Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Cell ; 184(2): 534-544.e11, 2021 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-33373586

RESUMO

Determination of what is the specificity of subunits composing a protein complex is essential when studying gene variants on human pathophysiology. The pore-forming α-subunit KCNQ1, which belongs to the voltage-gated ion channel superfamily, associates to its ß-auxiliary subunit KCNE1 to generate the slow cardiac potassium IKs current, whose dysfunction leads to cardiac arrhythmia. Using pharmacology, gene invalidation, and single-molecule fluorescence assays, we found that KCNE1 fulfils all criteria of a bona fide auxiliary subunit of the TMEM16A chloride channel, which belongs to the anoctamin superfamily. Strikingly, assembly with KCNE1 switches TMEM16A from a calcium-dependent to a voltage-dependent ion channel. Importantly, clinically relevant inherited mutations within the TMEM16A-regulating domain of KCNE1 abolish the TMEM16A modulation, suggesting that the TMEM16A-KCNE1 current may contribute to inherited pathologies. Altogether, these findings challenge the dogma of the specificity of auxiliary subunits regarding protein complexes and questions ion channel classification.


Assuntos
Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Subunidades Proteicas/metabolismo , Animais , Anoctamina-1/metabolismo , Cálcio/metabolismo , Canais de Cloreto/metabolismo , Células HEK293 , Humanos , Túbulos Renais Proximais/metabolismo , Camundongos , Proteínas Mutantes/metabolismo , Peptídeos/metabolismo , Polimorfismo Genético , Canais de Potássio de Abertura Dependente da Tensão da Membrana/química , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Ligação Proteica , Domínios Proteicos , Sistema Renina-Angiotensina
2.
Proc Natl Acad Sci U S A ; 113(15): 4194-9, 2016 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-27035963

RESUMO

Twik-related K(+) channel 1 (TREK1), TREK2, and Twik-related arachidonic-acid stimulated K(+) channel (TRAAK) form the TREK subfamily of two-pore-domain K(+) (K2P) channels. Despite sharing up to 78% sequence homology and overlapping expression profiles in the nervous system, these channels show major differences in their regulation by physiological stimuli. For instance, TREK1 is inhibited by external acidification, whereas TREK2 is activated. Here, we investigated the ability of the members of the TREK subfamily to assemble to form functional heteromeric channels with novel properties. Using single-molecule pull-down (SiMPull) from HEK cell lysate and subunit counting in the plasma membrane of living cells, we show that TREK1, TREK2, and TRAAK readily coassemble. TREK1 and TREK2 can each heterodimerize with TRAAK, but do so less efficiently than with each other. We functionally characterized the heterodimers and found that all combinations form outwardly rectifying potassium-selective channels but with variable voltage sensitivity and pH regulation. TREK1-TREK2 heterodimers show low levels of activity at physiological external pH but, unlike their corresponding homodimers, are activated by both acidic and alkaline conditions. Modeling based on recent crystal structures, along with mutational analysis, suggests that each subunit within a TREK1-TREK2 channel is regulated independently via titratable His. Finally, TREK1/TRAAK heterodimers differ in function from TRAAK homodimers in two critical ways: they are activated by both intracellular acidification and alkalinization and are regulated by the enzyme phospholipase D2. Thus, heterodimerization provides a means for diversifying functionality through an expansion of the channel types within the K2P channels.


Assuntos
Canais de Potássio de Domínios Poros em Tandem/metabolismo , Linhagem Celular , Dimerização , Humanos , Concentração de Íons de Hidrogênio , Canais de Potássio de Domínios Poros em Tandem/química
3.
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
4.
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
5.
Nat Commun ; 14(1): 1160, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36859433

RESUMO

By endowing light control of neuronal activity, optogenetics and photopharmacology are powerful methods notably used to probe the transmission of pain signals. However, costs, animal handling and ethical issues have reduced their dissemination and routine use. Here we report LAKI (Light Activated K+ channel Inhibitor), a specific photoswitchable inhibitor of the pain-related two-pore-domain potassium TREK and TRESK channels. In the dark or ambient light, LAKI is inactive. However, alternating transdermal illumination at 365 nm and 480 nm reversibly blocks and unblocks TREK/TRESK current in nociceptors, enabling rapid control of pain and nociception in intact and freely moving mice and nematode. These results demonstrate, in vivo, the subcellular localization of TREK/TRESK at the nociceptor free nerve endings in which their acute inhibition is sufficient to induce pain, showing LAKI potential as a valuable tool for TREK/TRESK channel studies. More importantly, LAKI gives the ability to reversibly remote-control pain in a non-invasive and physiological manner in naive animals, which has utility in basic and translational pain research but also in in vivo analgesic drug screening and validation, without the need of genetic manipulations or viral infection.


Assuntos
Dor , Canais de Potássio de Domínios Poros em Tandem , Animais , Camundongos , Avaliação Pré-Clínica de Medicamentos , Nociceptores , Nematoides , Canais de Potássio de Domínios Poros em Tandem/antagonistas & inibidores
6.
J Exp Med ; 201(9): 1397-405, 2005 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-15867092

RESUMO

Here, we report the isolation of a human multipotent adipose-derived stem (hMADS) cell population from adipose tissue of young donors. hMADS cells display normal karyotype; have active telomerase; proliferate >200 population doublings; and differentiate into adipocytes, osteoblasts, and myoblasts. Flow cytometry analysis indicates that hMADS cells are CD44+, CD49b+, CD105+, CD90+, CD13+, Stro-1(-), CD34-, CD15-, CD117-, Flk-1(-), gly-A(-), CD133-, HLA-DR(-), and HLA-I(low). Transplantation of hMADS cells into the mdx mouse, an animal model of Duchenne muscular dystrophy, results in substantial expression of human dystrophin in the injected tibialis anterior and the adjacent gastrocnemius muscle. Long-term engraftment of hMADS cells takes place in nonimmunocompromised animals. Based on the small amounts of an easily available tissue source, their strong capacity for expansion ex vivo, their multipotent differentiation, and their immune-privileged behavior, our results suggest that hMADS cells will be an important tool for muscle cell-mediated therapy.


Assuntos
Tecido Adiposo/citologia , Diferenciação Celular , Distrofina/metabolismo , Regulação da Expressão Gênica , Imunocompetência/fisiologia , Células-Tronco Multipotentes/transplante , Animais , Criança , Pré-Escolar , Primers do DNA , Feminino , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Lactente , Cariotipagem , Masculino , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante Heterólogo
7.
Stem Cells ; 28(4): 753-64, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20135684

RESUMO

The differentiation of multipotent cells into undesirable lineages is a significant risk factor when performing cell therapy. In muscular diseases, myofiber loss can be associated with progressive fat accumulation that is one of the primary factors leading to decline of muscular strength. Therefore, to avoid any contribution of injected multipotent cells to fat deposition, we have searched for a highly myogenic but nonadipogenic muscle-derived cell population. We show that the myogenic marker CD56, which is the gold standard for myoblast-based therapy, was unable to separate muscle cells into myogenic and adipogenic fractions. Conversely, using the stem cell marker CD34, we were able to sort two distinct populations, CD34(+) and CD34(-), which have been thoroughly characterized in vitro and in vivo using an immunodeficient Rag2(-/-)gamma(c) (-/-) mouse model of muscle regeneration with or without adipose deposition. Our results demonstrate that both populations have equivalent capacities for in vitro amplification. The CD34(+) cells and CD34(-) cells exhibit equivalent myogenic potential, but only the CD34(-) population fails to differentiate into adipocytes in vitro and in vivo after transplantation into regenerative fat muscle. These data indicate that the muscle-derived cells constitute a heterogeneous population of cells with various differentiation potentials. The simple CD34 sorting allows isolation of myogenic cells with no adipogenic potential and therefore could be of high interest for cell therapy when fat is accumulated in diseased muscle.


Assuntos
Adipócitos/citologia , Técnicas de Cultura de Células/métodos , Adipócitos/imunologia , Adipócitos/metabolismo , Adolescente , Adulto , Animais , Antígenos CD34/imunologia , Diferenciação Celular , Linhagem da Célula , Separação Celular , Criança , Pré-Escolar , Feminino , Regulação da Expressão Gênica , Humanos , Lactente , Masculino , Camundongos , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/imunologia
8.
Mol Ther ; 17(6): 1064-72, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19352326

RESUMO

Muscle disorders such as Duchenne muscular dystrophy (DMD) still need effective treatments, and mesenchymal stem cells (MSCs) may constitute an attractive cell therapy alternative because they are multipotent and accessible in adult tissues. We have previously shown that human multipotent adipose-derived stem (hMADS) cells were able to restore dystrophin expression in the mdx mouse. The goal of this work was to improve the myogenic potential of hMADS cells and assess the impact on muscle repair. Forced expression of MyoD in vitro strongly induced myogenic differentiation while the adipogenic differentiation was inhibited. Moreover, MyoD-expressing hMADS cells had the capacity to fuse with DMD myoblasts and to restore dystrophin expression. Importantly, transplantation of these modified hMADS cells into injured muscles of immunodepressed Rag2(-/-)gammaC(-/-) mice resulted in a substantial increase in the number of hMADS cell-derived fibers. Our approach combined the easy access of MSCs from adipose tissue, the highly efficient lentiviral transduction of these cells, and the specific improvement of myogenic differentiation through the forced expression of MyoD. Altogether our results highlight the capacity of modified hMADS cells to contribute to muscle repair and their potential to deliver a repairing gene to dystrophic muscles.


Assuntos
Tecido Adiposo/citologia , Células-Tronco Multipotentes/metabolismo , Músculo Esquelético/citologia , Proteína MyoD/genética , Proteína MyoD/metabolismo , Adipogenia/genética , Adipogenia/fisiologia , Animais , Western Blotting , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Fusão Celular , Linhagem Celular , Células Cultivadas , Distrofina/metabolismo , Citometria de Fluxo , Vetores Genéticos/genética , Humanos , Imuno-Histoquímica , Lentivirus/genética , Masculino , Camundongos , Camundongos Endogâmicos mdx , Células-Tronco Multipotentes/citologia , Fibras Musculares Esqueléticas/citologia , Músculo Esquelético/metabolismo , Proteína MyoD/fisiologia , Mioblastos/citologia , Mioblastos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
Bioorg Med Chem ; 17(9): 3388-93, 2009 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-19369079

RESUMO

The one pot reaction of amino acids with diethylphosphite and formaldehyde yielded N,N-bis(phosphonomethyl)amino acids. This synthetic route does not require harsh reagents to cleave the ester group. The molecular structures of the new compounds were determined by X-ray diffraction methods. By employing DFT calculations the hydrolysis of the intermediate phosphonic esters to the respective acids could be explained by the decreasing P-OEt bond strength for C(alpha)-bisalkylated amino acids. Biological evaluation on the adipogenic and osteogenic differentiation of mesenchymal stem cells revealed no modification of the adipocyte differentiation, but inhibition of osteoblast formation at concentrations without detectable cytotoxicity.


Assuntos
Adipogenia/efeitos dos fármacos , Aminoácidos/síntese química , Aminoácidos/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Compostos Organofosforados/síntese química , Compostos Organofosforados/farmacologia , Osteogênese/efeitos dos fármacos , Aminoácidos/química , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Estrutura Molecular , Compostos Organofosforados/química , Difração de Raios X
10.
Biol Aujourdhui ; 213(1-2): 51-57, 2019.
Artigo em Francês | MEDLINE | ID: mdl-31274103

RESUMO

Migraine is a common, disabling neurological disorder with genetic, environmental and hormonal components and a prevalence estimated at ∼15%. Migraine episodes are notably related, among several factors, to electric hyperexcitability in sensory neurons. Their electrical activity is controlled by ion channels that generate current, specifically by the two-pore-domain potassium, K2P, channels, which inhibit electrical activity. Mutation in the gene encoding TRESK, a K2P channel, causes the formation of TRESK-MT1, the expected non-functional C-terminal truncated TRESK channel, and an additional unexpected protein, TRESK-MT2, which corresponds to a non-functional N-terminal truncated TRESK channel, through a mechanism called frameshift mutation-induced Alternative Translation Initiation (fsATI). TRESK-MT1 is inactive but TRESK-M2 targets two other ion channels, TREK1 and TREK2, inducing a great stimulation of the neuronal electrical activity that may cause migraines. These findings identify TREK1 and TREK2 as potential molecular targets for migraine treatment and suggest that fsATI should be considered as a distinct class of mutations.


Assuntos
Transtornos de Enxaqueca/genética , Canais de Potássio/genética , Animais , Humanos , Transtornos de Enxaqueca/metabolismo , Canais de Potássio/química , Canais de Potássio/classificação , Canais de Potássio/metabolismo , Canais de Potássio de Domínios Poros em Tandem/genética , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Multimerização Proteica/fisiologia , Transdução de Sinais/genética
11.
Neuron ; 101(2): 232-245.e6, 2019 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-30573346

RESUMO

It is often unclear why some genetic mutations to a given gene contribute to neurological disorders and others do not. For instance, two mutations have previously been found to produce a dominant negative for TRESK, a two-pore-domain K+ channel implicated in migraine: TRESK-MT, a 2-bp frameshift mutation, and TRESK-C110R. Both mutants inhibit TRESK, but only TRESK-MT increases sensory neuron excitability and is linked to migraine. Here, we identify a new mechanism, termed frameshift mutation-induced alternative translation initiation (fsATI), that may explain why only TRESK-MT is associated with migraine. fsATI leads to the production of a second protein fragment, TRESK-MT2, which co-assembles with and inhibits TREK1 and TREK2, two other two-pore-domain K+ channels, to increase trigeminal sensory neuron excitability, leading to a migraine-like phenotype in rodents. These findings identify TREK1 and TREK2 as potential molecular targets in migraine and suggest that fsATI should be considered as a distinct class of mutations.


Assuntos
Potenciais de Ação/genética , Transtornos de Enxaqueca/patologia , Mutação/genética , Neurônios/fisiologia , Canais de Potássio de Domínios Poros em Tandem/genética , Potenciais de Ação/efeitos dos fármacos , Animais , Células Cultivadas , Modelos Animais de Doenças , Feminino , Expressão Gênica/genética , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transtornos de Enxaqueca/induzido quimicamente , Transtornos de Enxaqueca/genética , Transtornos de Enxaqueca/fisiopatologia , Modelos Biológicos , Modelos Moleculares , Neurotransmissores/toxicidade , Óxido Nítrico/toxicidade , Oócitos , Canais de Potássio/genética , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Ratos , Ratos Sprague-Dawley , Xenopus
12.
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
13.
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
14.
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
15.
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.

16.
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
17.
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
18.
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
19.
Genome Biol ; 11(8): R80, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20678241

RESUMO

BACKGROUND: The current epidemic of obesity has caused a surge of interest in the study of adipose tissue formation. While major progress has been made in defining the molecular networks that control adipocyte terminal differentiation, the early steps of adipocyte development and the embryonic origin of this lineage remain largely unknown. RESULTS: Here we performed genome-wide analysis of gene expression during adipogenesis of mouse embryonic stem cells (ESCs). We then pursued comprehensive bioinformatic analyses, including de novo functional annotation and curation of the generated data within the context of biological pathways, to uncover novel biological functions associated with the early steps of adipocyte development. By combining in-depth gene regulation studies and in silico analysis of transcription factor binding site enrichment, we also provide insights into the transcriptional networks that might govern these early steps. CONCLUSIONS: This study supports several biological findings: firstly, adipocyte development in mouse ESCs is coupled to blood vessel morphogenesis and neural development, just as it is during mouse development. Secondly, the early steps of adipocyte formation involve major changes in signaling and transcriptional networks. A large proportion of the transcription factors that we uncovered in mouse ESCs are also expressed in the mouse embryonic mesenchyme and in adipose tissues, demonstrating the power of our approach to probe for genes associated with early developmental processes on a genome-wide scale. Finally, we reveal a plethora of novel candidate genes for adipocyte development and present a unique resource that can be further explored in functional assays.


Assuntos
Adipócitos/citologia , Adipogenia/genética , Células-Tronco Embrionárias/citologia , Perfilação da Expressão Gênica , Animais , Sítios de Ligação , Biologia Computacional/métodos , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Estudos de Associação Genética , Genoma , Camundongos , Fatores de Transcrição
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA