Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 23(2)2022 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-35054926

RESUMEN

Obesity prevalence is increasing worldwide, leading to cardiometabolic morbidities. Adipocyte dysfunction, impairing white adipose tissue (WAT) expandability and metabolic flexibility, is central in the development of obesity-related metabolic complications. Rare syndromes of lipodystrophy characterized by an extreme paucity of functional adipose tissue should be considered as primary adipocyte dysfunction diseases. Berardinelli-Seip congenital lipodystrophy (BSCL) is the most severe form with a near absence of WAT associated with cardiometabolic complications such as insulin resistance, liver steatosis, dyslipidemia, and cardiomyopathy. Twenty years ago, mutations in the BSCL2 gene have been identified as the cause of BSCL in human. BSCL2 encodes seipin, an endoplasmic reticulum (ER) anchored protein whose function was unknown back then. Studies of seipin knockout mice or rats demonstrated how seipin deficiency leads to severe lipodystrophy and to cardiometabolic complications. At the cellular levels, seipin is organized in multimers that are particularly enriched at ER/lipid droplet and ER/mitochondria contact sites. Seipin deficiency impairs both adipocyte differentiation and mature adipocyte maintenance. Experiments using adipose tissue transplantation in seipin knockout mice and tissue-specific deletion of seipin have provided a large body of evidence that liver steatosis, cardiomyopathy, and renal injury, classical diabetic complications, are all consequences of lipodystrophy. Rare adipocyte dysfunctions such as in BSCL are the key paradigm to unravel the pathways that control adipocyte homeostasis. The knowledge gathered through the study of these pathologies may bring new strategies to maintain and improve adipose tissue expandability.


Asunto(s)
Adipocitos/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/deficiencia , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Fenotipo , Adipogénesis , Tejido Adiposo/metabolismo , Animales , Complicaciones de la Diabetes , Modelos Animales de Enfermedad , Estudios de Asociación Genética/métodos , Humanos , Resistencia a la Insulina , Metabolismo de los Lípidos , Lipodistrofia/diagnóstico , Lipodistrofia/etiología , Lipodistrofia/metabolismo , Lipólisis , Roedores , Índice de Severidad de la Enfermedad
2.
EMBO J ; 35(24): 2699-2716, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27879284

RESUMEN

Seipin is an endoplasmic reticulum (ER) membrane protein implicated in lipid droplet (LD) biogenesis and mutated in severe congenital lipodystrophy (BSCL2). Here, we show that seipin is stably associated with nascent ER-LD contacts in human cells, typically via one mobile focal point per LD Seipin appears critical for such contacts since ER-LD contacts were completely missing or morphologically aberrant in seipin knockout and BSCL2 patient cells. In parallel, LD mobility was increased and protein delivery from the ER to LDs to promote LD growth was decreased. Moreover, while growing LDs normally acquire lipid and protein constituents from the ER, this process was compromised in seipin-deficient cells. In the absence of seipin, the initial synthesis of neutral lipids from exogenous fatty acid was normal, but fatty acid incorporation into neutral lipids in cells with pre-existing LDs was impaired. Together, our data suggest that seipin helps to connect newly formed LDs to the ER and that by stabilizing ER-LD contacts seipin facilitates the incorporation of protein and lipid cargo into growing LDs in human cells.


Asunto(s)
Retículo Endoplásmico/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Gotas Lipídicas/metabolismo , Células Cultivadas , Subunidades gamma de la Proteína de Unión al GTP/genética , Técnicas de Inactivación de Genes , Humanos , Modelos Biológicos
3.
Eur J Pediatr ; 179(10): 1653-1654, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32710302

RESUMEN

Although the patient has provided consent for publication of this case report and accompanying images, after publication of this article it has come to the authors' attention that Fig. 1 needs changes to better protect the privacy of the patient. A modified Fig. 1 is included in this Erratum. The original Fig. 1 has been removed to protect the patient's privacy.

4.
Eur J Pediatr ; 174(7): 975-80, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25994244

RESUMEN

UNLABELLED: Berardinelli-Seip congenital lipodystrophy (BSCL) is an uncommon autosomal recessive disorder. Patients with BSCL present with a distinct phenotype since subcutaneous fat is largely lacking and musculature has become more prominent. During childhood, diabetes and acanthosis nigricans evolve and female patients may develop hirsutism. Different genes encoding this entity have been described. Achalasia is a rare esophageal motility disorder, characterized by its distinct motility pattern with absent or incomplete lower esophageal sphincter (LES) relaxations. The exact cause of achalasia is yet unknown. Here, we describe a patient with achalasia in the context of BSCL, which might be linked by a shared pathophysiologic background, as evaluated in this case report. CONCLUSION: In a BSCL patient presenting with gastrointestinal symptoms, a motility disorder of the gastrointestinal tract should be considered. WHAT IS KNOWN: • Berardinelli-Seip congenital lipodystrophy (BSCL) and achalasia are both disorders characterized by low prevalence. What is New: • Co-existence of both diseases is described in this report. Linkage by a potential common pathophysiologic background is discussed in this paper.


Asunto(s)
Acalasia del Esófago/complicaciones , Lipodistrofia Generalizada Congénita/complicaciones , Adolescente , Codón sin Sentido , Acalasia del Esófago/diagnóstico , Acalasia del Esófago/genética , Femenino , Humanos , Lipodistrofia Generalizada Congénita/genética , Proteínas de Unión al ARN/genética
5.
N Engl J Med ; 364(8): 740-8, 2011 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-21345103

RESUMEN

Perilipin is the most abundant adipocyte-specific protein that coats lipid droplets, and it is required for optimal lipid incorporation and release from the droplet. We identified two heterozygous frameshift mutations in the perilipin gene (PLIN1) in three families with partial lipodystrophy, severe dyslipidemia, and insulin-resistant diabetes. Subcutaneous fat from the patients was characterized by smaller-than-normal adipocytes, macrophage infiltration, and fibrosis. In contrast to wild-type perilipin, mutant forms of the protein failed to increase triglyceride accumulation when expressed heterologously in preadipocytes. These findings define a novel dominant form of inherited lipodystrophy and highlight the serious metabolic consequences of a primary defect in the formation of lipid droplets in adipose tissue.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Mutación del Sistema de Lectura , Hipertrigliceridemia/genética , Lipodistrofia Parcial Familiar/genética , Fosfoproteínas/deficiencia , Fosfoproteínas/genética , Acantosis Nigricans/genética , Adulto , Proteínas Portadoras , Femenino , Genes Dominantes , Heterocigoto , Humanos , Resistencia a la Insulina/genética , Persona de Mediana Edad , Linaje , Perilipina-1
6.
Curr Atheroscler Rep ; 16(9): 437, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25047893

RESUMEN

Lipodystrophies are rare acquired and genetic disorders characterized by the selective loss of adipose tissue. One key metabolic feature of patients with congenital inherited lipodystrophy is hypertriglyceridemia. The precise mechanisms by which the lack of adipose tissue causes dyslipidemia remain largely unknown. In recent years, new insights have arisen from data obtained in vitro in adipocytes, yeast, drosophila, and very recently in several genetically modified mouse models of generalized lipodystrophy. A common metabolic pathway involving accelerated lipolysis and defective energy storage seems to contribute to the dyslipidemia associated with congenital generalized lipodystrophy syndromes, although the pathophysiological changes may vary with the nature of the mutation involved. Therapeutic management of dyslipidemia in patients with lipodystrophy is primarily based on specific approaches using recombinant leptin therapy. Preclinical studies suggest a potential efficacy of thiazolidinediones that remains to be assessed in dedicated clinical trials.


Asunto(s)
Dislipidemias/congénito , Predisposición Genética a la Enfermedad , Hipertrigliceridemia/congénito , Lipodistrofia/congénito , Lipodistrofia/diagnóstico , Tejido Adiposo/metabolismo , Animales , Dislipidemias/complicaciones , Dislipidemias/diagnóstico , Humanos , Hipertrigliceridemia/complicaciones , Lipodistrofia/metabolismo , Mutación/genética
7.
Front Endocrinol (Lausanne) ; 13: 785819, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35250856

RESUMEN

Lipodystrophies belong to the heterogenous group of syndromes in which the primary defect is a generalized or partial absence of adipose tissue, which may be congenital or acquired in origin. Lipodystrophy should be considered in patients manifesting the combination of insulin resistance (with or without overt diabetes), dyslipidemia and fatty liver. Lipodystrophies are classified according to the etiology of the disease (genetic or acquired) and to the anatomical distribution of adipose tissue (generalized or partial). The mechanism of adipose tissue loss is specific to each syndrome, depending on the biological function of the mutated gene. Mice models, together with cellular studies have permitted clarification of the mechanisms by which human mutations deeply compromise adipocyte homeostasis. In addition, rodent models have proven to be crucial in deciphering the cardiometabolic consequences of the lack of adipose tissue such as NAFLD, muscle insulin resistance and cardiomyopathy. More precisely, tissue-specific transgenic and knockout mice have brought new tools to distinguish phenotypic traits that are the consequences of lipodystrophy from those that are cell-autonomous. In this review, we discuss the mice models of lipodystrophy including those of inherited human syndromes of generalized and partial lipodystrophy. We present how these models have demonstrated the central role of white adipose tissue in energetic homeostasis in general, including insulin sensitivity and lipid handling in particular. We underscore the differences reported with the human phenotype and discuss the limit of rodent models in recapitulating adipose tissue primary default. Finally, we present how these mice models have highlighted the function of the causative-genes and brought new insights into the pathophysiology of the cardiometabolic complications associated with lipodystrophy.


Asunto(s)
Enfermedades Cardiovasculares , Resistencia a la Insulina , Lipodistrofia , Enfermedad del Hígado Graso no Alcohólico , Tejido Adiposo , Animales , Enfermedades Cardiovasculares/complicaciones , Modelos Animales de Enfermedad , Humanos , Resistencia a la Insulina/genética , Lipodistrofia/genética , Ratones , Ratones Noqueados , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Síndrome
8.
Cell Rep ; 38(2): 110213, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-35021082

RESUMEN

Deficiency of the endoplasmic reticulum (ER) protein seipin results in generalized lipodystrophy by incompletely understood mechanisms. Here, we report mitochondrial abnormalities in seipin-deficient patient cells. A subset of seipin is enriched at ER-mitochondria contact sites (MAMs) in human and mouse cells and localizes in the vicinity of calcium regulators SERCA2, IP3R, and VDAC. Seipin association with MAM calcium regulators is stimulated by fasting-like stimuli, while seipin association with lipid droplets is promoted by lipid loading. Acute seipin removal does not alter ER calcium stores but leads to defective mitochondrial calcium import accompanied by a widespread reduction in Krebs cycle metabolites and ATP levels. In mice, inducible seipin deletion leads to mitochondrial dysfunctions preceding the development of metabolic complications. Together, these data suggest that seipin controls mitochondrial energy metabolism by regulating mitochondrial calcium influx at MAMs. In seipin-deficient adipose tissue, reduced ATP production compromises adipocyte properties, contributing to lipodystrophy pathogenesis.


Asunto(s)
Adipocitos/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Mitocondrias/metabolismo , Tejido Adiposo/metabolismo , Animales , Calcio/metabolismo , Línea Celular , Retículo Endoplásmico/metabolismo , Estrés del Retículo Endoplásmico , Metabolismo Energético/fisiología , Subunidades gamma de la Proteína de Unión al GTP/deficiencia , Subunidades gamma de la Proteína de Unión al GTP/fisiología , Humanos , Gotas Lipídicas/metabolismo , Metabolismo de los Lípidos/fisiología , Lípidos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL
9.
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
10.
Endocr Dev ; 19: 1-20, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20551664

RESUMEN

Human lipodystrophies represent a heterogeneous group of diseases characterized by generalized or partial fat loss, with fat hypertrophy in other depots when partial. Insulin resistance, dyslipidemia and diabetes are generally associated, leading to early complications. Genetic forms are uncommon: recessive generalized congenital lipodystrophies result in most cases from mutations in the genes encoding seipin or the 1-acyl-glycerol-3-phosphate-acyltransferase 2(AGPAT2). Dominant partial familial lipodystrophies result from mutations in genes encoding the nuclear protein lamin A/C or the adipose transcription factor PPARgamma. Importantly, lamin A/Cmutations are also responsible for metabolic laminopathies, resembling the metabolic syndrome and progeria, a syndrome of premature aging. A number of lipodystrophic patients remain undiagnosed at the genetic level. Acquired lipodystrophy can be generalized, resembling congenital forms, or partial, as the Barraquer-Simons syndrome, with loss of fat in the upper part of the body contrasting with accumulation in the lower part. Although their etiology is generally unknown, they could be associated with signs of autoimmunity. The most common forms of lipodystrophies are iatrogenic. In human immunodeficiency virus-infected patients, some first-generation antiretroviral drugs were strongly related with peripheral lipoatrophy and metabolic alterations. Partial lipodystrophy also characterize patients with endogenous or exogenous long-term corticoid excess. Treatment of fat redistribution can sometimes benefit from plastic surgery. Lipid and glucose alterations are difficult to control leading to early occurrence of diabetic, cardiovascular and hepatic complications.


Asunto(s)
Tejido Adiposo/patología , Tejido Adiposo/fisiopatología , Lipodistrofia , 1-Acilglicerol-3-Fosfato O-Aciltransferasa/genética , Humanos , Lamina Tipo A/genética , Lipodistrofia/genética , Lipodistrofia/patología , Lipodistrofia/fisiopatología , PPAR gamma/genética
11.
J Clin Endocrinol Metab ; 93(4): 1129-34, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18211975

RESUMEN

CONTEXT: Berardinelli-Seip congenital lipodystrophy (BSCL) is a rare recessive disease characterized by near absence of adipose tissue, resulting in severe dyslipidemia and insulin resistance. In most reported cases, BSCL is due to alterations in either seipin, of unknown function, or 1-acylglycerol-3-phosphate acyltransferase-beta (AGPAT2), which catalyzes the formation of phosphatidic acid. OBJECTIVE: We sought to determine the genetic origin of the unexplained cases of BSCL. We thus sequenced CAV1, encoding caveolin-1, as a candidate gene involved in insulin signaling and lipid homeostasis. CAV1 is a key structural component of plasma membrane caveolae, and Cav1-deficient mice display progressive loss of adipose tissue and insulin resistance. DESIGN: We undertook phenotyping studies and molecular screening of CAV1 in four patients with BSCL with no mutation in the genes encoding either seipin or AGPAT2. RESULTS: A homozygous nonsense mutation (p.Glu38X) was identified in CAV1 in a patient with BSCL born from a consanguineous union. This mutation affects both the alpha- and beta-CAV1 isoforms and ablates CAV1 expression in skin fibroblasts. Detailed magnetic resonance imaging of the proband confirmed near total absence of both sc and visceral adipose tissue, with only vestigial amounts in the dorsal sc regions. In keeping with the lack of adipose tissue, the proband was also severely insulin resistant and dyslipidemic. In addition, the proband had mild hypocalcemia likely due to vitamin D resistance. CONCLUSIONS: These findings identify CAV1 as a new BSCL-related gene and support a critical role for caveolins in human adipocyte function.


Asunto(s)
Caveolina 1/genética , Codón sin Sentido , Lipodistrofia Generalizada Congénita/genética , Adipocitos/fisiología , Tejido Adiposo/metabolismo , Adulto , Caveolina 1/fisiología , Femenino , Humanos
12.
Clin Endocrinol (Oxf) ; 68(4): 547-554, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18076675

RESUMEN

AIMS/HYPOTHESIS: Partial lipodystrophy (PL) is most commonly characterized by loss of subcutaneous fat in the extremities with preservation of truncal fat and is associated with insulin resistance, diabetes and hyperlipidaemia. Recombinant human leptin (r-metHuLeptin) therapy has been shown to be effective in treating metabolic abnormalities associated with congenital or acquired generalized lipodystrophy and PL associated with lamin A/C (LMNA) gene mutations or highly active antiretroviral therapy (HAART). Our aim was to assess the effectiveness of leptin therapy in treating metabolic complications of PL associated with heterozygous peroxisome proliferator activated receptor gamma (PPARG) mutations. This is the first report to detail the clinical response of a patient with PL due to a PPARG mutation treated with r-metHuLeptin. METHODS: A 36-year-old female with PL associated with a heterozygous PPARG mutation complicated by poorly controlled diabetes and severe, refractory hypertriglyceridaemia was enrolled in a National Institutes of Health (NIH) protocol to evaluate the role of r-metHuLeptin in lipodystrophy. The patient received escalating doses of r-metHuLeptin until a dose 0.12 mg/kg/day was reached. Metabolic parameters, including serum chemistries, fasting blood glucose, glycated haemoglobin (HbA1c), lipid profile, an oral glucose tolerance test (OGTT), an insulin tolerance test (ITT), liver volume, percentage body fat and energy expenditure were followed at regular time intervals over 18 months of therapy. RESULTS: Eighteen months of r-MetHuLeptin therapy was associated with a marked improvement in glucose homeostasis as evidenced by normalization of the fasting blood glucose (baseline = 8.3 mmol/l; 18 months = 4.9 mmol/l), lowering of HbA1c (baseline = 9.9%; 18 months = 7.2%) and improved tolerance to an oral glucose load. In addition, a striking amelioration in the patient's refractory, severe hypertriglyceridaemia was observed (baseline = 21.15 mmol/l; 18 months = 5.96 mmol/l). CONCLUSION: r-MetHuLeptin is effective in treating metabolic complications associated with PL due to PPARG mutations. In the context of previously published work, our findings suggest that the response to r-MetHuLeptin is independent of the aetiology in lipodystrophy.


Asunto(s)
Leptina/análogos & derivados , Lipodistrofia/tratamiento farmacológico , Lipodistrofia/genética , PPAR gamma/genética , Adulto , Femenino , Humanos , Leptina/administración & dosificación , Resultado del Tratamiento
13.
Artículo en Inglés | MEDLINE | ID: mdl-30420836

RESUMEN

Type 2 diabetes (T2D) is a major risk factor for heart failure. Diabetic cardiomyopathy (DC) is characterized by diastolic dysfunction and left ventricular hypertrophy. Epidemiological data suggest that hyperglycaemia contributes to the development of DC. Several cellular pathways have been implicated in the deleterious effects of high glucose concentrations in the heart: oxidative stress, accumulation of advanced glycation end products (AGE), and chronic hexosamine biosynthetic pathway (HBP) activation. In the present review, we focus on the effect of chronic activation of the HBP on diabetic heart function. The HBP supplies N-acetylglucosamine moiety (O-GlcNAc) that is O-linked by O-GlcNAc transferase (OGT) to proteins on serine or threonine residues. This post-translational protein modification modulates the activity of the targeted proteins. In the heart, acute activation of the HBP in response to ischaemia-reperfusion injury appears to be protective. Conversely, chronic activation of the HBP in the diabetic heart affects Ca2+ handling, contractile properties, and mitochondrial function and promotes stress signaling, such as left ventricular hypertrophy and endoplasmic reticulum stress. Many studies have shown that O-GlcNAc impairs the function of key protein targets involved in these pathways, such as phospholamban, calmodulin kinase II, troponin I, and FOXO1. The data show that excessive O-GlcNAcylation is a major trigger of the glucotoxic events that affect heart function under chronic hyperglycaemia. Supporting this finding, pharmacological or genetic inhibition of the HBP in the diabetic heart improves heart function. In addition, the SGLT2 inhibitor dapagliflozin, a glucose lowering agent, has recently been shown to lower cardiac HBP in a lipodystophic T2D mice model and to concomitantly improve the diastolic dysfunction of these mice. Therefore, targeting cardiac-excessive O-GlcNAcylation or specific target proteins represents a potential therapeutic option to treat glucotoxicity in the diabetic heart.

14.
Diabetes ; 66(4): 1030-1040, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28052965

RESUMEN

Type 2 diabetes mellitus (T2DM) is a well-recognized independent risk factor for heart failure. T2DM is associated with altered cardiac energy metabolism, leading to ectopic lipid accumulation and glucose overload, the exact contribution of these two parameters remaining unclear. To provide new insight into the mechanism driving the development of diabetic cardiomyopathy, we studied a unique model of T2DM: lipodystrophic Bscl2-/- (seipin knockout [SKO]) mice. Echocardiography and cardiac magnetic resonance imaging revealed hypertrophic cardiomyopathy with left ventricular dysfunction in SKO mice, and these two abnormalities were strongly correlated with hyperglycemia. Surprisingly, neither intramyocardial lipid accumulation nor lipotoxic hallmarks were detected in SKO mice. [18F]Fludeoxyglucose positron emission tomography showed increased myocardial glucose uptake. Consistently, the O-GlcNAcylated protein levels were markedly increased in an SKO heart, suggesting a glucose overload. To test this hypothesis, we treated SKO mice with the hypoglycemic sodium-glucose cotransporter 2 (SGLT2) inhibitor dapagliflozin and the insulin sensitizer pioglitazone. Both treatments reduced the O-GlcNAcylated protein levels in SKO mice, and dapagliflozin successfully prevented the development of hypertrophic cardiomyopathy. Our data demonstrate that glucotoxicity by itself can trigger cardiac dysfunction and that a glucose-lowering agent can correct it. This result will contribute to better understanding of the potential cardiovascular benefits of SGLT2 inhibitors.


Asunto(s)
Compuestos de Bencidrilo/farmacología , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Cardiomiopatías Diabéticas , Glucósidos/farmacología , Corazón/efectos de los fármacos , Hipoglucemiantes/farmacología , Lipodistrofia , Tiazolidinedionas/farmacología , Función Ventricular/efectos de los fármacos , Animales , Compuestos de Bencidrilo/uso terapéutico , Glucemia/metabolismo , Cardiomiopatía Hipertrófica , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animales de Enfermedad , Ecocardiografía , Fluorodesoxiglucosa F18 , Subunidades gamma de la Proteína de Unión al GTP , Glucósidos/uso terapéutico , Corazón/diagnóstico por imagen , Proteínas de Unión al GTP Heterotriméricas/genética , Hiperglucemia , Hipoglucemiantes/uso terapéutico , Lipodistrofia/genética , Imagen por Resonancia Magnética , Ratones , Ratones Noqueados , Miocardio/metabolismo , Pioglitazona , Tomografía de Emisión de Positrones , Radiofármacos , Inhibidores del Cotransportador de Sodio-Glucosa 2 , Disfunción Ventricular Izquierda
15.
C R Biol ; 329(8): 639-52; discussion 653-5, 2006 Aug.
Artículo en Francés | MEDLINE | ID: mdl-16860281

RESUMEN

Lipodystrophic syndromes regroup a heterogeneous group of genetic or acquired diseases. Lipodystrophy, an altered development and/or repartition of body fat, is associated with alterations of lipid and glucose metabolism with insulin resistance. Genetic forms, rare, can be generalized and recessive resulting from mutations in the seipin or AGPAT2 gene. Partial lipodystrophies are dominant and observed in patients mutated in the gene encoding PPAR-gamma or lamin A/C, a gene seen also mutated in patients with syndromes of premature aging. Acquired forms are common and regroup the highly prevalent Metabolic Syndrome, hypercorticism together with lipodystrophy related to antiretroviral treatment of HIV-infected patients.


Asunto(s)
Tejido Adiposo/fisiopatología , Síndrome de Lipodistrofia Asociada a VIH/fisiopatología , Lipodistrofia/fisiopatología , Tejido Adiposo/patología , Animales , Modelos Animales de Enfermedad , Síndrome de Lipodistrofia Asociada a VIH/congénito , Humanos , Lipodistrofia/clasificación , Lipodistrofia/congénito , Lipodistrofia/genética , Ratones
16.
J Clin Endocrinol Metab ; 101(7): 2892-904, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27144934

RESUMEN

CONTEXT: Mutations in PTRF encoding cavin-1 are responsible for congenital generalized lipodystrophy type 4 (CGL4) characterized by lipoatrophy, insulin resistance, dyslipidemia, and muscular dystrophy. Cavin-1 cooperates with caveolins to form the plasma membrane caveolae, which are involved in cellular trafficking and signalling and in lipid turnover. OBJECTIVE: We sought to identify PTRF mutations in patients with CGL and to determine their impact on insulin sensitivity, adipose differentiation, and cellular autophagy. DESIGN AND PATIENTS: We performed phenotyping studies and molecular screening of PTRF in two unrelated families with CGL. Cellular studies were conducted in cultured skin fibroblasts from the two probands and from control subjects, and in murine 3T3-F442A preadipocytes. Knockdown of cavin-1 or ATG5 was obtained by small interfering RNA-mediated silencing. RESULTS: We identified two new PTRF homozygous mutations (p.Asp59Val or p.Gln157Hisfs*52) in four patients with CGL4 presenting with generalized lipoatrophy and associated metabolic abnormalities. In probands' fibroblasts, cavin-1 expression was undetectable and caveolin-1 and -2 barely expressed. Ultrastructural analysis revealed a loss of membrane caveolae and the presence of numerous cytoplasmic autophagosomes. Patients' cells also showed increased autophagic flux and blunted insulin signaling. These results were reproduced by PTRF knockdown in control fibroblasts and in 3T3-F442A preadipocytes. Cavin-1 deficiency also impaired 3T3-F442A adipocyte differentiation. Suppression of autophagy by small interfering RNA-mediated silencing of ATG5 improved insulin sensitivity and adipocyte differentiation. CONCLUSIONS: This study showed that cavin-1 deficiency resulted in maladaptative autophagy that contributed to insulin resistance and altered adipocyte differentiation. These new pathophysiological mechanisms could open new therapeutic perspectives for adipose tissue diseases including CGL4.


Asunto(s)
Tejido Adiposo/fisiología , Autofagia/fisiología , Caveolina 1/genética , Lipodistrofia Generalizada Congénita/genética , Lipodistrofia Generalizada Congénita/fisiopatología , Adipogénesis/genética , Adolescente , Adulto , Animales , Autofagia/genética , Caveolina 1/deficiencia , Diferenciación Celular/genética , Células Cultivadas , Niño , Preescolar , Consanguinidad , Femenino , Fibroblastos/fisiología , Humanos , Resistencia a la Insulina/genética , Masculino , Ratones , Persona de Mediana Edad , Adulto Joven
17.
Atherosclerosis ; 250: 52-6, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27179706

RESUMEN

BACKGROUND AND AIMS: Familial hypobetalipoproteinemia (FHBL) is a co-dominant disorder characterized by decreased plasma levels of LDL-cholesterol and apolipoprotein B (ApoB). Currently, genetic diagnosis in FHBL relies largely on Sanger sequencing to identify APOB and PCSK9 gene mutations and on western blotting to detect truncated ApoB species. METHODS: Here, we applied targeted enrichment and next-generation sequencing (NGS) on a panel of three FHBL genes and two abetalipoproteinemia genes (APOB, PCSK9, ANGPTL3, MTTP and SAR1B). RESULTS: In this study, we identified five likely pathogenic heterozygous rare variants. These include four novel nonsense mutations in APOB (p.Gln845*, p.Gln2571*, p.Cys2933* and p.Ser3718*) and a rare variant in PCSK9 (Minor Allele Frequency <0.1%). The affected family members tested were shown to be carriers, suggesting co-segregation with low LDL-C. CONCLUSIONS: Our study further demonstrates that NGS is a reliable and practical approach for the molecular screening of FHBL-causative genes that may provide a mean for deciphering the genetic basis in FHBL.


Asunto(s)
Apolipoproteína B-100/genética , Hipobetalipoproteinemias/genética , Mutación , Abetalipoproteinemia/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , LDL-Colesterol/genética , Codón sin Sentido , Biblioteca de Genes , Variación Genética , Heterocigoto , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Hipobetalipoproteinemias/diagnóstico , Persona de Mediana Edad , Linaje , Fenotipo , Proproteína Convertasa 9/genética , Reproducibilidad de los Resultados , Análisis de Secuencia de ADN , Adulto Joven
18.
Diabetes ; 65(11): 3410-3417, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27554469

RESUMEN

Fibroblast growth factor 21 (FGF21) was shown to improve metabolic homeostasis, at least partly by controlling white adipocyte profile and adiponectin secretion. Here, we studied its effect on adipocyte dysfunction in the context of Berardinelli-Seip congenital lipodystrophy (BSCL) linked to seipin deficiency. Bscl2-/- mice displayed a progressive adipose tissue loss with aging as evidenced by the altered profile of residual fat pads and the decrease in adiponectin plasma levels in 12- vs. 4-week-old animals. Aiming to prevent this impairment, we treated 6-week-old Bscl2-/- mice with an FGF21 analog (LY2405319) for a period of 28 days. FGF21 treatment increased adiponectin plasma levels and normalized insulin sensitivity in Bscl2-/- mice by improving the white adipose tissue gene expression pattern. To further decipher the molecular pathways altered by seipin deficiency in mature adipocytes, we developed a unique inducible seipin knockdown cell line (SKD). SKD showed chronic activation of the p38 MAPK pathway associated with apoptotic cell death. Interestingly, FGF21 treatment exerted an antistress effect on SKD cells, reducing p38 MAPK phosphorylation and limiting mature adipocyte loss. Our data demonstrate that FGF21 treatment improves the metabolic profile of Bscl2-/- lipodystrophic mice, partly by improving mature adipocyte maintenance through suppression of cellular stress via inhibition of p38 MAPK activity.


Asunto(s)
Adipocitos Blancos/metabolismo , Factores de Crecimiento de Fibroblastos/farmacología , Proteínas de Unión al GTP Heterotriméricas/deficiencia , Células 3T3-L1 , Adipocitos Blancos/efectos de los fármacos , Adiponectina/sangre , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Western Blotting , Factores de Crecimiento de Fibroblastos/química , Subunidades gamma de la Proteína de Unión al GTP , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Homeostasis/efectos de los fármacos , Homeostasis/genética , Homeostasis/fisiología , Ratones , Ratones Noqueados , ARN Mensajero/genética
19.
Diabetes ; 51(4): 1291-3, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11916958

RESUMEN

Congenital generalized lipodystrophy (CGL) is a rare disorder characterized by the absence of body fat and insulin resistance and accompanied by other features, including acanthosis nigricans, organomegaly, hyperandrogenism, and diabetes. We have examined case subjects from 11 families in Oman with CGL. All subjects were the progeny of consanguineous marriages; therefore, a homozygosity mapping strategy was used to investigate the reported loci, 11q13 and 9q34. Three subjects could be linked to 11q13, and mutations were found within the seipin gene. An additional eight subjects were linked to 9q34, but the locus was in a 9-cM interval with no known microsatellites, so further fine mapping was not possible. However, two sibships (four subjects) did not map to either locus, raising the possibility of more than two lipodystrophy loci within the Oman population.


Asunto(s)
Cromosomas Humanos Par 11 , Cromosomas Humanos Par 13 , Cromosomas Humanos Par 9 , Lipodistrofia/genética , Mapeo Cromosómico , Consanguinidad , Femenino , Ligamiento Genético , Marcadores Genéticos , Humanos , Lipodistrofia/congénito , Masculino , Repeticiones de Microsatélite , Omán , Linaje
20.
Diabetes ; 52(6): 1573-8, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12765973

RESUMEN

Berardinelli-Seip congenital lipodystrophy (BSCL) is a heterogeneous genetic disease characterized by near absence of adipose tissue and severe insulin resistance. We have previously identified mutations in the seipin gene in a subset of our patients' cohort. Recently, disease-causing mutations in AGPAT2 have been reported in BSCL patients. In this study, we have performed mutation screening in AGPAT2 and the related AGPAT1 in patients with BSCL or other forms of lipodystrophy who have no detectable mutation in the seipin gene. We found 38 BSCL patients from 30 families with mutations in AGPAT2. Three of the known mutations were frequently found in our families. Of the eight new alterations, six are null mutations and two are missense mutations (Glu172Lys and Ala238Gly). All the patients harboring AGPAT2 mutations presented with typical features of BSCL. We did not find mutations in patients with other forms of lipodystrophies, including the syndromes of Lawrence, Dunnigan, and Barraquer-Simons, or with type A insulin resistance. In conclusion, mutations in the seipin gene and AGPAT2 are confined to the BSCL phenotype. Because we found mutations in 92 of the 94 BSCL patients studied, the seipin gene and AGPAT2 are the two major genes involved in the etiology of BSCL.


Asunto(s)
Aciltransferasas/genética , Cromosomas Humanos Par 9 , Subunidades gamma de la Proteína de Unión al GTP , Lipodistrofia/genética , Mutación , 1-Acilglicerol-3-Fosfato O-Aciltransferasa , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Secuencia de Bases , Cartilla de ADN , Femenino , Proteínas de Unión al GTP Heterotriméricas/genética , Humanos , Lipodistrofia/enzimología , Masculino , Datos de Secuencia Molecular , Linaje
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA