Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
J Cell Physiol ; 230(12): 3076-83, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25989607

RESUMO

White adipose tissue (WAT) can be found in different locations in the body, and these different adipose deposits exhibit specific physiopathological importance according to the subcutaneous or abdominal locations. We have shown previously the presence of functional hematopoietic stem/progenitor cells (HSPC) in subcutaneous adipose tissue (SCAT). These cells exhibit a specific hematopoietic activity that contributes to the renewal of the immune cell compartment within this adipose deposit. In this study, we investigated whether HSPC can be found in visceral adipose tissue (VAT) and whether a putative difference in in situ hematopoiesis may be related to anatomical location and to site-specific immune cell content in VAT compared to SCAT. Therein, we identified for the first time the presence of HSPC in VAT. Using both in vitro assays and in vivo competitive repopulation experiments with sorted HSPC from VAT or SCAT, we showed that the hematopoietic activity of HSPC was lower in VAT, compared to SCAT. In addition, this altered hematopoietic activity of HSPC in VAT was due to their microenvironment, and may be related to a specific combination of secreted factors and extracellular matrix molecules expressed by adipose derived stromal cells. Our results indicate that WAT specific hematopoietic activity may be generalized to all adipose deposits, although with specificity according to the fat pad location. Considering the abundance of WAT in the body, this emphasizes the potential importance of this hematopoietic activity in physiopathological situations.


Assuntos
Hematopoese Extramedular , Células-Tronco Hematopoéticas/fisiologia , Gordura Intra-Abdominal/fisiologia , Gordura Subcutânea/fisiologia , Animais , Comunicação Celular , Linhagem da Célula , Proliferação de Células , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/imunologia , Gordura Intra-Abdominal/citologia , Gordura Intra-Abdominal/imunologia , Masculino , Camundongos Endogâmicos C57BL , Fenótipo , Nicho de Células-Tronco , Gordura Subcutânea/citologia , Gordura Subcutânea/imunologia
2.
Stem Cells ; 31(7): 1309-20, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23533182

RESUMO

Adipose tissue (AT) has become accepted as a source of multipotent progenitor cells, the adipose stromal cells (ASCs). In this regard, considerable work has been performed to harvest and characterize this cell population as well as to investigate the mechanisms by which transplanted ASCs mediate tissue regeneration. In contrast the endogenous release of native ASCs by AT has been poorly investigated. In this work, we show that native ASCs egress from murine AT. Indeed, we demonstrated that the release of native ASCs from AT can be evidenced both using an ex vivo perfusion model that we set up and in vivo. Such a mobilization process is controlled by CXCR4 chemokine receptor. In addition, once mobilized from AT, circulating ASCs were found to navigate through lymph fluid and to home into lymph nodes (LN). Therefore, we demonstrated that, during the LN activation, the fat depot encapsulating the activated LN releases native ASCs, which in turn invade the activated LN. Moreover, the ASCs invading the LN were visualized in close physical interaction with podoplanin and ER-TR7 positive structures corresponding to the stromal network composing the LN. This dynamic was impaired with CXCR4 neutralizing antibody. Taken together, these data provide robust evidences that native ASCs can traffic in vivo and that AT might provide stromal cells to activated LNs.


Assuntos
Tecido Adiposo/citologia , Linfonodos/citologia , Células-Tronco Mesenquimais/citologia , Tecido Adiposo/metabolismo , Animais , Diferenciação Celular/fisiologia , Processos de Crescimento Celular/fisiologia , Quimiocina CXCL12/metabolismo , Imunofenotipagem , Linfonodos/metabolismo , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Receptores CXCR4/metabolismo
3.
Nat Commun ; 15(1): 7940, 2024 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-39266552

RESUMO

Dedifferentiated and Well-differentiated liposarcoma are characterized by a systematic amplification of the Murine Double Minute 2 (MDM2) oncogene. We demonstrate that p53-independent metabolic functions of chromatin-bound MDM2 are exacerbated in liposarcoma and mediate an addiction to serine metabolism to sustain tumor growth. However, the origin of exogenous serine remains unclear. Here, we show that elevated serine levels in mice harboring liposarcoma-patient derived xenograft, released by distant muscle is essential for liposarcoma cell survival. Repressing interleukine-6 expression, or treating liposarcoma cells with Food and Drugs Administration (FDA) approved anti-interleukine-6 monoclonal antibody, decreases de novo serine synthesis in muscle, impairs proliferation, and increases cell death in vitro and in vivo. This work reveals a metabolic crosstalk between muscle and liposarcoma tumor and identifies anti-interleukine-6 as a plausible treatment for liposarcoma patients.


Assuntos
Proliferação de Células , Lipossarcoma , Proteínas Proto-Oncogênicas c-mdm2 , Serina , Lipossarcoma/metabolismo , Lipossarcoma/patologia , Lipossarcoma/genética , Animais , Humanos , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/genética , Camundongos , Linhagem Celular Tumoral , Serina/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Feminino , Masculino
4.
Med Sci (Paris) ; 39 Hors série n° 1: 15-21, 2023 Nov.
Artigo em Francês | MEDLINE | ID: mdl-37975765

RESUMO

Fibro-adipogenic progenitors (FAPs) are resident mesenchymal stromal cells (MSCs) of skeletal muscle. They play a crucial role in muscle homeostasis and regeneration through their paracrine activity. Recent technological advances in single-cell RNA sequencing have allowed the characterization of the heterogeneity within this cell population. In this article, we will present the different subpopulations of FAPs under basal, injury, or degenerative conditions, as well as their associated functions in mice and humans. We will then discuss the potential extramuscular origin of a post-injury FAP population. Indeed, our recent work demonstrates that MSCs from adipose tissue, infiltrating the muscle, could contribute to FAP heterogeneity.


Title: Et si l'origine des progéniteurs fibro-adipeux contribuait à leur hétérogénéité dans le muscle ? Abstract: Les progéniteurs fibro-adipogéniques (FAPs), cellules stromales mésenchymateuses (CSMs) résidentes du muscle squelettique, jouent un rôle crucial dans l'homéostasie et la régénération musculaire via leur activité paracrine. Les avancées technologiques récentes dans le domaine du séquençage de l'ARN en cellule unique ont permis la description de l'hétérogénéité de cette population cellulaire. Dans cet article, nous présenterons les différentes sous-populations de FAPs en condition basale, lésionnelle ou de dégénérescence, ainsi que leurs fonctions associées chez la souris et l'homme. Nous discuterons ensuite de l'origine extra-musculaire possible d'une population de FAPs post-lésionnelle. En effet, nos travaux récents démontrent que des CSMs provenant du tissu adipeux et infiltrées dans le muscle pourraient participer à l'hétérogénéité des FAPs.


Assuntos
Adipogenia , Células-Tronco Mesenquimais , Humanos , Camundongos , Animais , Músculo Esquelético , Tecido Adiposo , Diferenciação Celular/fisiologia
5.
Nat Commun ; 14(1): 80, 2023 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-36604419

RESUMO

Fibro-adipogenic progenitors (FAPs) play a crucial role in skeletal muscle regeneration, as they generate a favorable niche that allows satellite cells to perform efficient muscle regeneration. After muscle injury, FAP content increases rapidly within the injured muscle, the origin of which has been attributed to their proliferation within the muscle itself. However, recent single-cell RNAseq approaches have revealed phenotype and functional heterogeneity in FAPs, raising the question of how this differentiation of regenerative subtypes occurs. Here we report that FAP-like cells residing in subcutaneous adipose tissue (ScAT), the adipose stromal cells (ASCs), are rapidly released from ScAT in response to muscle injury. Additionally, we find that released ASCs infiltrate the damaged muscle, via a platelet-dependent mechanism and thus contribute to the FAP heterogeneity. Moreover, we show that either blocking ASCs infiltration or removing ASCs tissue source impair muscle regeneration. Collectively, our data reveal that ScAT is an unsuspected physiological reservoir of regenerative cells that support skeletal muscle regeneration, underlining a beneficial relationship between muscle and fat.


Assuntos
Músculo Esquelético , Doenças Musculares , Humanos , Tecido Adiposo , Diferenciação Celular/genética , Adipogenia/genética
6.
Stem Cells ; 29(11): 1837-48, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21922607

RESUMO

Although adipocyte terminal differentiation has been extensively studied, the early steps of adipocyte development and the embryonic origin of this lineage remain largely unknown. Here we describe a novel role for the pre-B-cell leukemia transcription factor one (PBX1) in adipocyte development using both mouse embryonic stem cells (mESCs) and human multipotent adipose-derived stem (hMADS) cells. We show that Pbx1(-/-) mESCs are unable to generate adipocytes, despite normal expression of neuroectoderm and neural crest (NC) markers. Early adipocyte lineage markers are not induced in Pbx1(-/-) mESCs, suggesting that Pbx1 controls the generation and/or the maintenance of adipocyte progenitors (APs) from the NC. We further characterize the function of PBX1 in postnatal adipogenesis and show that silencing of PBX1 expression in hMADS cells reduces their proliferation by preventing their entry in the S phase of the cell cycle. Furthermore, it promotes differentiation of hMADS cells into adipocytes and partially substitutes for glucocorticoids and rosiglitazone, two key proadipogenic agents. These effects involve direct modulation of PPARγ activity, most likely through regulation of the biosynthesis of PPARγ natural endogenous ligand(s). Together, our data suggest that PBX1 regulates adipocyte development at multiple levels, promoting the generation of NC-derived APs during embryogenesis, while favoring APs proliferation and preventing their commitment to the adipocyte lineage in postnatal life.


Assuntos
Adipócitos/citologia , Adipócitos/metabolismo , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição/metabolismo , Animais , Western Blotting , Ciclo Celular/genética , Ciclo Celular/fisiologia , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Proliferação de Células , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Proteínas de Homeodomínio/genética , Camundongos , Fator de Transcrição 1 de Leucemia de Células Pré-B , RNA Interferente Pequeno , Reação em Cadeia da Polimerase em Tempo Real , Fatores de Transcrição/genética
7.
Cells ; 11(6)2022 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-35326442

RESUMO

The epicardial adipose tissue (EAT) is the visceral fat depot of the heart which is highly plastic and in direct contact with myocardium and coronary arteries. Because of its singular proximity with the myocardium, the adipokines and pro-inflammatory molecules secreted by this tissue may directly affect the metabolism of the heart and coronary arteries. Its accumulation, measured by recent new non-invasive imaging modalities, has been prospectively associated with the onset and progression of coronary artery disease (CAD) and atrial fibrillation in humans. Recent studies have shown that EAT exhibits beige fat-like features, and express uncoupling protein 1 (UCP-1) at both mRNA and protein levels. However, this thermogenic potential could be lost with age, obesity and CAD. Here we provide an overview of the physiological and pathophysiological relevance of EAT and further discuss whether its thermogenic properties may serve as a target for obesity therapeutic management with a specific focus on the role of immune cells in this beiging phenomenon.


Assuntos
Tecido Adiposo , Doença da Artéria Coronariana , Adipocinas/metabolismo , Tecido Adiposo/metabolismo , Doença da Artéria Coronariana/metabolismo , Humanos , Obesidade/metabolismo , Pericárdio/metabolismo
8.
Diagn Interv Imaging ; 103(7-8): 353-359, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35292217

RESUMO

PURPOSE: The purpose of this study was to assess the capabilities of a deep learning (DL) tool to discriminate between type 1 facioscapulo-humeral dystrophy (FSHD1) and myositis using whole-body muscle magnetic resonance imaging (MRI) examination without the need for visual grading of muscle signal changes. MATERIALS AND METHODS: A total of 40 patients who underwent whole-body MRI examination that included T1-weighted and STIR sequences were included. There were 19 patients with proven FSHD1 (9 men, 10 women; mean age, 47.7 ± 18.0 [SD] years; age range: 20-72 years) and 21 patients with myositis fulfilling European Neuromuscular Centre criteria and European League Against Rheumatism and American College of Rheumatology criteria (11 men, 10 women; mean age, 59.3 ± 17.0 [SD]; age range: 19-78 years). Based on thigh, calf, and shoulder sections a supervised training of a neural network was performed and its diagnostic performance was studied using a 5-fold cross validation method and compared to the results obtained by two radiologists specialized in musculoskeletal imaging. RESULTS: The DL tool was able to differentiate FSHD1 from myositis with a correct classification percentage respectively of 69 % (95% CI: 39-99), 75% (95% CI: 48-100) and 77% (95% CI: 60-94) when thigh only, thigh and calf or the thigh, calf, and shoulder MR images were analyzed. The percentages of correct classification of the two radiologists for these later MR images were 38/40 (95%) and 35/40 (87.5%), respectively; with no differences with DL tool correct classification (P = 0.41 and P > 0.99, respectively). Among the seven patients who were misclassified by the radiologists, the DL tool correctly classified six of them. CONCLUSION: A DL tool was developed to discriminate between FSHD1 and myositis using whole-body MRI with performances equivalent to those achieved by two radiologists. This study provides a proof of concept of the effectiveness of a DL approach to distinguish between two myopathies using MRI with a small amount of data, and no prior muscle signal changes grading.


Assuntos
Aprendizado Profundo , Distrofia Muscular Facioescapuloumeral , Miosite , Adulto , Idoso , Feminino , Humanos , Imageamento por Ressonância Magnética/métodos , Masculino , Pessoa de Meia-Idade , Músculo Esquelético/diagnóstico por imagem , Distrofia Muscular Facioescapuloumeral/diagnóstico por imagem , Distrofia Muscular Facioescapuloumeral/patologia , Miosite/diagnóstico por imagem , Miosite/patologia , Adulto Jovem
9.
Arterioscler Thromb Vasc Biol ; 29(10): 1608-14, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19644053

RESUMO

OBJECTIVE: Adipose tissue (AT) plays a major role in the low-grade inflammatory state associated with obesity. The aim of the present study was to characterize the human AT lymphocytes (ATLs) and to analyze their interactions with adipocytes. METHODS AND RESULTS: Human ATL subsets were characterized by flow cytometry in subcutaneous ATs from 92 individuals with body mass index (BMI) ranging from 19 to 43 kg/m(2) and in paired biopsies of subcutaneous and visceral AT from 45 class II/III obese patients. CD3(+) ATLs were composed of effector and memory CD4(+) helper and CD8(+) cytotoxic T cells. The number of ATLs correlated positively with BMI and was higher in visceral than subcutaneous AT. Mature adipocytes stimulated the migration of ATLs and released the chemokine CCL20, the receptor of which (CCR6) was expressed in ATLs. The expression of adipocyte CCL20 was positively correlated with BMI and increased in visceral compared to subcutaneous adipocytes. ATLs expressed inflammatory markers and released interferon gamma (IFN gamma). Progenitor and adipocyte treatment with ATL-conditioned media reduced the insulin-mediated upregulation of lipogenic enzymes, an effect involving IFN gamma. CONCLUSIONS: Therefore, crosstalk occurs between adipocytes and lymphocytes within human AT involving T cell chemoattraction by adipocytes and modulation of lipogenesis by ATLs.


Assuntos
Adipócitos/imunologia , Quimiocina CCL20/fisiologia , Lipogênese , Obesidade/imunologia , Linfócitos T/fisiologia , Adiposidade , Adulto , Índice de Massa Corporal , Complexo CD3/análise , Quimiocina CCL20/análise , Feminino , Citometria de Fluxo , Humanos , Imunofenotipagem , Interferon gama/fisiologia , Pessoa de Meia-Idade , Gordura Subcutânea/imunologia
10.
Front Cell Dev Biol ; 8: 598520, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33490065

RESUMO

To coordinate specialized organs, inter-tissue communication appeared during evolution. Consequently, individual organs communicate their states via a vast interorgan communication network (ICN) made up of peptides, proteins, and metabolites that act between organs to coordinate cellular processes under homeostasis and stress. However, the nature of the interorgan signaling could be even more complex and involve mobilization mechanisms of unconventional cells that are still poorly described. Mesenchymal stem/stromal cells (MSCs) virtually reside in all tissues, though the biggest reservoir discovered so far is adipose tissue where they are named adipose stromal cells (ASCs). MSCs are thought to participate in tissue maintenance and repair since the administration of exogenous MSCs is well known to exert beneficial effects under several pathological conditions. However, the role of endogenous MSCs is barely understood. Though largely debated, the presence of circulating endogenous MSCs has been reported in multiple pathophysiological conditions, but the significance of such cell circulation is not known and therapeutically untapped. In this review, we discuss current knowledge on the circulation of native MSCs, and we highlight recent findings describing MSCs as putative key components of the ICN.

11.
Trends Endocrinol Metab ; 19(4): 130-7, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18337116

RESUMO

Human fat cell lipolysis was, until recently, thought to be mediated exclusively by a cAMP-dependent protein kinase (PKA)-regulated pathway under the control of catecholamines and insulin. We have shown that atrial- and B-type natriuretic peptides (ANP and BNP respectively) stimulate lipolysis in human fat cells through a cGMP-dependent protein kinase (PKG) signaling pathway independent of cAMP production and PKA activity. Pharmacological or physiological (exercise) increases in plasma ANP levels stimulate lipid mobilization in humans. This pathway becomes important during chronic treatment with beta-adrenoceptor antagonists, which inhibit catecholamine-induced lipolysis but enhance cardiac ANP release. These findings have metabolic implications and point to potential problems when natriuretic peptide secretion is altered or during therapeutic use of recombinant BNP.


Assuntos
GMP Cíclico/farmacologia , Lipólise/efeitos dos fármacos , Peptídeos Natriuréticos/farmacologia , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Animais , Biomarcadores/análise , Humanos , Lipólise/fisiologia , Modelos Biológicos
12.
Cell Rep ; 27(2): 323-333.e5, 2019 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-30970240

RESUMO

Ectopic lipid deposition (ELD) is defined by excess fat storage in locations not classically associated with adipose tissue (AT) storage. ELD is positively correlated with insulin resistance and increased risk of metabolic disorders. ELD appears as lipid droplets or adipocytes, whose cell origin is unknown. We previously showed that subcutaneous AT (ScAT) releases adipocyte progenitors into the circulation. Here, we demonstrate that triggering or preventing the release of adipocyte precursors from ScAT directly promoted or limited ectopic adipocyte formation in skeletal muscle in mice. Importantly, obesity-associated metabolic disorders could be mimicked by causing adipocyte precursor release without a high-fat diet. Finally, during nutrient overload, adipocyte progenitors exited ScAT, where their retention signals (CXCR4/CXCL12 axis) were greatly decreased, and further infiltrated skeletal muscles. These data provide insights into the formation of ELD associated with calorie overload and highlight adipocyte progenitor trafficking as a potential target in the treatment of metabolic diseases.


Assuntos
Gordura Subcutânea/metabolismo , Animais , Humanos , Absorção Intramuscular , Camundongos , Células Estromais/metabolismo
13.
J Clin Endocrinol Metab ; 93(10): 4098-106, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18682517

RESUMO

CONTEXT: Adipocyte formation in human adult adipose tissue (hAT) originates from resident progenitor cell differentiation in the stroma vascular fraction of the AT. The processes involved in the self-renewal of this cell population remain to be defined. OBJECTIVE: The objective was to study in situ and in vitro hAT progenitor cell (defined as CD34(+)/CD31(-) cells) proliferation. DESIGN AND PARTICIPANTS: In situ progenitor cell proliferation was assessed by immunohistochemistry and flow cytometry analyses on hAT from lean to obese subjects using the proliferation marker Ki-67. The effects of adipokines, hypoxia, and conditioned media (CM) from adipocytes, capillary endothelial cells, and macrophages isolated by an immunoselection approach were studied on hAT progenitor cell growth. Cell death in hAT was assessed by the terminal deoxynucleotidyl transferase-mediated dUTP-fluorescein end labeling method. RESULTS: Ki-67-positive staining was observed in AT progenitor cells. Fat mass enlargement in obese patients was associated with an increased Ki-67(+) progenitor cell population together with a new fraction of small adipocytes and increased cell death. HIF-1alpha mRNA expression in freshly harvested progenitor cells was positively correlated with body mass index. Adipocyte- and capillary endothelial cell-CM, hypoxia, leptin, IL-6, lysophosphatidic acid, and vascular endothelial growth factor, all increased hAT progenitor cell proliferation in vitro. Macrophage-CM had an antiproliferative effect that was suppressed by an antioxidant. CONCLUSIONS: The fraction of proliferative progenitor cells in adult hAT is modulated by the degree of adiposity. Changes in the progenitor cell microenvironment involving adipokines, hypoxia, and oxidative stress might play a key role in the control of the self-renewal of the local pool of AT progenitor cells.


Assuntos
Tecido Adiposo/fisiologia , Proliferação de Células , Líquido Extracelular/fisiologia , Desenvolvimento Humano/fisiologia , Células-Tronco/fisiologia , Adipocinas/farmacologia , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Adulto , Antígenos CD34/genética , Antígenos CD34/metabolismo , Morte Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Proliferação de Células/efeitos dos fármacos , Tamanho Celular/efeitos dos fármacos , Células Cultivadas , Meios de Cultivo Condicionados/farmacologia , Feminino , Humanos , Obesidade/genética , Obesidade/metabolismo , Obesidade/patologia , Oxigênio/farmacologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/genética , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo
14.
Stem Cells ; 25(9): 2269-76, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17525234

RESUMO

The native CD34+/CD31- cell population present in the stroma-vascular fraction of human adipose tissue (hAT) displays progenitor cell properties since they exhibit adipocyte- and endothelial cell-like phenotypes under appropriate stimuli. To analyze the signals within hAT regulating their phenotypes, the influence of hAT-derived capillary endothelial cells (CECs) was studied on the chemotaxis and differentiation of the hAT-CD34+/CD31- cells. Conditioned medium from hAT-CECs led to a strong chemotaxis of the hAT-CD34+/CD31- cells that was inhibited with pretreatments with pertussis toxin, CXCR-4 antagonist, or neutralizing antibodies. Furthermore, hAT-CECs produced and secreted the CXCR-4 ligand, that is, the stromal derived factor-1 (SDF-1). Finally, hAT-CECs induced the differentiation of hAT-CD34+/CD31- cells toward an endothelial cell (EC) phenotype. Indeed, hAT-CECs and -CD34+/CD31- cell coculture stimulated in a two-dimensional system the expression of the EC CD31 marker by the hAT-progenitor cells and, in a three-dimensional approach, the formation of capillary-like structures via a SDF-1/CXCR-4 dependent pathway. Thus, the migration and differentiation of hAT progenitor cells are modulated by hAT-CEC-derived factors. SDF-1, which is secreted by hAT-derived CECs, and its receptor CXCR-4, expressed by hAT-derived progenitor cells, may promote chemotaxis and differentiation of hAT-derived progenitor cells and thus contribute to the formation of the vascular network during the development of hAT.


Assuntos
Tecido Adiposo/citologia , Células-Tronco Adultas/citologia , Antígenos CD34/metabolismo , Diferenciação Celular , Quimiocina CXCL12/fisiologia , Quimiotaxia/fisiologia , Células Endoteliais/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Tecido Adiposo/metabolismo , Células-Tronco Adultas/metabolismo , Células Cultivadas , Quimiocina CXCL12/metabolismo , Humanos , Receptores CXCR4/metabolismo , Receptores CXCR4/fisiologia
15.
Mol Metab ; 13: 24-29, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29789270

RESUMO

OBJECTIVE: Surplus dietary fat cannot be converted into other macronutrient forms or excreted, so has to be stored or oxidized. Healthy mammals store excess energy in the form of triacylgycerol (TAG) in lipid droplets within adipocytes rather than oxidizing it, and thus ultimately gain weight. The 'overflow hypothesis' posits that the capacity to increase the size and number of adipocytes is finite and that when this limit is exceeded, fat accumulates in ectopic sites and leads to metabolic disease. METHODS: Here we studied the energetic and biochemical consequences of short-term (2-day) excess fat ingestion in a lipodystrophic (A-ZIP/F-1) mouse model in which adipose capacity is severely restricted. RESULTS: In wildtype littermates, this acute exposure to high fat diets resulted in excess energy intake and weight gain without any significant changes in macronutrient oxidation rates, glucose, TAG, or insulin levels. In contrast, hyperphagic lipodystrophic mice failed to gain weight; rather, they significantly increased hepatic steatosis and fat oxidation. This response was associated with a significant increase in hyperglycemia, hyperinsulinemia, glucosuria, hypertriglyceridemia, and worsening insulin tolerance. CONCLUSIONS: These data suggest that when adipose storage reserves are saturated, excess fat intake necessarily increases fat oxidation and induces oxidative substrate competition which exacerbates insulin resistance resolving any residual energy surplus through excretion of glucose.


Assuntos
Gorduras na Dieta/metabolismo , Lipídeos/fisiologia , Lipodistrofia/metabolismo , Adipócitos , Tecido Adiposo , Adiposidade , Animais , Dieta Hiperlipídica , Ingestão de Energia , Metabolismo Energético , Fígado Gorduroso , Glucose , Hiperinsulinismo , Resistência à Insulina/fisiologia , Lipodistrofia/fisiopatologia , Fígado , Camundongos , Obesidade , Oxirredução , Aumento de Peso
16.
J Endocrinol ; 236(3): 137-150, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29339381

RESUMO

The incorporation of plasma triglyceride (TG) fatty acids to white adipose tissue (WAT) depends on lipoprotein lipase (LPL), which is regulated by angiopoietin-like protein-4 (ANGPTL-4), an unfolding molecular chaperone that converts active LPL dimers into inactive monomers. The production of ANGPTL-4 is promoted by fasting and repressed by feeding. We hypothesized that the postprandial hormone cholecystokinin (CCK) facilitates the storage of dietary TG fatty acids in WAT by regulating the activity of the LPL/ANGPTL-4 axis and that it does so by acting directly on CCK receptors in adipocytes. We report that administration of CCK-8 (a bioactive fragment of CCK) to rats: (i) reduces plasma ANGTPL-4 levels; (ii) represses Angptl-4 expression in WAT and (iii) simultaneously enhances LPL activity in this tissue without inducing Lpl expression. In vivo CCK-8 effects are specifically antagonized by the CCK-2 receptor (CCK-2R) antagonist, L-365,260. Moreover, CCK-8 downregulates Angptl-4 expression in wild-type pre-adipocytes, an effect that is not observed in engineered pre-adipocytes lacking CCK-2R. These effects have functional consequences as CCK-8 was found to promote the uptake of dietary fatty acids by WAT, as demonstrated by means of proton nuclear magnetic resonance (1H-NMR). The efficacy of acute CCK-8 administration was not reduced after chronic CCK-8 treatment. Moreover, the effects of CCK-8 on WAT were not associated to the increase of circulating insulin. Our results show that cholecystokinin promotes lipid storage in WAT by acting on adipocyte CCK-2R, suggesting a pivotal role for CCK in TG homeostasis.


Assuntos
Tecido Adiposo Branco/metabolismo , Colecistocinina/fisiologia , Ácidos Graxos/metabolismo , Triglicerídeos/metabolismo , Proteína 4 Semelhante a Angiopoietina/antagonistas & inibidores , Proteína 4 Semelhante a Angiopoietina/sangue , Proteína 4 Semelhante a Angiopoietina/genética , Animais , Gorduras na Dieta/metabolismo , Expressão Gênica , Lipase Lipoproteica/genética , Lipase Lipoproteica/metabolismo , Masculino , Período Pós-Prandial , Ratos , Ratos Sprague-Dawley , Receptor de Colecistocinina B/efeitos dos fármacos , Receptor de Colecistocinina B/fisiologia , Sincalida/administração & dosagem , Sincalida/farmacologia
17.
Compr Physiol ; 7(3): 1051-1082, 2017 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-28640452

RESUMO

Epicardial adipose tissue (EAT) is a small but very biologically active ectopic fat depot that surrounds the heart. Given its rapid metabolism, thermogenic capacity, unique transcriptome, secretory profile, and simply measurability, epicardial fat has drawn increasing attention among researchers attempting to elucidate its putative role in health and cardiovascular diseases. The cellular crosstalk between epicardial adipocytes and cells of the vascular wall or myocytes is high and suggests a local role for this tissue. The balance between protective and proinflammatory/profibrotic cytokines, chemokines, and adipokines released by EAT seem to be a key element in atherogenesis and could represent a future therapeutic target. EAT amount has been found to predict clinical coronary outcomes. EAT can also modulate cardiac structure and function. Its amount has been associated with atrial fibrillation, coronary artery disease, and sleep apnea syndrome. Conversely, a beiging fat profile of EAT has been identified. In this review, we describe the current state of knowledge regarding the anatomy, physiology and pathophysiological role of EAT, and the factors more globally leading to ectopic fat development. We will also highlight the most recent findings on the origin of this ectopic tissue, and its association with cardiac diseases. © 2017 American Physiological Society. Compr Physiol 7:1051-1082, 2017.


Assuntos
Tecido Adiposo/metabolismo , Doenças Cardiovasculares/etiologia , Metabolismo dos Lipídeos , Obesidade/etiologia , Pericárdio/metabolismo , Adipocinas/genética , Adipocinas/metabolismo , Tecido Adiposo/diagnóstico por imagem , Animais , Humanos , Pericárdio/citologia , Pericárdio/diagnóstico por imagem
18.
Arterioscler Thromb Vasc Biol ; 25(10): 2032-42, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16123323

RESUMO

In normal and obese humans, lipid mobilization and systemic nonesterified fatty acid levels are thought to be acutely controlled by catecholamines (ie, epinephrine and norepinephrine) and insulin. Natriuretic peptides (NPs) are known to play a key role in the regulation of salt and water balance and blood pressure homeostasis. They are involved in the pathophysiology of hypertension and heart failure. NPs have recently been found to exert potent lipolytic effects (ie, activating the breakdown of stored triacylglycerols) in isolated human fat cells and to promote lipid mobilization in vivo. Atrial natriuretic peptide increases the intracellular 3', 5'-cyclic guanosine monophosphate (cGMP) concentration which activates cGMP-dependent protein kinase leading to perilipin and hormone-sensitive lipase phosphorylation and lipolysis. NPs promote lipid mobilization when administered intravenously. NPs are also responsible for the residual lipid-mobilizing action observed under oral beta-blockade in subjects performing physical exercise. NPs are therefore novel factors which may open promising research pathways to explain the control of lipid mobilization in physiological and pathological conditions. The metabolic impact of altered production and circulation of NPs remains to be established. The potential influence of NPs on the development of lipid disorders, obesity-related cardiovascular events, and cardiac cachexia will be discussed in this review.


Assuntos
Fator Natriurético Atrial/metabolismo , Ácidos Graxos não Esterificados/metabolismo , Metabolismo dos Lipídeos/fisiologia , Lipólise/fisiologia , Síndrome Metabólica/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Obesidade/metabolismo
19.
Diabetes ; 53(5): 1285-92, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15111498

RESUMO

Obesity has been suggested to be a low-grade systemic inflammatory state, therefore we studied the interaction between human adipocytes and monocytes via adipose tissue (AT)-derived capillary endothelium. Cells composing the stroma-vascular fraction (SVF) of human ATs were characterized by fluorescence-activated cell sorter (FACS) analysis and two cell subsets (resident macrophages and endothelial cells [ECs]) were isolated using antibody-coupled microbeads. Media conditioned by mature adipocytes maintained in fibrin gels were applied to AT-derived ECs. Thereafter, the expression of endothelial adhesion molecules was analyzed as well as the adhesion and transmigration of human monocytes. FACS analysis showed that 11% of the SVF is composed of CD14(+)/CD31(+) cells, characterized as resident macrophages. A positive correlation was found between the BMI and the percentage of resident macrophages, suggesting that fat tissue growth is associated with a recruitment of blood monocytes. Incubation of AT-derived ECs with adipocyte-conditioned medium resulted in the upregulation of EC adhesion molecules and the increased chemotaxis of blood monocytes, an effect mimicked by recombinant human leptin. These results indicate that adipokines, such as leptin, activate ECs, leading to an enhanced diapedesis of blood monocytes, and suggesting that fat mass growth might be linked to inflammatory processes.


Assuntos
Adipócitos/fisiologia , Tecido Adiposo/citologia , Macrófagos/citologia , Monócitos/fisiologia , Adipócitos/metabolismo , Tecido Adiposo/irrigação sanguínea , Capilares , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Senescência Celular , Meios de Cultivo Condicionados/farmacologia , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/fisiologia , Humanos
20.
Diabetes ; 51(2): 293-300, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11812735

RESUMO

Hormone-sensitive lipase (HSL) catalyzes the rate-limiting step in the mobilization of fatty acids from adipose tissue, thus determining the supply of energy substrates in the body. HSL mRNA was positively regulated by glucose in human adipocytes. Pools of stably transfected 3T3-F442A adipocytes were generated with human adipocyte HSL promoter fragments from -2,400/+38 to -31/+38 bp linked to the luciferase gene. A glucose-responsive region was mapped within the proximal promoter (-137 bp). Electromobility shift assays showed that upstream stimulatory factor (USF)-1 and USF2 and Sp1 and Sp3 bound to a consensus E-box and two GC-boxes in the -137-bp region. Cotransfection of the -137/+38 construct with USF1 and USF2 expression vectors produced enhanced luciferase activity. Moreover, HSL mRNA levels were decreased in USF1- and USF2-deficient mice. Site-directed mutagenesis of the HSL promoter showed that the GC-boxes, although contributing to basal promoter activity, were dispensable for glucose responsiveness. Mutation of the E-box led to decreased promoter activity and suppression of the glucose response. Analogs and metabolites were used to determine the signal metabolite of the glucose response. The signal is generated downstream of glucose-6-phosphate in the glycolytic pathway before the triose phosphate step.


Assuntos
Adipócitos/metabolismo , Esterol Esterase/genética , Transcrição Gênica/fisiologia , Células 3T3 , Tecido Adiposo/fisiologia , Animais , Sequência de Bases/genética , Sítios de Ligação/genética , Proteínas Estimuladoras de Ligação a CCAAT/genética , Células Cultivadas , Proteínas de Ligação a DNA/genética , Espaço Extracelular/metabolismo , Feminino , Expressão Gênica , Glucose/metabolismo , Glucose/farmacologia , Humanos , Camundongos , Mutagênese Sítio-Dirigida , Mutação/fisiologia , Concentração Osmolar , Regiões Promotoras Genéticas/fisiologia , Esterol Esterase/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1 , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Transcrição Gênica/efeitos dos fármacos , Transfecção , Fatores Estimuladores Upstream
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA