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

Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Stem Cells ; 33(8): 2564-73, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25827082

RESUMO

Adipose tissue expansion is well-orchestrated to fulfill the energy demand. It results from adipocyte hypertrophy and hyperplasia due to adipose progenitor cell (APC) expansion and differentiation. Chronic low grade inflammation and hypoxia take place in obese adipose tissue microenvironment. Both of these events were shown to impact the APC pool by promoting increased self-renewal along with a decrease in the APC differentiation potential. However, no common target has been identified so far. Here we show that the immediate early response 3 gene (IER3) is preferentially expressed in APCs and is essential for APC proliferation and self-renewal. Experiments based on RNA interference revealed that impairing IER3 expression altered cell proliferation through ERK1/2 phosphorylation and clonogenicity. IER3 expression was induced by Activin A, which plays a crucial role in adipocyte differentiation as well as by a decrease in oxygen tension through HIF1-induced transcriptional activation. Interestingly, high levels of IER3 were detected in native APCs (CD34+/CD31- cells) isolated from obese patients and conditioned media from obese adipose tissue-macrophages stimulated its expression. Overall, these results indicate that IER3 is a key player in expanding the pool of APC while highlighting the role of distinct effectors found in an obese microenvironment in this process.


Assuntos
Tecido Adiposo/metabolismo , Proteínas Reguladoras de Apoptose/biossíntese , Regulação da Expressão Gênica/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas de Membrana/biossíntese , Nicho de Células-Tronco/fisiologia , Células-Tronco/metabolismo , Tecido Adiposo/citologia , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Células-Tronco/citologia
2.
Stem Cells ; 32(6): 1459-67, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24302443

RESUMO

Identification of molecular mechanisms involved in generation of different types of adipocytes is progressing substantially in mice. However, much less is known regarding characterization of brown (BAP) and white adipocyte progenitors (WAPs) in humans, highlighting the need for an in vitro model of human adipocyte development. Here, we report a procedure to selectively derive BAP and WAPs from human-induced pluripotent stem cells. Molecular characterization of APs of both phenotypes revealed that BMP4, Hox8, Hoxc9, and HoxA5 genes were specifically expressed in WAPs, whereas expression of PRDM16, Dio2, and Pax3 marked BAPs. We focused on Pax3 and we showed that expression of this transcription factor was enriched in human perirenal white adipose tissue samples expressing UCP1 and in human classical brown fat. Finally, functional experiments indicated that Pax3 was a critical player of human AP fate as its ectopic expression led to convert WAPs into brown-like APs. Together, these data support a model in which Pax3 is a new marker of human BAPs and a molecular mediator of their fate. The findings of this study could lead to new anti-obesity therapies based on the recruitment of APs and constitute a platform for investigating in vitro the developmental origins of human white and brown adipocytes.


Assuntos
Adipócitos Marrons/citologia , Adipócitos Brancos/citologia , Diferenciação Celular , Células-Tronco Pluripotentes Induzidas/citologia , Fatores de Transcrição Box Pareados/metabolismo , Adipócitos Marrons/efeitos dos fármacos , Adipócitos Marrons/metabolismo , Adipócitos Brancos/efeitos dos fármacos , Adipócitos Brancos/metabolismo , Adipogenia/efeitos dos fármacos , Idoso de 80 Anos ou mais , Animais , Diferenciação Celular/efeitos dos fármacos , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Camundongos , Fator de Transcrição PAX3 , Fenótipo , Tretinoína/farmacologia
3.
Biochem Biophys Res Commun ; 430(3): 871-5, 2013 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-23268339

RESUMO

Human adipose-derived stem cell populations express cell surface markers such as CD105, CD73, CD146 and CD140a/PDFGRα. However, it was unclear whether these markers could discriminate subpopulations of undifferentiated cells and whether the expression of these markers is modulated during differentiation. To address this issue, we analysed the immunophenotype of cultured human multipotent adipose derived stem (hMADS) cell populations at different adipocyte differentiation steps. We found that 100% of undifferentiated cells expressed CD73 and CD105. In contrast, CD146 and CD140a/PDFGRα marked two different subpopulations of cells. CD140a/PDGFRα subpopulation was regulated by FGF2, a critical factor of human adipose-derived stem cell self-renewal. During differentiation, CD73 was maintained and marked lipid-laden cells, whereas CD105 expression was inhibited in fully differentiated cells. The percentage of CD146 and CD140a/PDFGRα-positive cells declined as soon as cells had undergone differentiation. Altogether, these data support the notion that expanded adipose-derived stem cells are heterogeneous mixtures of cells and cell surface markers studied can discriminate subpopulations.


Assuntos
Adipócitos/citologia , Adipogenia/fisiologia , Tecido Adiposo/citologia , Membrana Celular/metabolismo , Células-Tronco/citologia , 5'-Nucleotidase/biossíntese , 5'-Nucleotidase/genética , Adipogenia/efeitos dos fármacos , Tecido Adiposo/efeitos dos fármacos , Antígenos CD/biossíntese , Antígenos CD/genética , Biomarcadores/metabolismo , Antígeno CD146/biossíntese , Antígeno CD146/genética , Linhagem Celular , Endoglina , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fator 2 de Crescimento de Fibroblastos/farmacologia , Citometria de Fluxo , Proteínas Ligadas por GPI/biossíntese , Proteínas Ligadas por GPI/genética , Humanos , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/biossíntese , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptores de Superfície Celular/biossíntese , Receptores de Superfície Celular/genética
4.
BMC Cell Biol ; 9: 11, 2008 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-18271953

RESUMO

BACKGROUND: Multipotent stem cells exist within adipose tissue throughout life. An abnormal recruitment of these adipose precursor cells could participate to hyperplasia of adipose tissue observed in severe obesity or to hypoplasia of adipose tissue observed in lipodystrophy. Therefore, pharmacological molecules that control the pool of stem cells in adipose tissue are of great interest. Glycogen Synthase Kinase (GSK) 3 has been previously described as involved in differentiation of preadipose cells and might be a potential therapeutic target to modulate proliferation and differentiation of adipocyte precursors. However, the impact of GSK3 inhibition on human adipose-derived stem cells remained to be investigated. The aim of this study was to investigate GSK3 as a possible target for pharmacological inhibition of stem cell adipogenesis. To reach this goal, we studied the effects of pharmacological inhibitors of GSK3, i.e. lithium chloride (LiCl) and BIO on proliferation and adipocyte differentiation of multipotent stem cells derived from human adipose tissue. RESULTS: Our results showed that GSK3 inhibitors inhibited proliferation and clonogenicity of human stem cells, strongly suggesting that GSK3 inhibitors could be potent regulators of the pool of adipocyte precursors in adipose tissue. The impact of GSK3 inhibition on differentiation of hMADS cells was also investigated. Adipogenic and osteogenic differentiations were inhibited upon hMADS treatment with BIO. Whereas a chronic treatment was required to inhibit osteogenesis, a treatment that was strictly restricted to the early step of differentiation was sufficient to inhibit adipogenesis. CONCLUSION: These results demonstrated the feasibility of a pharmacological approach to regulate adipose-derived stem cell function and that GSK3 could represent a potential target for controlling adipocyte precursor pool under conditions where fat tissue formation is impaired.


Assuntos
Adipócitos/citologia , Tecido Adiposo/citologia , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Células-Tronco/citologia , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Adipogenia/efeitos dos fármacos , Adipogenia/fisiologia , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Pré-Escolar , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Imuno-Histoquímica , Lactente , Cloreto de Lítio/farmacologia , Masculino , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo
5.
Plast Reconstr Surg ; 139(3): 685e-692e, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28234838

RESUMO

BACKGROUND: Autologous fat grafting has become an essential procedure in breast reconstructive surgery. However, molecular knowledge of different adipose donor sites remains inadequate. Tissue regeneration studies have shown that it is essential to match the Hox code of transplanted cells and host tissues to achieve correct repair. This study aims to provide a better molecular understanding of adipose tissue. METHODS: Over the course of 1 year, the authors prospectively included 15 patients and studied seven adipose areas: chin, breast, arm, abdomen, thigh, hip, and knee. The first step consisted of the surgical harvesting of adipose tissue. RNA was then extracted and converted into cDNA to study gene expression levels of 10 targeted genes by real-time polymerase chain reaction. RESULTS: Forty samples from Caucasian women with a mean age of 48 years were studied. The expression of PAX3, a marker of neuroectodermal origin, was significantly higher in the breast, with a decreasing gradient from the upper to lower areas of the body. An inverse gradient was found for the expression of HOXC10. This expression profile was statistically significant for the areas of the thigh and knee compared with the breast (p < 0.0083). CONCLUSIONS: Breast fat may have a specific embryologic origin compared with the knee and thigh. The reinjection of adipocytes from the infraumbilical area leads to the transfer of cells highly expressing HOXC10. This study raises questions about the safety of this procedure, and future studies will be required to examine molecular modifications of adipose cells transferred to a heterotopic location. CLINICAL QUESTION/LEVEL OF EVIDENCE: Therapeutic, V.


Assuntos
Tecido Adiposo , Mama/anatomia & histologia , Regulação da Expressão Gênica no Desenvolvimento , Genes Homeobox/fisiologia , Procedimentos de Cirurgia Plástica/métodos , Sítio Doador de Transplante , Feminino , Expressão Gênica , Humanos , Pessoa de Meia-Idade , Estudos Prospectivos
6.
Sci Rep ; 7(1): 2954, 2017 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-28592806

RESUMO

Autologous fat grafting is a gold standard therapy for soft tissue defects, but is hampered by unpredictable postoperative outcomes. Fat graft enrichment with adipose-derived stromal cell (ASCs) was recently reported to enhance graft survival. Platelet-rich plasma (PRP) has also emerged as a biologic scaffold that promotes fat graft viability. Combined ASC/PRP fat grafting enrichment is thus a promising new regenerative medicine approach. The effects of PRP on ASC proliferation are well documented, but the impact of PRP on ASC differentiation has yet to be investigated in depth to further elucidate the PRP clinical effects. Here we analyzed the human ASC fate upon PRP treatment. PRP was found to sharply reduce the potential of ASCs to undergo differentiation into adipocytes. Interestingly, the PRP anti-adipogenic effect was accompanied by the generation of myofibroblast-like cells. Among the various factors released from PRP, TGFß pathway activators played a critical role in both the anti-adipogenic and pro-myofibroblastic PRP effects. Overall, these data suggest that PRP participates in maintaining a pool of ASCs and in the repair process by promoting ASC differentiation into myofibroblast-like cells. TGFß may provide an important target pathway to improve PRP clinical outcomes.


Assuntos
Adipogenia , Tecido Adiposo/citologia , Diferenciação Celular , Miofibroblastos/citologia , Miofibroblastos/metabolismo , Plasma Rico em Plaquetas/metabolismo , Células Estromais/citologia , Células Estromais/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Adipócitos/citologia , Adipócitos/metabolismo , Idoso , Benzamidas/farmacologia , Células Cultivadas , Dioxóis/farmacologia , Feminino , Humanos , Lactente , Masculino , Fenótipo
7.
Methods Mol Biol ; 329: 341-51, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16846003

RESUMO

Adipocytes and osteoblasts are derived from a common precursor cell. It has been proposed that the bone loss commonly seen during aging or in the pathology of osteoporosis might be partly caused by a deregulation of the normal balance between osteoblast and adipocyte differentiation. In vitro differentiation of mouse embryonic stem cells toward the adipogenic and the osteogenic lineages provides a powerful system for testing effects of compounds on the developmental switch between adipogenesis and osteogenesis and identification of pharmacological targets. In this chapter, an improved protocol to commit mouse embryonic stem cells into both lineages at a correct rate is detailed.


Assuntos
Adipócitos/citologia , Diferenciação Celular , Embrião de Mamíferos/citologia , Osteoblastos/citologia , Células-Tronco Pluripotentes/citologia , Adipócitos/metabolismo , Adipogenia/genética , Animais , Sequência de Bases , Técnicas de Cultura de Células/métodos , Primers do DNA/genética , Avaliação Pré-Clínica de Medicamentos , Expressão Gênica , Camundongos , Osteoblastos/metabolismo , Osteogênese/genética , Células-Tronco Pluripotentes/metabolismo , RNA/genética , RNA/isolamento & purificação , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
Sci Rep ; 6: 32490, 2016 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-27577850

RESUMO

Human induced pluripotent stem cells (hiPSCs) show great promise for obesity treatment as they represent an unlimited source of brown/brite adipose progenitors (BAPs). However, hiPSC-BAPs display a low adipogenic capacity compared to adult-BAPs when maintained in a traditional adipogenic cocktail. The reasons of this feature are unknown and hamper their use both in cell-based therapy and basic research. Here we show that treatment with TGFß pathway inhibitor SB431542 together with ascorbic acid and EGF were required to promote hiPSCs-BAP differentiation at a level similar to adult-BAP differentiation. hiPSC-BAPs expressed the molecular identity of adult-UCP1 expressing cells (PAX3, CIDEA, DIO2) with both brown (ZIC1) and brite (CD137) adipocyte markers. Altogether, these data highlighted the critical role of TGFß pathway in switching off hiPSC-brown adipogenesis and revealed novel factors to unlock their differentiation. As hiPSC-BAPs display similarities with adult-BAPs, it opens new opportunities to develop alternative strategies to counteract obesity.


Assuntos
Adipócitos Marrons/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Transdução de Sinais/genética , Fator de Crescimento Transformador beta/antagonistas & inibidores , Adipócitos Marrons/citologia , Adipócitos Marrons/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Ácido Ascórbico/farmacologia , Benzamidas/farmacologia , Biomarcadores/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Dioxóis/farmacologia , Fator de Crescimento Epidérmico/farmacologia , Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Iodeto Peroxidase/genética , Iodeto Peroxidase/metabolismo , Fator de Transcrição PAX3/genética , Fator de Transcrição PAX3/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/genética , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo , Iodotironina Desiodinase Tipo II
9.
Stem Cells Int ; 2015: 592090, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25733979

RESUMO

Animal study findings have revealed that individual fat depots are not functionally equivalent and have different embryonic origins depending on the anatomic location. Mouse bone regeneration studies have also shown that it is essential to match the Hox code of transplanted cells and host tissues to achieve correct repair. However, subcutaneous fat depots from any donor site are often used in autologous fat grafting. Our study was thus carried out to determine the embryonic origins of human facial (chin) and limb (knee) fat depots and whether they had similar features and molecular matching patterns. Paired chin and knee fat depots were harvested from 11 subjects and gene expression profiles were determined by DNA microarray analyses. Adipose-derived stromal cells (ASCs) from both sites were isolated and analyzed for their capacity to proliferate, form clones, and differentiate. Chin and knee fat depots expressed a different HOX code and could have different embryonic origins. ASCs displayed a different phenotype, with chin-ASCs having the potential to differentiate into brown-like adipocytes, whereas knee-ASCs differentiated into white adipocytes. These results highlighted different features for these two fat sites and indicated that donor site selection might be an important factor to be considered when applying adipose tissue in cell-based therapies.

10.
Diabetes ; 63(10): 3253-65, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24789919

RESUMO

The presence of brown adipose tissue (BAT) in human adults opens attractive perspectives to treat metabolic disorders. Indeed, BAT dissipates energy as heat via uncoupling protein (UCP)1. Brown adipocytes are located in specific deposits or can emerge among white fat through the so-called browning process. Although numerous inducers have been shown to drive this process, no study has investigated whether it could be controlled by specific metabolites. Here, we show that lactate, an important metabolic intermediate, induces browning of murine white adipose cells with expression of functional UCP1. Lactate-induced browning also occurs in human cells and in vivo. Lactate controls Ucp1 expression independently of hypoxia-inducible factor-1α and PPARα pathways but requires active PPARγ signaling. We demonstrate that the lactate effect on Ucp1 is mediated by intracellular redox modifications as a result of lactate transport through monocarboxylate transporters. Further, the ketone body ß-hydroxybutyrate, another metabolite that impacts redox state, is also a strong browning inducer. Because this redox-dependent increase in Ucp1 expression promotes an oxidative phenotype with mitochondria, browning appears as an adaptive mechanism to alleviate redox pressure. Our findings open new perspectives for the control of adipose tissue browning and its physiological relevance.


Assuntos
Adipócitos Marrons/metabolismo , Adipócitos Brancos/metabolismo , Adipogenia/fisiologia , Animais , Metabolismo Energético/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Oxirredução , Consumo de Oxigênio/fisiologia , PPAR gama/metabolismo , Células-Tronco
11.
Acta Cir Bras ; 27(4): 283-9, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22534801

RESUMO

PURPOSE: To evaluate the implant of human adipose derived stem cells (ADSC) delivered in hyaluronic acid gel (HA), injected in the subcutaneous of athymic mice. METHODS: Control implants -HA plus culture media was injected in the subcutaneous of the left sub scapular area of 12 athymic mice. ADSC implants: HA plus ADSC suspended in culture media was injected in the subcutaneous, at the contra lateral area, of the same animals. With eight weeks, animals were sacrificed and the recovered implants were processed for extraction of genomic DNA, and histological study by hematoxilin-eosin staining and immunufluorescence using anti human vimentin and anti von Willebrand factor antibodies. CONTROLS: Not visualized at the injection site. An amorphous substance was observed in hematoxilin-eosin stained sections. Human vimentin and anti von Willebrand factor were not detected. No human DNA was detected. ADSC implants - A plug was visible at the site of injection. Fusiform cells were observed in sections stained by hematoxilin- eosin and both human vimentin and anti von Willebrand factor were detected by immunofluorescence. The presence of human DNA was confirmed. CONCLUSION: The delivery of human adipose derived stem cells in preparations of hyaluronic acid assured cells engraftment at the site of injection.


Assuntos
Adipócitos/transplante , Tecido Adiposo/citologia , Ácido Hialurônico/administração & dosagem , Transplante de Células-Tronco/métodos , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Adulto , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Feminino , Imunofluorescência , Humanos , Implantes Experimentais , Injeções Subcutâneas/métodos , Camundongos , Camundongos Nus , Modelos Animais , Engenharia Tecidual/métodos , Vimentina/análise , Fator de von Willebrand/análise , Fator de von Willebrand/antagonistas & inibidores
12.
Am J Stem Cells ; 1(1): 42-7, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23671796

RESUMO

Adipose tissue is an alternative source of mesenchymal stem cells and human adipose-derived stem cells (ASCs) display an attractive and substantial therapeutic potential when transplanted in animal models. To this end, an understanding of ASC biology is necessary and the knowledge of mechanisms that maintain ASCs in an undifferentiated state with no loss of differentiation potential during ex vivo expansion represents a crucial step. However, these mechanisms remain to be identified because appropriate human cellular models are scant. In this review we will describe a cellular model isolated from human adipose tissue displaying all the features of stem cells. Then, we will focus on the identification of intrinsic and extrinsic factors regulating the balance between human ASC proliferation and differentiation. We will point out the role of factors secreted by undifferentiated ASCs, such a FGF2, activin A, BMP4, Hedgehog molecules and secreted by adipose tissue macrophages. Finally, we will outline the role of miRNAs in these processes.

13.
PLoS One ; 7(2): e31274, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22355352

RESUMO

OBJECTIVE: The present study was undertaken to characterize the remodeling phenotype of human adipose tissue (AT) macrophages (ATM) and to analyze their paracrine effects on AT progenitor cells. RESEARCH DESIGN AND METHODS: The phenotype of ATM, immunoselected from subcutaneous (Sc) AT originating from subjects with wide range of body mass index and from paired biopsies of Sc and omental (Om) AT from obese subjects, was studied by gene expression analysis in the native and activated states. The paracrine effects of ScATM on the phenotype of human ScAT progenitor cells (CD34(+)CD31(-)) were investigated. RESULTS: Two main ATM phenotypes were distinguished based on gene expression profiles. For ScAT-derived ATM, obesity and adipocyte-derived factors favored a pro-fibrotic/remodeling phenotype whereas the OmAT location and hypoxic culture conditions favored a pro-angiogenic phenotype. Treatment of native human ScAT progenitor cells with ScATM-conditioned media induced the appearance of myofibroblast-like cells as shown by expression of both α-SMA and the transcription factor SNAIL, an effect mimicked by TGFß1 and activinA. Immunohistochemical analyses showed the presence of double positive α-SMA and CD34 cells in the stroma of human ScAT. Moreover, the mRNA levels of SNAIL and SLUG in ScAT progenitor cells were higher in obese compared with lean subjects. CONCLUSIONS: Human ATM exhibit distinct pro-angiogenic and matrix remodeling/fibrotic phenotypes according to the adiposity and the location of AT, that may be related to AT microenvironment including hypoxia and adipokines. Moreover, human ScAT progenitor cells have been identified as target cells for ScATM-derived TGFß and as a potential source of fibrosis through their induction of myofibroblast-like cells.


Assuntos
Tecido Adiposo/metabolismo , Macrófagos/metabolismo , Miofibroblastos/citologia , Obesidade/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Tecido Adiposo/citologia , Biomarcadores/metabolismo , Western Blotting , Composição Corporal , Índice de Massa Corporal , Células Cultivadas , Perfilação da Expressão Gênica , Humanos , Técnicas Imunoenzimáticas , Macrófagos/citologia , Miofibroblastos/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Omento/citologia , Omento/metabolismo , Fenótipo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Gordura Subcutânea/citologia , Gordura Subcutânea/metabolismo , Fator de Crescimento Transformador beta/genética
14.
Methods Mol Biol ; 702: 419-27, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21082419

RESUMO

In this chapter, we describe a method to isolate and to expand multipotent adipose-derived stem (hMADS) cells from human adipose tissue. We also describe culture conditions to differentiate them into adipocytes at a high rate. This culture system provides a powerful means for studying the first steps of human adipose cell development and a route for investigating effects of drugs on the biology of adipocytes. Finally, we provide a protocol to investigate gene function during proliferation and differentiation of hMADS cells by means of siRNA-mediated gene silencing approaches or forced expression by transducing hMADS cells permissive to infection with murine retrovirus vectors.


Assuntos
Tecido Adiposo/citologia , Técnicas de Cultura de Células/métodos , Células-Tronco Multipotentes/citologia , Adipócitos/citologia , Diferenciação Celular , Proliferação de Células , Separação Celular , Forma Celular , Congelamento , Inativação Gênica , Humanos , Células-Tronco Multipotentes/metabolismo , Células-Tronco Multipotentes/virologia , Nitrogênio , RNA Interferente Pequeno/metabolismo , Retroviridae/genética , Transfecção
15.
Genome Biol ; 12(7): R64, 2011 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-21767385

RESUMO

BACKGROUND: In severe obesity, as well as in normal development, the growth of adipose tissue is the result of an increase in adipocyte size and numbers, which is underlain by the stimulation of adipogenic differentiation of precursor cells. A better knowledge of the pathways that regulate adipogenesis is therefore essential for an improved understanding of adipose tissue expansion. As microRNAs (miRNAs) have a critical role in many differentiation processes, our study aimed to identify the role of miRNA-mediated gene silencing in the regulation of adipogenic differentiation. RESULTS: We used deep sequencing to identify small RNAs that are differentially expressed during adipogenesis of adipose tissue-derived stem cells. This approach revealed the un-annotated miR-642a-3p as a highly adipocyte-specific miRNA. We then focused our study on the miR-30 family, which was also up-regulated during adipogenic differentiation and for which the role in adipogenesis had not yet been elucidated. Inhibition of the miR-30 family blocked adipogenesis, whilst over-expression of miR-30a and miR-30d stimulated this process. We additionally showed that both miR-30a and miR-30d target the transcription factor RUNX2, and stimulate adipogenesis via the modulation of this major regulator of osteogenesis. CONCLUSIONS: Overall, our data suggest that the miR-30 family plays a central role in adipocyte development. Moreover, as adipose tissue-derived stem cells can differentiate into either adipocytes or osteoblasts, the down-regulation of the osteogenesis regulator RUNX2 represents a plausible mechanism by which miR-30 miRNAs may contribute to adipogenic differentiation of adipose tissue-derived stem cells.


Assuntos
Adipócitos/metabolismo , Adipogenia/genética , MicroRNAs/metabolismo , Especificidade de Órgãos/genética , Diferenciação Celular/genética , Subunidade alfa 1 de Fator de Ligação ao Core/antagonistas & inibidores , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Regulação da Expressão Gênica no Desenvolvimento , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , MicroRNAs/química , Osteogênese/genética , Análise de Sequência de RNA , Regulação para Cima/genética
16.
Diabetes ; 59(10): 2513-21, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20530742

RESUMO

OBJECTIVE: Growth of white adipose tissue takes place in normal development and in obesity. A pool of adipose progenitors is responsible for the formation of new adipocytes and for the potential of this tissue to expand in response to chronic energy overload. However, factors controlling self-renewal of human adipose progenitors are largely unknown. We investigated the expression profile and the role of activin A in this process. RESEARCH DESIGN AND METHODS: Expression of INHBA/activin A was investigated in three types of human adipose progenitors. We then analyzed at the molecular level the function of activin A during human adipogenesis. We finally investigated the status of activin A in adipose tissues of lean and obese subjects and analyzed macrophage-induced regulation of its expression. RESULTS: INHBA/activin A is expressed by adipose progenitors from various fat depots, and its expression dramatically decreases as progenitors differentiate into adipocytes. Activin A regulates the number of undifferentiated progenitors. Sustained activation or inhibition of the activin A pathway impairs or promotes, respectively, adipocyte differentiation via the C/EBPß-LAP and Smad2 pathway in an autocrine/paracrine manner. Activin A is expressed at higher levels in adipose tissue of obese patients compared with the expression levels in lean subjects. Indeed, activin A levels in adipose progenitors are dramatically increased by factors secreted by macrophages derived from obese adipose tissue. CONCLUSIONS: Altogether, our data show that activin A plays a significant role in human adipogenesis. We propose a model in which macrophages that are located in adipose tissue regulate adipose progenitor self-renewal through activin A.


Assuntos
Ativinas/fisiologia , Tecido Adiposo/citologia , Glucosefosfato Desidrogenase/genética , Obesidade Mórbida/patologia , Células-Tronco/citologia , Magreza/patologia , Ativinas/genética , Ativinas/farmacologia , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/patologia , Adulto , Diferenciação Celular , Divisão Celular , RNA Polimerases Dirigidas por DNA/efeitos dos fármacos , RNA Polimerases Dirigidas por DNA/genética , Dexametasona/farmacologia , Regulação da Expressão Gênica , Glucosefosfato Desidrogenase/efeitos dos fármacos , Humanos , Obesidade Mórbida/genética , Obesidade Mórbida/prevenção & controle , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/efeitos dos fármacos , Células-Tronco/patologia , Proteína de Ligação a TATA-Box/efeitos dos fármacos , Proteína de Ligação a TATA-Box/genética
18.
Stem Cells Dev ; 18(3): 457-63, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18690793

RESUMO

Key events leading to terminal differentiation of preadipocytes into adipocytes have been identified in recent years. However, signaling pathways involved in the decision of stem cells to follow the adipogenic lineage have not yet been characterized. We have previously shown that differentiating mouse embryonic stem (mES) cells give rise to functional adipocytes upon an early treatment with retinoic acid (RA). The goal of this work was to identify regulators of RA-induced commitment of mES cells to the adipocyte lineage. First, we investigated the role of RA receptor (RAR) isotypes in the induction of mES cell adipogenesis. Using synthetic retinoids selective of RAR isotypes, we show that RARbeta activation is both sufficient and necessary to trigger commitment of mES cells to adipocytes. Then, we performed a small-scale drug screening to find signaling pathways involved in RARbeta-induced mES cell adipogenesis. We show that pharmacological inhibitors of glycogen synthase kinase (GSK) 3, completely inhibit RARbeta-induced adipogenesis in mES cells. This finding uncovers the requirement of active GSK3 in RARbeta-induced commitment of mES cells toward the adipocyte lineage. Finally, we investigated the role of the Wnt pathway, in which GSK3 is a critical negative regulator, in adipocyte commitment by analyzing Wnt pathway activity in RA- and RARbeta-induced mES cell adipogenesis. Our results suggest that although RARbeta and active GSK3 are required for RA-induced adipogenesis, they might be acting through a Wnt pathway-independent mechanism.


Assuntos
Adipócitos/fisiologia , Adipogenia/fisiologia , Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/fisiologia , Quinase 3 da Glicogênio Sintase/metabolismo , Receptores do Ácido Retinoico/metabolismo , Adipócitos/citologia , Animais , Linhagem Celular , Linhagem da Célula , Células-Tronco Embrionárias/citologia , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/genética , Camundongos , Receptores do Ácido Retinoico/genética , Transdução de Sinais/fisiologia , Tretinoína/química , Tretinoína/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
19.
Curr Protoc Cell Biol ; Chapter 23: Unit 23.4, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18228505

RESUMO

The authors describe protocols for culture conditions in which mouse ES cells can be maintained in an undifferentiated state or committed to undergo adipocyte differentiation at a high rate and in a highly reproducible fashion. There is also a protocol for maintaining and differentiating human adult stem cells, isolated form adipose tissue and from bone marrow, into adipocytes. These culture systems provide a powerful means for studying the first step of adipose cell development and a means to investigate effects of drugs on the biology of adipocytes. There are also protocols for detection of adipocytes and analysis of their gene expression.


Assuntos
Adipócitos/citologia , Células-Tronco Adultas/citologia , Técnicas de Cultura de Células/métodos , Células-Tronco Embrionárias/citologia , Células-Tronco Multipotentes/citologia , Adulto , Células-Tronco Adultas/efeitos dos fármacos , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Diferenciação Celular , Células Cultivadas/citologia , Células Cultivadas/efeitos dos fármacos , Técnicas de Cocultura , Meios de Cultura/farmacologia , Células-Tronco Embrionárias/efeitos dos fármacos , Perfilação da Expressão Gênica/métodos , Humanos , Camundongos , Células-Tronco Multipotentes/efeitos dos fármacos
20.
Acta cir. bras ; 27(4): 283-289, Apr. 2012. ilus
Artigo em Inglês | LILACS | ID: lil-622351

RESUMO

PURPOSE: To evaluate the implant of human adipose derived stem cells (ADSC) delivered in hyaluronic acid gel (HA), injected in the subcutaneous of athymic mice. METHODS: Control implants -HA plus culture media was injected in the subcutaneous of the left sub scapular area of 12 athymic mice. ADSC implants: HA plus ADSC suspended in culture media was injected in the subcutaneous, at the contra lateral area, of the same animals. With eight weeks, animals were sacrificed and the recovered implants were processed for extraction of genomic DNA, and histological study by hematoxilin-eosin staining and immunufluorescence using anti human vimentin and anti von Willebrand factor antibodies. RESULTS: Controls: Not visualized at the injection site. An amorphous substance was observed in hematoxilin-eosin stained sections. Human vimentin and anti von Willebrand factor were not detected. No human DNA was detected. ADSC implants - A plug was visible at the site of injection. Fusiform cells were observed in sections stained by hematoxilin- eosin and both human vimentin and anti von Willebrand factor were detected by immunofluorescence. The presence of human DNA was confirmed. CONCLUSION: The delivery of human adipose derived stem cells in preparations of hyaluronic acid assured cells engraftment at the site of injection.


OBJETIVO: Avaliar o implante de células tronco do tecido adiposo humano (CTTAH) em gel de ácido hialurônico (AH), injetados no tecido subcutâneo de camundongos atímicos. MÉTODOS: Implantes controle - HA com meio de cultura foram injetados no tecido subcutâneo da região infraescapular esquerda de 12 camundongos atímicos. Implantes de CTTAH: HA com CTTAH suspensas em meio de cultura foi injetado no subcutâneo da região contra lateral, dos mesmos animais. Com oito semanas, os animais foram sacrificados e os implantes recuperados foram processados para extração de DNA genômico, estudo histológico por coloração por hematoxilina eosina e imnuoflurescência utilizando anticorpos anti vimentina humana e anti fator de von Willebrand. RESULTADOS: Controles - implantes não visualizados no local da injeção. Uma substância amorfa foi observada nos cortes corados por hematoxilina eosina. Vimentina humana e fator anti von Willebrand não foram identificados. DNA humano não foi detectado. Implantes de CTTAH - Uma massa era visível no local da injeção. Células fusiformes foram observadas nos corte corados com hematoxilina eosina. Tanto vimentina humana quanto fator de von Willebrand foram identificados pela imunofluorescência. A presença de DNA humano foi confirmada. CONCLUSÃO: O implante de células tronco do tecido adiposo humano em veículo de ácido hialurônico gel assegurou a manutenção das células no local do implante.


Assuntos
Adulto , Animais , Feminino , Humanos , Camundongos , Adipócitos/transplante , Tecido Adiposo/citologia , Ácido Hialurônico/administração & dosagem , Transplante de Células-Tronco/métodos , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Células Cultivadas , Proliferação de Células/efeitos dos fármacos , Imunofluorescência , Implantes Experimentais , Injeções Subcutâneas/métodos , Camundongos Nus , Modelos Animais , Engenharia Tecidual/métodos , Vimentina/análise , Fator de von Willebrand/análise , Fator de von Willebrand/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA