Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-38899472

RESUMEN

BACKGROUND: Pathogenic variants in PLIN1-encoding PLIN1 (perilipin-1) are responsible for an autosomal dominant form of familial partial lipodystrophy (FPL) associated with severe insulin resistance, hepatic steatosis, and important hypertriglyceridemia. This study aims to decipher the mechanisms of hypertriglyceridemia associated with PLIN1-related FPL. METHODS: We performed an in vivo lipoprotein kinetic study in 6 affected patients compared with 13 healthy controls and 8 patients with type 2 diabetes. Glucose and lipid parameters, including plasma LPL (lipoprotein lipase) mass, were measured. LPL mRNA and protein expression were evaluated in abdominal subcutaneous adipose tissue from patients with 5 PLIN1-mutated FPL and 3 controls. RESULTS: Patients with PLIN1-mutated FPL presented with decreased fat mass, insulin resistance, and diabetes (glycated hemoglobin A1c, 6.68±0.70% versus 7.48±1.63% in patients with type 2 diabetes; mean±SD; P=0.27). Their plasma triglycerides were higher (5.96±3.08 mmol/L) than in controls (0.76±0.27 mmol/L; P<0.0001) and patients with type 2 diabetes (2.94±1.46 mmol/L, P=0.006). Compared with controls, patients with PLIN1-related FPL had a significant reduction of the indirect fractional catabolic rate of VLDL (very-low-density lipoprotein)-apoB100 toward IDL (intermediate-density lipoprotein)/LDL (low-density lipoprotein; 1.79±1.38 versus 5.34±2.45 pool/d; P=0.003) and the indirect fractional catabolic rate of IDL-apoB100 toward LDL (2.14±1.44 versus 7.51±4.07 pool/d; P=0.005). VLDL-apoB100 production was not different between patients with PLIN1-related FPL and controls. Compared with patients with type 2 diabetes, patients with PLIN1-related FPL also showed a significant reduction of the catabolism of both VLDL-apoB100 (P=0.031) and IDL-apoB100 (P=0.031). Plasma LPL mass was significantly lower in patients with PLIN1-related FPL than in controls (21.03±10.08 versus 55.76±13.10 ng/mL; P<0.0001), although the LPL protein expression in adipose tissue was similar. VLDL-apoB100 and IDL-apoB100 indirect fractional catabolic rates were negatively correlated with plasma triglycerides and positively correlated with LPL mass. CONCLUSIONS: We show that hypertriglyceridemia associated with PLIN1-related FPL results from a marked decrease in the catabolism of triglyceride-rich lipoproteins (VLDL and IDL). This could be due to a pronounced reduction in LPL availability, related to the decreased adipose tissue mass.

2.
Am J Physiol Endocrinol Metab ; 320(2): E219-E233, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33252251

RESUMEN

Beiging is an attractive therapeutic strategy to fight against obesity and its side metabolic complications. The loss of function of the nuclear transcription factor RORα has been related to a lean phenotype with higher thermogenesis in sg/sg mice lacking this protein. Here we show that pharmacological modulation of RORα activity exerts reciprocal and cell-autonomous effect on UCP1 expression ex vivo, in cellulo, and in vivo. The RORα inverse-agonist SR3335 upregulated UCP1 expression in brown and subcutaneous white adipose tissue (scWAT) explants of wild-type (WT) mice, whereas the RORα agonist SR1078 had the opposite effect. We confirmed the reciprocal action of these synthetic RORα ligands on gene expression, mitochondrial mass, and uncoupled oxygen consumption rate in cultured murine and human adipocytes. Time course analysis revealed stepwise variation in gene expression, first of TLE3, an inhibitor of the thermogenic program, followed by a reciprocal effect on PRDM16 and UCP1. Finally, RORα ligands were shown to be useful tools to modulate in vivo UCP1 expression in scWAT with associated changes in this fat depot mass. SR3335 and SR1078 provoked the opposite effects on the WT mice body weight, but without any effect on sg/sg mice. This slimming effect of SR3335 was related to an increased adaptive thermogenesis of the mice, as assessed by the rectal temperature of cold-stressed mice and induction of UCP1 in scWAT, as well as by indirect calorimetry in presence or not of a ß3-adrenoceptor agonist. These data confirmed that RORα ligands could be useful tools to modulate thermogenesis and energy homeostasis.NEW & NOTEWORTHY The regulation of adipose tissue browning was not fully deciphered and required further studies explaining how the regulation of this process may be of interest for tackling obesity and related metabolic disorders. Our data confirmed the involvement of the transcription factor RORα in the regulation of nonshivering thermogenesis, and importantly, revealed the possibility to in vivo modulate its activity by synthetic ligands with beneficial consequences on fat mass and body weight of the mice.


Asunto(s)
Tejido Adiposo Pardo/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/agonistas , Sulfonamidas/farmacología , Termogénesis/efectos de los fármacos , Tiofenos/farmacología , Adipocitos/efectos de los fármacos , Adipocitos/fisiología , Tejido Adiposo Pardo/fisiología , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/fisiología , Adulto , Animales , Benzamidas/farmacología , Transdiferenciación Celular/efectos de los fármacos , Células Cultivadas , Respuesta al Choque por Frío/efectos de los fármacos , Respuesta al Choque por Frío/fisiología , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/fisiología , Tiazoles/farmacología
3.
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
4.
J Antimicrob Chemother ; 71(11): 3212-3221, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27516476

RESUMEN

OBJECTIVES: Use of ART containing HIV PIs has previously been associated with toxicity in subcutaneous adipose tissue (SAT), potentially contributing to the development of lipodystrophy and insulin resistance. However, the effect of PIs on SAT function in ART-naive patients independent of other ART classes is unknown. This study aimed to elucidate the effect of initiating PI-only ART on SAT function in ART-naive subjects. METHODS: In the HIVNAT-019 study, 48 HIV-infected, ART-naive Thai adults commencing PI-only ART comprising lopinavir/ritonavir/saquinavir for 24 weeks underwent assessments of fasting metabolic parameters and body composition. In a molecular substudy, 20 subjects underwent SAT biopsies at weeks 0, 2 and 24 for transcriptional, protein, mitochondrial DNA (mtDNA) and histological analyses. ClinicalTrials.gov registration number: NCT00400738. RESULTS: Over 24 weeks, limb fat increased (+416.4 g, P = 0.023), coinciding with larger adipocytes as indicated by decreased adipocyte density in biopsies (-32.3 cells/mm2, P = 0.047) and increased mRNA expression of adipogenesis regulator PPARG at week 2 (+58.1%, P = 0.003). Increases in mtDNA over 24 weeks (+600 copies/cell, P = 0.041), decreased NRF1 mRNA expression at week 2 (-33.7%, P < 0.001) and increased COX2/COX4 protein ratio at week 24 (+288%, P = 0.038) indicated improved mitochondrial function. Despite decreased AKT2 mRNA at week 2 (-28.6%, P = 0.002) and increased PTPN1 mRNA at week 24 (+50.3%, P = 0.016) suggesting insulin resistance, clinical insulin sensitivity [by homeostasis model assessment (HOMA-IR)] was unchanged. CONCLUSIONS: Initiation of PI-only ART showed little evidence of SAT toxicity, the changes observed being consistent with a return to health rather than contributing to lipodystrophy.


Asunto(s)
Infecciones por VIH/tratamiento farmacológico , Inhibidores de la Proteasa del VIH/efectos adversos , Inhibidores de la Proteasa del VIH/uso terapéutico , Grasa Subcutánea/efectos de los fármacos , Grasa Subcutánea/fisiología , Adulto , Biopsia , ADN Mitocondrial/análisis , Femenino , Perfilación de la Expresión Génica , Histocitoquímica , Humanos , Masculino , Proteoma/análisis , Tailandia
5.
Arterioscler Thromb Vasc Biol ; 33(4): 829-38, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23393388

RESUMEN

OBJECTIVE: Inactivating peroxisome proliferator-activated receptor-γ (PPARγ) mutations lead to a syndrome of familial partial lipodystrophy (FPLD3) associated with early-onset severe hypertension. PPARγ can repress the vascular renin-angiotensin system (RAS) and angiotensin II receptor 1 expression. We evaluated the relationships between PPARγ inactivation and cellular RAS using FPLD3 patients' cells and human vascular smooth muscle cells expressing mutant or wild-type PPARγ. Approach and Results- We identified 2 novel PPARG mutations, R165T and L339X, located in the DNA and ligand-binding domains of PPARγ, respectively in 4 patients from 2 FPLD3 families. In cultured skin fibroblasts and peripheral blood mononuclear cells from the 4 patients and healthy controls, we compared markers of RAS activation, oxidative stress, and inflammation, and tested the effect of modulators of PPARγ and angiotensin II receptor 1. We studied the impact of the 2 mutations on the transcriptional activity of PPARγ and on the vascular RAS in transfected human vascular smooth muscle cells. Systemic RAS was not altered in patients. However, RAS markers were overexpressed in patients' fibroblasts and peripheral blood mononuclear cells, as in vascular cells expressing mutant PPARγ. Angiotensin II-mediated mitogen-activated protein kinase activity increased in patients' fibroblasts, consistent with RAS constitutive activation. Patients' cells also displayed oxidative stress and inflammation. PPARγ activation and angiotensin II receptor 1 mRNA silencing reversed RAS overactivation, oxidative stress, and inflammation, arguing for a role of angiotensin II receptor 1 in these processes. CONCLUSIONS: Two novel FPLD3-linked PPARG mutations are associated with a defective transrepression of cellular RAS leading to cellular dysfunction, which might contribute to the specific FPLD3-linked severe hypertension.


Asunto(s)
Hipertensión/genética , Lipodistrofia Parcial Familiar/genética , Mutación , PPAR gamma/genética , Sistema Renina-Angiotensina , Adulto , Anciano , Secuencia de Aminoácidos , Angiotensina II/metabolismo , Anilidas/farmacología , Animales , Estudios de Casos y Controles , Activación Enzimática , Femenino , Fibroblastos/metabolismo , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Hipertensión/metabolismo , Hipertensión/fisiopatología , Mediadores de Inflamación/metabolismo , Leucocitos Mononucleares/metabolismo , Lipodistrofia Parcial Familiar/metabolismo , Lipodistrofia Parcial Familiar/fisiopatología , Masculino , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Datos de Secuencia Molecular , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Estrés Oxidativo , PPAR gamma/efectos de los fármacos , PPAR gamma/metabolismo , Fenotipo , Interferencia de ARN , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo , Sistema Renina-Angiotensina/genética , Rosiglitazona , Índice de Severidad de la Enfermedad , Piel/metabolismo , Tiazolidinedionas/farmacología , Transfección , Adulto Joven
6.
Eur J Endocrinol ; 190(2): 151-164, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38245004

RESUMEN

OBJECTIVE: SOFT syndrome (MIM#614813), denoting Short stature, Onychodysplasia, Facial dysmorphism, and hypoTrichosis, is a rare primordial dwarfism syndrome caused by biallelic variants in POC1A, encoding a centriolar protein. SOFT syndrome, characterized by severe growth failure of prenatal onset and dysmorphic features, was recently associated with insulin resistance. This study aims to further explore its endocrinological features and pathophysiological mechanisms. DESIGN/METHODS: We present clinical, biochemical, and genetic features of 2 unrelated patients carrying biallelic pathogenic POC1A variants. Cellular models of the disease were generated using patients' fibroblasts and POC1A-deleted human adipose stem cells. RESULTS: Both patients present with clinical features of SOFT syndrome, along with hyperinsulinemia, diabetes or glucose intolerance, hypertriglyceridemia, liver steatosis, and central fat distribution. They also display resistance to the effects of IGF-1. Cellular studies show that the lack of POC1A protein expression impairs ciliogenesis and adipocyte differentiation, induces cellular senescence, and leads to resistance to insulin and IGF-1. An altered subcellular localization of insulin receptors and, to a lesser extent, IGF1 receptors could also contribute to resistance to insulin and IGF1. CONCLUSIONS: Severe growth retardation, IGF-1 resistance, and centripetal fat repartition associated with insulin resistance-related metabolic abnormalities should be considered as typical features of SOFT syndrome caused by biallelic POC1A null variants. Adipocyte dysfunction and cellular senescence likely contribute to the metabolic consequences of POC1A deficiency. SOFT syndrome should be included within the group of monogenic ciliopathies with metabolic and adipose tissue involvement, which already encompasses Bardet-Biedl and Alström syndromes.


Asunto(s)
Anomalías Múltiples , Ciliopatías , Resistencia a la Insulina , Insulinas , Humanos , Proteínas de Ciclo Celular/genética , Proteínas del Citoesqueleto/genética , Factor I del Crecimiento Similar a la Insulina , Resistencia a la Insulina/genética , Ciliopatías/genética , Anomalías Múltiples/genética
7.
Eur J Endocrinol ; 185(6): 841-854, 2021 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-34643546

RESUMEN

OBJECTIVE: CAV1 encodes caveolin-1, a major protein of plasma membrane microdomains called caveolae, involved in several signaling pathways. Caveolin-1 is also located at the adipocyte lipid droplet. Heterozygous pathogenic variants of CAV1 induce rare heterogeneous disorders including pulmonary arterial hypertension and neonatal progeroid syndrome. Only one patient was previously reported with a CAV1 homozygous pathogenic variant, associated with congenital generalized lipodystrophy (CGL3). We aimed to further delineate genetic transmission, clinical, metabolic, and cellular characteristics of CGL3. DESIGN/METHODS: In a large consanguineous kindred referred for CGL, we performed next-generation sequencing, as well as clinical, imagery, and metabolic investigations. We studied skin fibroblasts from the index case and the previously reported patient with CGL3. RESULTS: Four patients, aged 8 months to 18 years, carried a new homozygous p.(His79Glnfs*3) CAV1 variant. They all displayed generalized lipodystrophy since infancy, insulin resistance, low HDL-cholesterol, and/or high triglycerides, but no pulmonary hypertension. Two patients also presented at the age of 15 and 18 years with dysphagia due to achalasia, and one patient had retinitis pigmentosa. Heterozygous parents and relatives (n = 9) were asymptomatic, without any metabolic abnormality. Patients' fibroblasts showed a complete loss of caveolae and no protein expression of caveolin-1 and its caveolin-2 and cavin-1 partners. Patients' fibroblasts also displayed insulin resistance, increased oxidative stress, and premature senescence. CONCLUSIONS: The CAV1 null variant investigated herein leads to an autosomal recessive congenital lipodystrophy syndrome. Loss of caveolin-1 and/or caveolae induces specific manifestations including achalasia which requires specific management. Overlapping phenotypic traits between the different CAV1-related diseases require further studies.


Asunto(s)
Caveolina 1/genética , Acalasia del Esófago/genética , Lipodistrofia Generalizada Congénita/genética , Adolescente , Caveolas/patología , Caveolas/ultraestructura , Caveolina 1/metabolismo , Caveolina 2/metabolismo , Senescencia Celular , Niño , Preescolar , Consanguinidad , Dislipidemias/metabolismo , Acalasia del Esófago/patología , Femenino , Fibroblastos/patología , Fibroblastos/ultraestructura , Homocigoto , Humanos , Lactante , Lipodistrofia Generalizada Congénita/metabolismo , Lipodistrofia Generalizada Congénita/patología , Masculino , Microscopía Electrónica de Transmisión , Estrés Oxidativo , Linaje , Proteínas de Unión al ARN/metabolismo
8.
Front Endocrinol (Lausanne) ; 12: 803189, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35046902

RESUMEN

Lipodystrophy syndromes are rare diseases originating from a generalized or partial loss of adipose tissue. Adipose tissue dysfunction results from heterogeneous genetic or acquired causes, but leads to similar metabolic complications with insulin resistance, diabetes, hypertriglyceridemia, nonalcoholic fatty liver disease, dysfunctions of the gonadotropic axis and endocrine defects of adipose tissue with leptin and adiponectin deficiency. Diagnosis, based on clinical and metabolic investigations, and on genetic analyses, is of major importance to adapt medical care and genetic counseling. Molecular and cellular bases of these syndromes involve, among others, altered adipocyte differentiation, structure and/or regulation of the adipocyte lipid droplet, and/or premature cellular senescence. Lipodystrophy syndromes frequently present as systemic diseases with multi-tissue involvement. After an update on the main molecular bases and clinical forms of lipodystrophy, we will focus on topics that have recently emerged in the field. We will discuss the links between lipodystrophy and premature ageing and/or immuno-inflammatory aggressions of adipose tissue, as well as the relationships between lipomatosis and lipodystrophy. Finally, the indications of substitutive therapy with metreleptin, an analog of leptin, which is approved in Europe and USA, will be discussed.


Asunto(s)
Lipodistrofia , Adipocitos/fisiología , Tejido Adiposo/metabolismo , Envejecimiento Prematuro , Humanos , Inflamación/complicaciones , Resistencia a la Insulina , Leptina/análogos & derivados , Leptina/uso terapéutico , Lipodistrofia/tratamiento farmacológico , Lipodistrofia/etiología , Lipodistrofia/metabolismo , Lipodistrofia/patología , Lipomatosis/fisiopatología , Síndrome
9.
AIDS ; 35(10): 1625-1630, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-33831906

RESUMEN

OBJECTIVE: To evaluate the effect on anthropometric, metabolic and adipose tissue parameters of switching ART-controlled persons living with HIV (PLWH) from a protease inhibitor regimen to raltegravir/maraviroc. DESIGN: Sub-study of the ANRS157 ROCnRAL study with the investigation of subcutaneous abdominal adipose tissue (SCAT) biopsy at inclusion and study end. METHODS: We performed lipoaspiration of paired SCAT samples, histology on fresh/fixed samples and examined the transcriptomic profile analyzed using Illumina microarrays after RNA extraction. Statistical analyses used the Wilcoxon-paired test. RESULTS: The patients (n = 8) were mainly male (7/8), aged (mean ±â€Šstandard error of the mean) 54.9 ±â€Š1.2 years, BMI 26.1 ±â€Š1.2 kg/m2, CD4+ 699 ±â€Š56 cells/mm3, all viral load (VL) <50 copies/ml. After a follow-up of 6 ±â€Š0.5 months, all PLWH remained with VL <50 copies/ml. BMI, trunk and limb fat amounts were unchanged yet systemic insulin resistance increased. Adipose tissue histology was unchanged except for borderline increased adipocyte diameter (P = 0.1). Among the 16 094 RNA transcripts, 458 genes were up-regulated and 244 were down-regulated. Analyses of the Kyoto Encyclopedia of Genes and Genomes and Gene Ontology databases, evaluating modifications in the main functional pathways, revealed that genes related to immune recognition/function were less expressed as were genes encoding T-cell receptor and receptor signaling pathways. The gene expression profiles indicated decreased inflammation but genes involved in adipogenesis and insulin resistance were overexpressed. CONCLUSION: After 6 months of raltegravir/maraviroc, adipogenesis-related gene profile was enhanced in SCAT, in agreement with a tendency for increased adipocyte size. Enhanced SCAT insulin resistance-related profile was concordant with higher systemic insulin resistance. However, the immune activation/inflammation profile was globally lowered. We propose that raltegravir/maraviroc might favor SCAT gain but reduce inflammation/immune activation.


Asunto(s)
Fármacos Anti-VIH , Infecciones por VIH , Tejido Adiposo , Fármacos Anti-VIH/uso terapéutico , Infecciones por VIH/tratamiento farmacológico , Humanos , Masculino , Maraviroc , Raltegravir Potásico/uso terapéutico , Grasa Subcutánea
10.
Eur J Endocrinol ; 184(1): 155-168, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33112291

RESUMEN

OBJECTIVE: The term Multiple Symmetric Lipomatosis (MSL) describes a heterogeneous group of rare monogenic disorders and multifactorial conditions, characterized by upper-body adipose masses. Biallelic variants in LIPE encoding hormone-sensitive lipase (HSL), a key lipolytic enzyme, were implicated in three families worldwide. We aimed to further delineate LIPE-related clinical features and pathophysiological determinants. METHODS: A gene panel was used to identify pathogenic variants. The disease features were reviewed at the French lipodystrophy reference center. The immunohistological, ultrastructural, and protein expression characteristics of lipomatous tissue were determined in surgical samples from one patient. The functional impact of variants was investigated by developing a model of adipose stem cells (ASCs) isolated from lipomatous tissue. RESULTS: We identified new biallelic LIPE null variants in three unrelated patients referred for MSL and/or partial lipodystrophy. The hallmarks of the disease, appearing in adulthood, included lower-limb lipoatrophy, upper-body and abdominal pseudo-lipomatous masses, diabetes and/or insulin resistance, hypertriglyceridemia, liver steatosis, high blood pressure, and neuromuscular manifestations. Ophthalmological investigations revealed numerous auto-fluorescent drusen-like retinal deposits in all patients. Lipomatous tissue and patient ASCs showed loss of HSL and decreased expression of adipogenic and mature adipocyte markers. LIPE-mutated ASCs displayed impaired adipocyte differentiation, decreased insulin response, defective lipolysis, and mitochondrial dysfunction. CONSLUSIONS: Biallelic LIPE null variants result in a multisystemic disease requiring multidisciplinary care. Loss of HSL expression impairs adipocyte differentiation, consistent with the lipodystrophy/MSL phenotype and associated metabolic complications. Detailed ophthalmological examination could reveal retinal damage, further pointing to the nervous tissue as an important disease target.


Asunto(s)
Diferenciación Celular/genética , Lipodistrofia/genética , Lipomatosis Simétrica Múltiple/genética , Modelos Genéticos , Esterol Esterasa/genética , Adipocitos/fisiología , Tejido Adiposo/citología , Anciano , Alelos , Femenino , Variación Genética , Humanos , Persona de Mediana Edad , Fenotipo , Células Madre/fisiología , Síndrome
11.
Elife ; 102021 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-34342583

RESUMEN

Epoxide hydrolases (EHs) regulate cellular homeostasis through hydrolysis of epoxides to less-reactive diols. The first discovered EH was EPHX1, also known as mEH. EH functions remain partly unknown, and no pathogenic variants have been reported in humans. We identified two de novo variants located in EPHX1 catalytic site in patients with a lipoatrophic diabetes characterized by loss of adipose tissue, insulin resistance, and multiple organ dysfunction. Functional analyses revealed that these variants led to the protein aggregation within the endoplasmic reticulum and to a loss of its hydrolysis activity. CRISPR-Cas9-mediated EPHX1 knockout (KO) abolished adipocyte differentiation and decreased insulin response. This KO also promoted oxidative stress and cellular senescence, an observation confirmed in patient-derived fibroblasts. Metreleptin therapy had a beneficial effect in one patient. This translational study highlights the importance of epoxide regulation for adipocyte function and provides new insights into the physiological roles of EHs in humans.


Asunto(s)
Senescencia Celular/genética , Diabetes Mellitus Lipoatrófica/genética , Diabetes Mellitus Lipoatrófica/fisiopatología , Epóxido Hidrolasas/genética , Compuestos Epoxi/metabolismo , Adolescente , Adulto , Epóxido Hidrolasas/metabolismo , Femenino , Regulación Enzimológica de la Expresión Génica , Humanos , Hidrólisis , Mutación
12.
Ann Endocrinol (Paris) ; 81(1): 51-60, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31982105

RESUMEN

Lipodystrophic syndromes are acquired or genetic rare diseases, characterised by a generalised or partial lack of adipose tissue leading to metabolic alterations linked to strong insulin resistance. They encompass a variety of clinical entities due to primary defects in adipose differentiation, in the structure and/or regulation of the adipocyte lipid droplet, or due to immune-inflammatory aggressions, chromatin deregulations and/or mitochondrial dysfunctions affecting adipose tissue. Diagnosis is based on clinical examination, pathological context and comorbidities, and on results of metabolic investigations and genetic analyses, which together determine management and genetic counselling. Early lifestyle and dietary measures focusing on regular physical activity and avoiding excess energy intake are crucial. They are accompanied by multidisciplinary follow-up adapted to each clinical form. In case of hyperglycemia, antidiabetic medications, with metformin as a first-line therapy in adults, are used in addition to lifestyle and dietary modifications. When standard treatments have failed to control metabolic disorders, the orphan drug metreleptin, an analog of leptin, can be effective in certain forms of lipodystrophy syndrome. Metreleptin therapy indications, prescription and monitoring were recently defined in France, representing a major improvement in patient care.


Asunto(s)
Lipodistrofia/diagnóstico , Lipodistrofia/terapia , Adulto , Comorbilidad , Técnicas de Diagnóstico Endocrino , Endocrinología/métodos , Endocrinología/tendencias , Predisposición Genética a la Enfermedad , Humanos , Lipodistrofia/epidemiología , Lipodistrofia/genética , Síndrome
13.
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
14.
J Clin Lipidol ; 12(6): 1420-1435, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30158064

RESUMEN

BACKGROUND: Multiple symmetric lipomatosis (MSL) is characterized by upper-body lipomatous masses frequently associated with metabolic and neurological signs. MFN2 pathogenic variants were recently implicated in a very rare autosomal recessive form of MSL. MFN2 encodes mitofusin-2, a mitochondrial fusion protein previously involved in Charcot-Marie-Tooth neuropathy. OBJECTIVE: To investigate the clinical, metabolic, tissular, and molecular characteristics of MFN2-associated MSL. METHODS: We sequenced MFN2 in 66 patients referred for altered fat distribution with one or several lipomas or lipoma-like regions and performed clinical and metabolic investigations in patients with positive genetic testing. Lipomatous tissues were studied in 3 patients. RESULTS: Six patients from 5 families carried a homozygous p.Arg707Trp pathogenic variant, representing the largest reported series of MFN2-associated MSL. Patients presented both lipomatous masses and a lipodystrophic syndrome (lipoatrophy, low leptinemia and adiponectinemia, hypertriglyceridemia, insulin resistance and/or diabetes). Charcot-Marie-Tooth neuropathy was of highly variable clinical severity. Lipomatous tissue mainly contained hyperplastic unilocular adipocytes, with few multilocular cells. It displayed numerous mitochondrial alterations (increased number and size, structural defects). As compared to control subcutaneous fat, mRNA and protein expression of leptin and adiponectin was strikingly decreased, whereas the CITED1 and fibroblast growth factor 21 (FGF21) thermogenic markers were strongly overexpressed. Consistently, serum FGF21 was markedly increased, and 18F-FDG-PET-scan revealed increased fat metabolic activity. CONCLUSION: MFN2-related MSL is a novel mitochondrial lipodystrophic syndrome involving both lipomatous masses and lipoatrophy. Its complex neurological and metabolic phenotype justifies careful clinical evaluation and multidisciplinary care. Low leptinemia and adiponectinemia, high serum FGF21, and increased 18F-FDG body fat uptake may be disease markers.


Asunto(s)
Tejido Adiposo/patología , GTP Fosfohidrolasas/metabolismo , Lipomatosis Simétrica Múltiple/metabolismo , Lipomatosis Simétrica Múltiple/patología , Proteínas Mitocondriales/metabolismo , Adulto , Anciano , Femenino , GTP Fosfohidrolasas/genética , Humanos , Lipomatosis Simétrica Múltiple/diagnóstico por imagen , Lipomatosis Simétrica Múltiple/genética , Masculino , Persona de Mediana Edad , Proteínas Mitocondriales/genética , Tomografía de Emisión de Positrones , ARN Mensajero/genética , ARN Mensajero/metabolismo
15.
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
16.
Antivir Ther ; 19(8): 773-82, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24535489

RESUMEN

BACKGROUND: HIV-infected patients administered long-term ritonavir-boosted protease inhibitors (PIs) are at a greater risk for developing cardiovascular diseases. Endothelial dysfunction is an initiating event in HIV-associated atherosclerosis. Cultured endothelial cells can be used as a model to compare the endothelial toxicity of different PIs. METHODS: We compared the effect of darunavir (DRV), darunavir/ritonavir (DRV/r), lopinavir/ritonavir (LPV/r) and atazanavir/ritonavir (ATV/r), used at clinically relevant concentrations, on human coronary artery endothelial cell vascular function, oxidative stress, inflammation and senescence, and studied the effect of pravastatin on PI-induced alterations. RESULTS: Vascular endothelial cell function, evaluated by the expression of endothelial nitric oxide synthase and the production of nitric oxide and endothelin-1, was unaffected by DRV or DRV/r, but altered by LPV/r or ATV/r. DRV or DRV/r did not alter, or mildly induced oxidative stress and inflammation (phosphorylation of p65/RelA-NFκB, secretion of IL-6 and IL-8), while ATV/r and LPV/r induced a marked increase. Secretion of sICAM or sVCAM, indicative of altered cell integrity, was not or weakly altered by DRV or DRV/r, but increased by 2-3-fold by LPV/r or ATV/r. Similar results were observed regarding senescence markers: SA-ß-galactosidase activation and overexpression of phospho-p53, p16(ink4), p21(WAF-1) and prelamin A. Pravastatin could, in part, reverse PI-induced adverse effects. CONCLUSIONS: Ritonavir-boosted PIs differentially induced vascular endothelial cell dysfunction, reactive oxygen species production, inflammation and senescence with no effect or a mild effect of DRV/r, an intermediate effect of ATV/r, and a stronger effect of LPV/r. Statins could, in part, protect the cells from PI-induced endothelial dysfunction.


Asunto(s)
Fármacos Anti-VIH/farmacología , Células Endoteliales/efectos de los fármacos , Lopinavir/farmacología , Oligopéptidos/farmacología , Piridinas/farmacología , Ritonavir/farmacología , Sulfonamidas/farmacología , Sulfato de Atazanavir , Células Cultivadas , Senescencia Celular/efectos de los fármacos , Darunavir , Sinergismo Farmacológico , Células Endoteliales/metabolismo , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Óxido Nítrico/metabolismo , Estrés Oxidativo/efectos de los fármacos , Pravastatina/farmacología
17.
Antivir Ther ; 17(3): 549-56, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22293506

RESUMEN

BACKGROUND: Ritonavir-boosted protease inhibitors (PIs) could adversely affect metabolism and adipose tissue to different extents, depending on the molecule. Using drugs with minimal adverse metabolic effects is an important consideration in at-risk HIV-infected patients. In vitro adipocyte models can be useful for comparing the effects of different PIs. METHODS: We compared the effects of darunavir, darunavir/ritonavir, atazanavir/ritonavir and lopinavir/ritonavir in murine and human adipocytes on differentiation, mitochondrial function, reactive oxygen species (ROS) production and insulin sensitivity. RESULTS: In human and murine adipocytes, differentiation evaluated by lipid content and protein expression of adipogenic markers, mitochondrial function evaluated by aggregation of the cationic dye JC-1 and by 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyltetrazolium bromide lysis, and mitochondrial mass evaluated by MitoTracker fluorescence and the expression of mitochondrial proteins were unaffected by darunavir, mildly affected by darunavir/ritonavir and further altered by atazanavir/ritonavir and lopinavir/ritonavir. ROS production was unaltered by darunavir and darunavir/ritonavir but was increased by lopinavir/ritonavir and atazanavir/ritonavir. Regarding insulin sensitivity, darunavir and darunavir/ritonavir had no significant effect on insulin activation of protein kinase B (Akt/PKB) and MAP kinase and of glucose transport, whereas lopinavir/ritonavir and atazanavir/ritonavir partly impaired the effect of insulin. The effect of atazanavir/ritonavir was generally milder than that of lopinavir/ritonavir. CONCLUSIONS: The various PIs differentially modified adipocyte functions. Darunavir alone did not affect adipocyte functions and only modestly altered differentiation and mitochondrial function when associated with ritonavir. Lopinavir/ritonavir adversely affected differentiation and lipid content, mitochondrial function, ROS production and insulin sensitivity, and the effect of atazanavir/ritonavir was intermediate. Thus, in vitro, darunavir/ritonavir presented a safer metabolic profile on adipocytes than atazanavir/ritonavir and lopinavir/ritonavir.


Asunto(s)
Adipocitos/citología , Adipocitos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Inhibidores de la Proteasa del VIH/farmacología , Resistencia a la Insulina , Ritonavir/farmacología , Adipocitos/metabolismo , Animales , Sulfato de Atazanavir , Células Cultivadas , Darunavir , Inhibidores de la Proteasa del VIH/efectos adversos , Humanos , Lipólisis/efectos de los fármacos , Lopinavir/efectos adversos , Lopinavir/farmacología , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Oligopéptidos/efectos adversos , Oligopéptidos/farmacología , Piridinas/efectos adversos , Piridinas/farmacología , Especies Reactivas de Oxígeno/metabolismo , Ritonavir/efectos adversos , Sulfonamidas/efectos adversos , Sulfonamidas/farmacología
18.
J Clin Endocrinol Metab ; 96(4): E653-7, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21270328

RESUMEN

CONTEXT: Type B insulin resistance syndrome is a rare condition characterized by the presence of autoantibodies directed against the insulin receptor. It has been reported in association with autoimmune diseases such as systemic lupus erythematosus. OBJECTIVE: We report a case of type B insulin resistance syndrome in a patient with HIV infection on highly active antiretroviral therapy (HAART). PATIENT AND METHODS: A 27-yr-old African woman with ketosis-prone diabetes and HIV infection developed severe insulin resistance after the initiation of HAART. Standard oral glucose tolerance tests using 75 g of glucose performed 1, 2, and 3 months after the initiation of HAART showed severe hyperinsulinemia and hypoglycemia. Six months later, she developed symptomatic hyperglycemia resistant to high-dose insulin therapy. To determine the cause of insulin resistance, we assayed the titer of insulin receptor autoantibodies in the serum of the patient. RESULTS: Plasma insulin receptor autoantibodies were present at the time of marked hyperglycemia and insulin resistance, confirming the diagnosis of type B insulin resistance syndrome. Simultaneously the diagnosis of immune reconstitution inflammatory syndrome was established according to increased CD4 T cell count, decreased plasma HIV1-RNA level, and tuberculosis reactivation, shortly after institution of HAART. Corticosteroid therapy improved insulin resistance and hyperglycemia. CONCLUSION: We report the first case of type B insulin resistance syndrome associated with immune reconstitution inflammatory syndrome in an HIV-infected patient.


Asunto(s)
Infecciones por VIH/complicaciones , Síndrome Inflamatorio de Reconstitución Inmune/complicaciones , Síndrome Metabólico/complicaciones , Adulto , Terapia Antirretroviral Altamente Activa/efectos adversos , Cetoacidosis Diabética/complicaciones , Cetoacidosis Diabética/inmunología , Femenino , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/inmunología , Humanos , Síndrome Metabólico/inducido químicamente , Síndrome Metabólico/clasificación , Síndrome Metabólico/inmunología , Receptor de Insulina/inmunología
19.
Bull Cancer ; 95(1): 121-32, 2008 Jan.
Artículo en Francés | MEDLINE | ID: mdl-18230578

RESUMEN

During each cell division, DNA polymerase makes mistakes while copying DNA. These errors, more frequent at the level of repeated sequences called microsatellites are normally repaired by a system called MMR (mismatch repair). Tumors defective in their MMR system accumulate mutations (deletions and insertions of some nucleotides) at the level of microsatellites and are called MSI (microsatellite instability). Microsatellites are numerous and scattered throughout the genome, in coding and non-coding regions. The instability of non-coding microsatellites is not known to have a major role in the process of cell transformation, but is a good indicator of the MSI status. On the other hand, instability by deletion or insertion in a coding region leads to a frameshift within the gene containing the repeat. The consequence is, the more often, the inactivation of this gene that potentially plays a role in initiation and/or MSI tumor progression. The MSI phenotype was first described in about 15 % of colorectal cancers that maybe of sporadic or hereditary (Lynch syndrome, or HNPCC for hereditary non-polyposis colorectal cancer) origin. It is also associated with about 15 % of gastric and endometrial tumors, and to a lesser extent with other human tumors. Besides a fundamental interest because of its original transformation mechanism, the analysis of MSI tumors is also important for clinical reasons. It was indeed shown that MSI tumors were associated with a better prognosis than non-MSI (also called MSS for microsatellite stable) tumors, and responded differently to conventional chemotherapeutic drugs used for the management of colorectal cancers. All these points will be discussed in details in the present review.


Asunto(s)
Reparación de la Incompatibilidad de ADN , Inestabilidad de Microsatélites , Neoplasias/genética , Disparidad de Par Base/genética , Inestabilidad Cromosómica/genética , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales Hereditarias sin Poliposis/tratamiento farmacológico , Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Islas de CpG/genética , Criopreservación/métodos , Metilación de ADN , Neoplasias Endometriales/tratamiento farmacológico , Neoplasias Endometriales/genética , Femenino , Humanos , Enfermedades Inflamatorias del Intestino/genética , Mutación/genética , Neoplasias/tratamiento farmacológico , Fenotipo , Pronóstico , Estabilidad del ARN , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética , Fijación del Tejido/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA