Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 14(1): 1160, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36859433

RESUMEN

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.


Asunto(s)
Dolor , Canales de Potasio de Dominio Poro en Tándem , Animales , Ratones , Evaluación Preclínica de Medicamentos , Nociceptores , Nematodos , Canales de Potasio de Dominio Poro en Tándem/antagonistas & inhibidores
2.
Cell ; 184(2): 534-544.e11, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33373586

RESUMEN

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.


Asunto(s)
Canales de Potasio con Entrada de Voltaje/metabolismo , Subunidades de Proteína/metabolismo , Animales , Anoctamina-1/metabolismo , Calcio/metabolismo , Canales de Cloruro/metabolismo , Células HEK293 , Humanos , Túbulos Renales Proximales/metabolismo , Ratones , Proteínas Mutantes/metabolismo , Péptidos/metabolismo , Polimorfismo Genético , Canales de Potasio con Entrada de Voltaje/química , Canales de Potasio con Entrada de Voltaje/genética , Unión Proteica , Dominios Proteicos , Sistema Renina-Angiotensina
3.
Biol Aujourdhui ; 213(1-2): 51-57, 2019.
Artículo en Francés | MEDLINE | ID: mdl-31274103

RESUMEN

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.


Asunto(s)
Trastornos Migrañosos/genética , Canales de Potasio/genética , Animales , Humanos , Trastornos Migrañosos/metabolismo , Canales de Potasio/química , Canales de Potasio/clasificación , Canales de Potasio/metabolismo , Canales de Potasio de Dominio Poro en Tándem/genética , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Multimerización de Proteína/fisiología , Transducción de Señal/genética
4.
Neuron ; 101(2): 232-245.e6, 2019 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-30573346

RESUMEN

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.


Asunto(s)
Potenciales de Acción/genética , Trastornos Migrañosos/patología , Mutación/genética , Neuronas/fisiología , Canales de Potasio de Dominio Poro en Tándem/genética , Potenciales de Acción/efectos de los fármacos , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Expresión Génica/genética , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Trastornos Migrañosos/inducido químicamente , Trastornos Migrañosos/genética , Trastornos Migrañosos/fisiopatología , Modelos Biológicos , Modelos Moleculares , Neurotransmisores/toxicidad , Óxido Nítrico/toxicidad , Oocitos , Canales de Potasio/genética , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Ratas , Ratas Sprague-Dawley , Xenopus
5.
Proc Natl Acad Sci U S A ; 113(15): 4194-9, 2016 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-27035963

RESUMEN

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.


Asunto(s)
Canales de Potasio de Dominio Poro en Tándem/metabolismo , Línea Celular , Dimerización , Humanos , Concentración de Iones de Hidrógeno , Canales de Potasio de Dominio Poro en Tándem/química
6.
Diabetes ; 63(10): 3253-65, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24789919

RESUMEN

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.


Asunto(s)
Adipocitos Marrones/metabolismo , Adipocitos Blancos/metabolismo , Adipogénesis/fisiología , Animales , Metabolismo Energético/fisiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Oxidación-Reducción , Consumo de Oxígeno/fisiología , PPAR gamma/metabolismo , Células Madre
7.
Stem Cells ; 32(6): 1459-67, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24302443

RESUMEN

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.


Asunto(s)
Adipocitos Marrones/citología , Adipocitos Blancos/citología , Diferenciación Celular , Células Madre Pluripotentes Inducidas/citología , Factores de Transcripción Paired Box/metabolismo , Adipocitos Marrones/efectos de los fármacos , Adipocitos Marrones/metabolismo , Adipocitos Blancos/efectos de los fármacos , Adipocitos Blancos/metabolismo , Adipogénesis/efectos de los fármacos , Anciano de 80 o más Años , Animales , Diferenciación Celular/efectos de los fármacos , Femenino , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Masculino , Ratones , Factor de Transcripción PAX3 , Fenotipo , Tretinoina/farmacología
8.
Biochem Biophys Res Commun ; 430(3): 871-5, 2013 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-23268339

RESUMEN

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.


Asunto(s)
Adipocitos/citología , Adipogénesis/fisiología , Tejido Adiposo/citología , Membrana Celular/metabolismo , Células Madre/citología , 5'-Nucleotidasa/biosíntesis , 5'-Nucleotidasa/genética , Adipogénesis/efectos de los fármacos , Tejido Adiposo/efectos de los fármacos , Antígenos CD/biosíntesis , Antígenos CD/genética , Biomarcadores/metabolismo , Antígeno CD146/biosíntesis , Antígeno CD146/genética , Línea Celular , Endoglina , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Factor 2 de Crecimiento de Fibroblastos/farmacología , Citometría de Flujo , Proteínas Ligadas a GPI/biosíntesis , Proteínas Ligadas a GPI/genética , Humanos , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptores de Superficie Celular/biosíntesis , Receptores de Superficie Celular/genética
9.
PLoS One ; 7(2): e31274, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22355352

RESUMEN

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.


Asunto(s)
Tejido Adiposo/metabolismo , Macrófagos/metabolismo , Miofibroblastos/citología , Obesidad/metabolismo , Células Madre/citología , Células Madre/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Tejido Adiposo/citología , Biomarcadores/metabolismo , Western Blotting , Composición Corporal , Índice de Masa Corporal , Células Cultivadas , Perfilación de la Expresión Génica , Humanos , Técnicas para Inmunoenzimas , Macrófagos/citología , Miofibroblastos/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Epiplón/citología , Epiplón/metabolismo , Fenotipo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Grasa Subcutánea/citología , Grasa Subcutánea/metabolismo , Factor de Crecimiento Transformador beta/genética
10.
Am J Stem Cells ; 1(1): 42-7, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23671796

RESUMEN

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.

11.
Genome Biol ; 12(7): R64, 2011 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-21767385

RESUMEN

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.


Asunto(s)
Adipocitos/metabolismo , Adipogénesis/genética , MicroARNs/metabolismo , Especificidad de Órganos/genética , Diferenciación Celular/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/antagonistas & inhibidores , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Regulación del Desarrollo de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , MicroARNs/química , Osteogénesis/genética , Análisis de Secuencia de ARN , Regulación hacia Arriba/genética
12.
Methods Mol Biol ; 702: 419-27, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21082419

RESUMEN

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.


Asunto(s)
Tejido Adiposo/citología , Técnicas de Cultivo de Célula/métodos , Células Madre Multipotentes/citología , Adipocitos/citología , Diferenciación Celular , Proliferación Celular , Separación Celular , Forma de la Célula , Congelación , Silenciador del Gen , Humanos , Células Madre Multipotentes/metabolismo , Células Madre Multipotentes/virología , Nitrógeno , ARN Interferente Pequeño/metabolismo , Retroviridae/genética , Transfección
13.
Genome Biol ; 11(8): R80, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20678241

RESUMEN

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.


Asunto(s)
Adipocitos/citología , Adipogénesis/genética , Células Madre Embrionarias/citología , Perfilación de la Expresión Génica , Animales , Sitios de Unión , Biología Computacional/métodos , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Estudios de Asociación Genética , Genoma , Ratones , Factores de Transcripción
14.
Diabetes ; 59(10): 2513-21, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20530742

RESUMEN

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.


Asunto(s)
Activinas/fisiología , Tejido Adiposo/citología , Glucosafosfato Deshidrogenasa/genética , Obesidad Mórbida/patología , Células Madre/citología , Delgadez/patología , Activinas/genética , Activinas/farmacología , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/patología , Adulto , Diferenciación Celular , División Celular , ARN Polimerasas Dirigidas por ADN/efectos de los fármacos , ARN Polimerasas Dirigidas por ADN/genética , Dexametasona/farmacología , Regulación de la Expresión Génica , Glucosafosfato Deshidrogenasa/efectos de los fármacos , Humanos , Obesidad Mórbida/genética , Obesidad Mórbida/prevención & control , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/efectos de los fármacos , Células Madre/patología , Proteína de Unión a TATA-Box/efectos de los fármacos , Proteína de Unión a TATA-Box/genética
15.
Stem Cells ; 28(4): 753-64, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20135684

RESUMEN

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.


Asunto(s)
Adipocitos/citología , Técnicas de Cultivo de Célula/métodos , Adipocitos/inmunología , Adipocitos/metabolismo , Adolescente , Adulto , Animales , Antígenos CD34/inmunología , Diferenciación Celular , Linaje de la Célula , Separación Celular , Niño , Preescolar , Femenino , Regulación de la Expresión Génica , Humanos , Lactante , Masculino , Ratones , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/inmunología
16.
Bioorg Med Chem ; 17(9): 3388-93, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19369079

RESUMEN

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.


Asunto(s)
Adipogénesis/efectos de los fármacos , Aminoácidos/síntesis química , Aminoácidos/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Compuestos Organofosforados/síntesis química , Compuestos Organofosforados/farmacología , Osteogénesis/efectos de los fármacos , Aminoácidos/química , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Estructura Molecular , Compuestos Organofosforados/química , Difracción de Rayos X
17.
Mol Ther ; 17(6): 1064-72, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19352326

RESUMEN

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.


Asunto(s)
Tejido Adiposo/citología , Células Madre Multipotentes/metabolismo , Músculo Esquelético/citología , Proteína MioD/genética , Proteína MioD/metabolismo , Adipogénesis/genética , Adipogénesis/fisiología , Animales , Western Blotting , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Fusión Celular , Línea Celular , Células Cultivadas , Distrofina/metabolismo , Citometría de Flujo , Vectores Genéticos/genética , Humanos , Inmunohistoquímica , Lentivirus/genética , Masculino , Ratones , Ratones Endogámicos mdx , Células Madre Multipotentes/citología , Fibras Musculares Esqueléticas/citología , Músculo Esquelético/metabolismo , Proteína MioD/fisiología , Mioblastos/citología , Mioblastos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Stem Cells Dev ; 18(3): 457-63, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18690793

RESUMEN

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.


Asunto(s)
Adipocitos/fisiología , Adipogénesis/fisiología , Diferenciación Celular/fisiología , Células Madre Embrionarias/fisiología , Glucógeno Sintasa Quinasa 3/metabolismo , Receptores de Ácido Retinoico/metabolismo , Adipocitos/citología , Animales , Línea Celular , Linaje de la Célula , Células Madre Embrionarias/citología , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/genética , Ratones , Receptores de Ácido Retinoico/genética , Transducción de Señal/fisiología , Tretinoina/química , Tretinoina/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
19.
BMC Cell Biol ; 9: 11, 2008 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-18271953

RESUMEN

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.


Asunto(s)
Adipocitos/citología , Tejido Adiposo/citología , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Células Madre/citología , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Adipogénesis/efectos de los fármacos , Adipogénesis/fisiología , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Preescolar , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Inmunohistoquímica , Lactante , Cloruro de Litio/farmacología , Masculino , Células Madre/efectos de los fármacos , Células Madre/metabolismo
20.
Curr Protoc Cell Biol ; Chapter 23: Unit 23.4, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18228505

RESUMEN

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.


Asunto(s)
Adipocitos/citología , Células Madre Adultas/citología , Técnicas de Cultivo de Célula/métodos , Células Madre Embrionarias/citología , Células Madre Multipotentes/citología , Adulto , Células Madre Adultas/efectos de los fármacos , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Diferenciación Celular , Células Cultivadas/citología , Células Cultivadas/efectos de los fármacos , Técnicas de Cocultivo , Medios de Cultivo/farmacología , Células Madre Embrionarias/efectos de los fármacos , Perfilación de la Expresión Génica/métodos , Humanos , Ratones , Células Madre Multipotentes/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...