelial and stem cell markers CD31, CD34, CD133, and ABCG2. The freshly harvested SVF contained large numbers of CD34+ cells as well as cells expressing CD133 and ABCG2. Further analysis of the CD34+ cells revealed 2 CD34+ cell populations with differential expression of the endothelial cell marker CD31. Selection of the CD34+/CD31- cells by use of magnetic microbeads, followed by cell culture, demonstrated that this cell population could differentiate under appropriate conditions into endothelial cells. Moreover, in mouse ischemic hindlimb, intravenous injection of CD34(+)/CD31(-) cells was associated with an increase in the blood flow and the capillary density and an incorporation of the cells in the leg vasculature. CONCLUSIONS: Our data indicate the presence of a cell population within the SVF of human AT characterized as CD34+/CD31- exhibiting characteristics of endothelial progenitor cells. Therefore, human AT might represent a source of stem/progenitor cells useful for cell therapy to improve vasculogenesis in adults.


Asunto(s)
Tejido Adiposo/citología , Endotelio Vascular/citología , Isquemia/terapia , Células Madre/citología , Tejido Adiposo/irrigación sanguínea , Animales , Antígenos CD34/análisis , Biomarcadores/análisis , Diferenciación Celular , Proliferación Celular , Miembro Posterior/irrigación sanguínea , Humanos , Ratones , Ratones Desnudos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/análisis , Flujo Sanguíneo Regional , Células Madre/clasificación , Células Madre/metabolismo , Células del Estroma/metabolismo
20.
Int J Obes Relat Metab Disord ; 27(10): 1187-95, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14513066

RESUMEN

OBJECTIVE: Adipocyte hypertrophy combined with hyperplasia, observed during the growth of adipose tissue in obesity, might promote the occurrence of hypoxic areas within the tissue. The aim of the present study is to assess the influence of hypoxia on the expression and secretion of adipocyte-derived proangiogenic factors. DESIGN AND METHODS: Differentiated 3T3-F442A adipocytes were submitted either to ambient hypoxia (5% O(2)) or to chemically induced hypoxia by treatments with cobalt chloride or desferrioxamine. The activities of the matrix metalloproteinases 2 and 9 (MMP-2 and -9) were determined by gelatin zymography. The expression of vascular endothelial growth factor (VEGF), hypoxia inducible factor 1 alpha (HIF-1alpha), leptin, MMP-2 and -9 were studied by the use of Western blotting and RT-PCR analyses. RESULTS: Low oxygen pressure exposure and hypoxia mimics treatments were associated with increased glucose consumption and release of lactate in differentiated 3T3-F442A adipocytes. They also led to an upregulation of the expression of leptin, VEGF and MMPs. An enhanced accumulation of HIF-1alpha protein was observed in the hypoxic adipocyte nuclei. CONCLUSION: Hypoxia, in adipocytes, markedly enhances the expression of leptin, VEGF and MMPs and stimulates the HIF-1 pathway. The present data demonstrate that hypoxic adipocytes express more proangiogenic factors and suggest that hypoxia, if occurring in adipose tissue, might be a modulator of the angiogenic process.


Asunto(s)
Adipocitos/metabolismo , Proteínas Angiogénicas/metabolismo , Hipoxia de la Célula/fisiología , Células 3T3 , Animales , Western Blotting/métodos , Diferenciación Celular/fisiología , Glucosa/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia , Lactatos/metabolismo , Leptina/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Oxígeno/fisiología , ARN Mensajero/metabolismo , Factores de Transcripción/metabolismo , Regulación hacia Arriba/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA