Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Hum Mol Genet ; 27(8): 1447-1459, 2018 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-29438482

RESUMEN

The p.R482W hotspot mutation in A-type nuclear lamins causes familial partial lipodystrophy of Dunnigan-type (FPLD2), a lipodystrophic syndrome complicated by early onset atherosclerosis. Molecular mechanisms underlying endothelial cell dysfunction conferred by the lamin A mutation remain elusive. However, lamin A regulates epigenetic developmental pathways and mutations could perturb these functions. Here, we demonstrate that lamin A R482W elicits endothelial differentiation defects in a developmental model of FPLD2. Genome modeling in fibroblasts from patients with FPLD2 caused by the lamin A R482W mutation reveals repositioning of the mesodermal regulator T/Brachyury locus towards the nuclear center relative to normal fibroblasts, suggesting enhanced activation propensity of the locus in a developmental model of FPLD2. Addressing this issue, we report phenotypic and transcriptional alterations in mesodermal and endothelial differentiation of induced pluripotent stem cells we generated from a patient with R482W-associated FPLD2. Correction of the LMNA mutation ameliorates R482W-associated phenotypes and gene expression. Transcriptomics links endothelial differentiation defects to decreased Polycomb-mediated repression of the T/Brachyury locus and over-activation of T target genes. Binding of the Polycomb repressor complex 2 to T/Brachyury is impaired by the mutated lamin A network, which is unable to properly associate with the locus. This leads to a deregulation of vascular gene expression over time. By connecting a lipodystrophic hotspot lamin A mutation to a disruption of early mesodermal gene expression and defective endothelial differentiation, we propose that the mutation rewires the fate of several lineages, resulting in multi-tissue pathogenic phenotypes.


Asunto(s)
Células Endoteliales/metabolismo , Proteínas Fetales/genética , Regulación del Desarrollo de la Expresión Génica , Lamina Tipo A/genética , Lipodistrofia Parcial Familiar/genética , Proteínas del Grupo Polycomb/genética , Proteínas de Dominio T Box/genética , Adolescente , Adulto , Estudios de Casos y Controles , Diferenciación Celular/genética , Linaje de la Célula/genética , Células Endoteliales/patología , Femenino , Proteínas Fetales/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patología , Redes Reguladoras de Genes , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Lamina Tipo A/metabolismo , Lipodistrofia Parcial Familiar/metabolismo , Lipodistrofia Parcial Familiar/patología , Masculino , Mesodermo/metabolismo , Mesodermo/patología , Persona de Mediana Edad , Mutación , Proteínas del Grupo Polycomb/metabolismo , Cultivo Primario de Células , Unión Proteica , Transducción de Señal , Proteínas de Dominio T Box/metabolismo
2.
Curr Diab Rep ; 13(6): 757-67, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24026869

RESUMEN

Genetic lipodystrophic syndromes are rare diseases characterized by generalized or partial fat atrophy (lipoatrophy) associated with severe metabolic complications such as insulin resistance (IR), diabetes, dyslipidemia, nonalcoholic fatty liver disease, and ovarian hyperandrogenism. During the last 15 years, mutations in several genes have been shown to be responsible for monogenic forms of lipodystrophic syndromes, of autosomal dominant or recessive transmission. Although the molecular basis of lipodystrophies is heterogeneous, most mutated genes lead to impaired adipogenesis, adipocyte lipid storage, and/or formation or maintenance of the adipocyte lipid droplet (LD), showing that primary alterations of adipose tissue (AT) can result in severe systemic metabolic and endocrine consequences. The reduced expandability of AT alters its ability to buffer excess caloric intake, leading to ectopic lipid storage that impairs insulin signaling and other cellular functions ("lipotoxicity"). Genetic studies have also pointed out the close relationships between ageing, inflammatory processes, lipodystrophy, and IR.


Asunto(s)
Resistencia a la Insulina/fisiología , Lipodistrofia/genética , Envejecimiento/fisiología , Animales , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Humanos , Inflamación/genética , Inflamación/fisiopatología , Resistencia a la Insulina/genética , Lipodistrofia/fisiopatología
3.
J Clin Invest ; 131(1)2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-33393499

RESUMEN

LMNA mutations in patients are responsible for a dilated cardiomyopathy. Molecular mechanisms underlying the origin and development of the pathology are unknown. Herein, using mouse pluripotent embryonic stem cells (ESCs) and a mouse model both harboring the p.H222P Lmna mutation, we found early defects in cardiac differentiation of mutated ESCs and dilatation of mutated embryonic hearts at E13.5, pointing to a developmental origin of the disease. Using mouse ESCs, we demonstrated that cardiac differentiation of LmnaH222P/+ was impaired at the mesodermal stage. Expression of Mesp1, a mesodermal cardiogenic gene involved in epithelial-to-mesenchymal transition of epiblast cells, as well as Snai1 and Twist expression, was decreased in LmnaH222P/+ cells compared with WT cells in the course of differentiation. In turn, cardiomyocyte differentiation was impaired. ChIP assay of H3K4me1 in differentiating cells revealed a specific decrease of this histone mark on regulatory regions of Mesp1 and Twist in LmnaH222P/+ cells. Downregulation or inhibition of LSD1 that specifically demethylated H3K4me1 rescued the epigenetic landscape of mesodermal LmnaH222P/+ cells and in turn contraction of cardiomyocytes. Inhibition of LSD1 in pregnant mice or neonatal mice prevented cardiomyopathy in E13.5 LmnaH222P/H222P offspring and adults, respectively. Thus, LSD1 appeared to be a therapeutic target to prevent or cure dilated cardiomyopathy associated with a laminopathy.


Asunto(s)
Cardiomiopatías/enzimología , Cardiomiopatías/prevención & control , Histona Demetilasas/metabolismo , Laminopatías/complicaciones , Laminopatías/enzimología , Miocitos Cardíacos/enzimología , Sustitución de Aminoácidos , Animales , Cardiomiopatías/genética , Diferenciación Celular , Modelos Animales de Enfermedad , Histona Demetilasas/genética , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Laminopatías/genética , Ratones , Ratones Mutantes , Células Madre Embrionarias de Ratones/enzimología , Células Madre Embrionarias de Ratones/patología , Mutación Missense , Miocitos Cardíacos/patología
4.
Nat Metab ; 3(9): 1150-1162, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34531575

RESUMEN

Macrophages exhibit a spectrum of activation states ranging from classical to alternative activation1. Alternatively, activated macrophages are involved in diverse pathophysiological processes such as confining tissue parasites2, improving insulin sensitivity3 or promoting an immune-tolerant microenvironment that facilitates tumour growth and metastasis4. Recently, the metabolic regulation of macrophage function has come into focus as both the classical and alternative activation programmes require specific regulated metabolic reprogramming5. While most of the studies regarding immunometabolism have focussed on the catabolic pathways activated to provide energy, little is known about the anabolic pathways mediating macrophage alternative activation. In this study, we show that the anabolic transcription factor sterol regulatory element binding protein 1 (SREBP1) is activated in response to the canonical T helper 2 cell cytokine interleukin-4 to trigger the de novo lipogenesis (DNL) programme, as a necessary step for macrophage alternative activation. Mechanistically, DNL consumes NADPH, partitioning it away from cellular antioxidant defences and raising reactive oxygen species levels. Reactive oxygen species serves as a second messenger, signalling sufficient DNL, and promoting macrophage alternative activation. The pathophysiological relevance of this mechanism is validated by showing that SREBP1/DNL is essential for macrophage alternative activation in vivo in a helminth infection model.


Asunto(s)
Antioxidantes/metabolismo , Ácidos Grasos/biosíntesis , Macrófagos/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Animales , Dexametasona/farmacología , Humanos , Interleucina-4/farmacología , Lipopolisacáridos/farmacología , Activación de Macrófagos , Macrófagos/efectos de los fármacos , Ratones , Ratones Noqueados , Nippostrongylus/aislamiento & purificación , Nippostrongylus/patogenicidad , Células RAW 264.7 , Análisis de Secuencia de ARN/métodos , Infecciones por Strongylida/inmunología , Infecciones por Strongylida/parasitología , Regulación hacia Arriba
5.
Stem Cell Reports ; 16(3): 641-655, 2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33606988

RESUMEN

Increasing brown adipose tissue (BAT) mass and activation is a therapeutic strategy to treat obesity and complications. Obese and diabetic patients possess low amounts of BAT, so an efficient way to expand their mass is necessary. There is limited knowledge about how human BAT develops, differentiates, and is optimally activated. Accessing human BAT is challenging, given its low volume and anatomical dispersion. These constraints make detailed BAT-related developmental and functional mechanistic studies in humans virtually impossible. We have developed and characterized functionally and molecularly a new chemically defined protocol for the differentiation of human pluripotent stem cells (hPSCs) into brown adipocytes (BAs) that overcomes current limitations. This protocol recapitulates step by step the physiological developmental path of human BAT. The BAs obtained express BA and thermogenic markers, are insulin sensitive, and responsive to ß-adrenergic stimuli. This new protocol is scalable, enabling the study of human BAs at early stages of development.


Asunto(s)
Adipocitos Marrones/metabolismo , Adipogénesis , Tejido Adiposo Pardo/metabolismo , Técnicas de Cultivo de Célula/métodos , Células Madre Pluripotentes/metabolismo , Termogénesis , Factores de Transcripción/metabolismo , Biomarcadores/metabolismo , Diferenciación Celular , Línea Celular , Regulación del Desarrollo de la Expresión Génica , Humanos , Reproducibilidad de los Resultados
6.
PLoS One ; 15(1): e0226924, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31971958

RESUMEN

OBJECTIVES: Aging HIV-infected antiretroviral-treatment (ART)-controlled patients often present cardiovascular and metabolic comorbidities. Thus, it is mandatory that life-long used ART has no cardiometabolic toxicity. Protease inhibitors have been associated with cardiometabolic risk, integrase-strand-transfer-inhibitors (INSTI) with weight gain and the CCR5 inhibitor maraviroc with improved vascular function. We have previously reported that the INSTI dolutegravir and maraviroc improved, and ritonavir-boosted atazanavir(atazanavir/r) worsened, inflammation and senescence in human coronary artery endothelial cells (HCAEC)s from adult controls. Here, we analyzed the pathways involved in the drugs' effects on inflammation, senescence and also insulin resistance. METHODS: We analyzed the involvement of the anti-inflammatory SIRT-1 pathway in HCAECs. Then, we performed a transcriptomic analysis of the effect of dolutegravir, maraviroc and atazanavir/r and used siRNA-silencing to address ubiquitin-specific-peptidase-18 (USP18) involvement into ART effects. RESULTS: Dolutegravir reduced inflammation by decreasing NFκB activation and IL-6/IL-8/sICAM-1/sVCAM-1 secretion, as did maraviroc with a milder effect. However, when SIRT-1 was inhibited by splitomicin, the drugs anti-inflammatory effects were maintained, indicating that they were SIRT-1-independant. From the transcriptomic analysis we selected USP18, previously shown to decrease inflammation and insulin-resistance. USP18-silencing enhanced basal inflammation and senescence. Maraviroc still inhibited NFκB activation, cytokine/adhesion molecules secretion and senescence but the effects of dolutegravir and atazanavir/r were lost, suggesting that they involved USP18. Otherwise, in HCAECs, dolutegravir improved and atazanavir/r worsened insulin resistance while maraviroc had no effect. In USP18-silenced cells, basal insulin resistance was increased, but dolutegravir and atazanavir/r kept their effect on insulin sensitivity, indicating that USP18 was dispensable. CONCLUSION: USP18 reduced basal inflammation, senescence and insulin resistance in coronary endothelial cells. Dolutegravir and atazanavir/r, but not maraviroc, exerted opposite effects on inflammation and senescence that involved USP18. Otherwise, dolutegravir improved and atazanavir/r worsened insulin resistance independently of USP18. Thus, in endothelial cells, dolutegravir and atazanavir/r oppositely affected pathways leading to inflammation, senescence and insulin resistance.


Asunto(s)
Fármacos Anti-VIH/efectos adversos , Vasos Coronarios/citología , Infecciones por VIH/tratamiento farmacológico , FN-kappa B/metabolismo , Sirtuina 1/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Fármacos Anti-VIH/uso terapéutico , Sulfato de Atazanavir/efectos adversos , Sulfato de Atazanavir/uso terapéutico , Células Cultivadas , Comorbilidad , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/metabolismo , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Femenino , Compuestos Heterocíclicos con 3 Anillos/efectos adversos , Compuestos Heterocíclicos con 3 Anillos/uso terapéutico , Humanos , Resistencia a la Insulina , Masculino , Maraviroc/efectos adversos , Maraviroc/uso terapéutico , Oxazinas , Piperazinas , Piridonas , Ritonavir/efectos adversos , Ritonavir/uso terapéutico , Transducción de Señal , Ubiquitina Tiolesterasa/genética
7.
Artículo en Inglés | MEDLINE | ID: mdl-29852279

RESUMEN

The adipose organ portrays adipocytes of diverse tones: white, brown and beige, each type with distinct functions. Adipocytes orchestrate their adaptation and expansion to provide storage to excess nutrients, the quick mobilisation of fuel to supply peripheral functional demands, insulation, and, in their thermogenic form, heat generation to maintain core body temperature. Thermogenic adipocytes could be targets for anti-obesity and anti-diabetic therapeutic approaches aiming to restore adipose tissue functionality and increase energy dissipation. However, for thermogenic adipose tissue to become therapeutically relevant, a better understanding of its development and origins, its progenitors and their characteristics and the composition of its niche, is essential. Also crucial is the identification of stimuli and molecules promoting its specific differentiation and activation. Here we highlight the structural/cellular differences between human and rodent brown adipose tissue and discuss how obesity and metabolic complication affects brown and beige cells as well as how they could be targeted to improve their activation and improve global metabolic homeostasis. Finally, we describe the limitations of current research models and the advantages of new emerging approaches.


Asunto(s)
Tejido Adiposo Beige/fisiología , Tejido Adiposo Pardo/fisiología , Adipocitos/fisiología , Adipoquinas/fisiología , Animales , Humanos , Termogénesis
8.
Trends Endocrinol Metab ; 29(1): 5-7, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29110964

RESUMEN

Obesity is a major health problem without satisfactory pharmacological treatment. A promising strategy is to promote energy dissipation by activating brown/beige adipose tissue. However, for this strategy to succeed it requires improving the transferability amongst cellular, murine, and human systems and bridging the gap between basic and clinical research.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Investigación Biomédica , Obesidad/metabolismo , Termogénesis/fisiología , Animales , Humanos
9.
Nucleus ; 9(1): 235-248, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29578370

RESUMEN

Mutations in LMNA, encoding A-type lamins, are responsible for laminopathies including muscular dystrophies, lipodystrophies, and premature ageing syndromes. LMNA mutations have been shown to alter nuclear structure and stiffness, binding to partners at the nuclear envelope or within the nucleoplasm, gene expression and/or prelamin A maturation. LMNA-associated lipodystrophic features, combining generalized or partial fat atrophy and metabolic alterations associated with insulin resistance, could result from altered adipocyte differentiation or from altered fat structure. Recent studies shed some light on how pathogenic A-type lamin variants could trigger lipodystrophy, metabolic complications, and precocious cardiovascular events. Alterations in adipose tissue extracellular matrix and TGF-beta signaling could initiate metabolic inflexibility. Premature senescence of vascular cells could contribute to cardiovascular complications. In affected families, metabolic alterations occur at an earlier age across generations, which could result from epigenetic deregulation induced by LMNA mutations. Novel cellular models recapitulating adipogenic developmental pathways provide scalable tools for disease modeling and therapeutic screening.


Asunto(s)
Lamina Tipo A/genética , Lipodistrofia , Mutación , Humanos , Lamina Tipo A/metabolismo , Lipodistrofia/tratamiento farmacológico , Lipodistrofia/genética , Lipodistrofia/metabolismo
10.
Diabetes ; 66(6): 1470-1478, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28270520

RESUMEN

Activation of thermogenic beige adipocytes has recently emerged as a promising therapeutic target in obesity and diabetes. Relevant human models for beige adipocyte differentiation are essential to implement such therapeutic strategies. We report a straightforward and efficient protocol to generate functional human beige adipocytes from human induced pluripotent stem cells (hiPSCs). Without overexpression of exogenous adipogenic genes, our method recapitulates an adipogenic developmental pathway through successive mesodermal and adipogenic progenitor stages. hiPSC-derived adipocytes are insulin sensitive and display beige-specific markers and functional properties, including upregulation of thermogenic genes, increased mitochondrial content, and increased oxygen consumption upon activation with cAMP analogs. Engraftment of hiPSC-derived adipocytes in mice produces well-organized and vascularized adipose tissue, capable of ß-adrenergic-responsive glucose uptake. Our model of human beige adipocyte development provides a new and scalable tool for disease modeling and therapeutic screening.


Asunto(s)
Adipocitos Beige/metabolismo , Tejido Adiposo/metabolismo , Técnicas de Reprogramación Celular/métodos , Glucosa/metabolismo , Células Madre Pluripotentes Inducidas/citología , Resistencia a la Insulina , Obesidad , Termogénesis/genética , Adipocitos Beige/citología , Adipocitos Beige/efectos de los fármacos , Adipocitos Beige/trasplante , Adipogénesis , Tejido Adiposo/efectos de los fármacos , Agonistas Adrenérgicos beta/farmacología , Animales , Trasplante de Células , Fluorodesoxiglucosa F18 , Perfilación de la Expresión Génica , Humanos , Isoproterenol/farmacología , Ratones , Mitocondrias/metabolismo , Consumo de Oxígeno , ARN Mensajero/metabolismo , Radiofármacos , Reacción en Cadena en Tiempo Real de la Polimerasa , Regulación hacia Arriba
13.
Nat Commun ; 6: 6749, 2015 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-25851587

RESUMEN

Short- and long-scales intra- and inter-chromosomal interactions are linked to gene transcription, but the molecular events underlying these structures and how they affect cell fate decision during embryonic development are poorly understood. One of the first embryonic cell fate decisions (that is, mesendoderm determination) is driven by the POU factor OCT4, acting in concert with the high-mobility group genes Sox-2 and Sox-17. Here we report a chromatin-remodelling mechanism and enhancer function that mediate cell fate switching. OCT4 alters the higher-order chromatin structure at both Sox-2 and Sox-17 loci. OCT4 titrates out cohesin and switches the Sox-17 enhancer from a locked (within an inter-chromosomal Sox-2 enhancer/CCCTC-binding factor CTCF/cohesin loop) to an active (within an intra-chromosomal Sox-17 promoter/enhancer/cohesin loop) state. SALL4 concomitantly mobilizes the polycomb complexes at the Soxs loci. Thus, OCT4/SALL4-driven cohesin- and polycombs-mediated changes in higher-order chromatin structure mediate instruction of early cell fate in embryonic cells.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Embrión de Mamíferos/metabolismo , Corazón/embriología , Células Madre Embrionarias Humanas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Factores de Transcripción SOX/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factor de Unión a CCCTC , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Proteínas Fluorescentes Verdes , Proteínas HMGB/genética , Proteínas HMGB/metabolismo , Humanos , Ratones , Proteínas de Neoplasias , Células Madre Pluripotentes , Complejo Represivo Polycomb 1/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Proteínas del Grupo Polycomb/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Represoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción SOXB1/metabolismo , Factores de Transcripción SOXF/genética , Factores de Transcripción SOXF/metabolismo , Factores de Transcripción/metabolismo , Cohesinas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA