Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 37(11): e23211, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37773757

RESUMEN

ARL15, a small GTPase protein, was linked to metabolic traits in association studies. We aimed to test the Arl15 gene as a functional candidate for metabolic traits in the mouse. CRISPR/Cas9 germline knockout (KO) of Arl15 showed that homozygotes were postnatal lethal and exhibited a complete cleft palate (CP). Also, decreased cell migration was observed from Arl15 KO mouse embryonic fibroblasts (MEFs). Metabolic phenotyping of heterozygotes showed that females had reduced fat mass on a chow diet from 14 weeks of age. Mild body composition phenotypes were also observed in heterozygous mice on a high-fat diet (HFD)/low-fat diet (LFD). Females on a HFD showed reduced body weight, gonadal fat depot weight and brown adipose tissue (BAT) weight. In contrast, in the LFD group, females showed increased bone mineral density (BMD), while males showed a trend toward reduced BMD. Clinical biochemistry analysis of plasma on HFD showed transient lower adiponectin at 20 weeks of age in females. Urinary and plasma Mg2+ concentrations were not significantly different. Our phenotyping data showed that Arl15 is essential for craniofacial development. Adult metabolic phenotyping revealed potential roles in brown adipose tissue and bone development.


Asunto(s)
Fisura del Paladar , Masculino , Femenino , Ratones , Animales , Técnicas de Inactivación de Genes , Fisura del Paladar/genética , Fisura del Paladar/metabolismo , Fibroblastos/metabolismo , Dieta Alta en Grasa , Tejido Adiposo Pardo/metabolismo , Adiponectina/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados
2.
Diabetes Obes Metab ; 25(11): 3161-3170, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37435697

RESUMEN

AIM: To investigate whether mesna-sodium-2-mercaptoethane sulfonate) can reduce diet-induced fat gain in mice, and to assess the safety of single ascending mesna doses in humans to find the dose associated with lowering of plasma tCys by at least 30%. METHODS: C3H/HeH mice were shifted to a high-fat diet ± mesna in drinking water; body composition was measured at weeks 0, 2 and 4. In an open, phase I, single ascending dose study, oral mesna (400, 800, 1200, 1600 mg) was administered to 17 men with overweight or obesity. Mesna and tCys concentrations were measured repeatedly for a duration of 48 hours postdosing in plasma, as well as in 24-hour urine. RESULTS: Compared with controls, mesna-treated mice had lower tCys and lower estimated mean fat mass gain from baseline (week 2: 4.54 ± 0.40 vs. 6.52 ± 0.36 g; week 4: 6.95 ± 0.35 vs. 8.19 ± 0.34 g; Poverall = .002), but similar lean mass gain. In men with overweight, mesna doses of 400-1600 mg showed dose linearity and were well tolerated. Mesna doses of 800 mg or higher decreased plasma tCys by 30% or more at nadir (4h post-dosing). With increasing mesna dose, tCys AUC0-12h decreased (Ptrend < .001), and urine tCys excretion increased (Ptrend = .004). CONCLUSIONS: Mesna reduces diet-induced fat gain in mice. In men with overweight, single oral doses of mesna (800-1600 mg) were well tolerated and lowered plasma tCys efficiently. The effect of sustained tCys-lowering by repeated mesna administration on weight loss in humans deserves investigation.


Asunto(s)
Cisteína , Mesna , Humanos , Masculino , Mesna/farmacología , Ratones Endogámicos C3H , Obesidad/tratamiento farmacológico , Sobrepeso/complicaciones , Sobrepeso/tratamiento farmacológico , Animales , Ratones , Ensayos Clínicos Fase I como Asunto
3.
Am J Physiol Gastrointest Liver Physiol ; 319(3): G345-G360, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32755310

RESUMEN

The pathogenesis of nonalcoholic fatty liver disease (NAFLD) and the progression to nonalcoholic steatohepatitis (NASH) and increased risk of hepatocellular carcinoma remain poorly understood. Additionally, there is increasing recognition of the extrahepatic manifestations associated with NAFLD and NASH. We demonstrate that intervention with the American lifestyle-induced obesity syndrome (ALIOS) diet in male and female mice recapitulates many of the clinical and transcriptomic features of human NAFLD and NASH. Male and female C57BL/6N mice were fed either normal chow (NC) or ALIOS from 11 to 52 wk and underwent comprehensive metabolic analysis throughout the duration of the study. From 26 wk, ALIOS-fed mice developed features of hepatic steatosis, inflammation, and fibrosis. ALIOS-fed mice also had an increased incidence of hepatic tumors at 52 wk compared with those fed NC. Hepatic transcriptomic analysis revealed alterations in multiple genes associated with inflammation and tissue repair in ALIOS-fed mice. Ingenuity Pathway Analysis confirmed dysregulation of metabolic pathways as well as those associated with liver disease and cancer. In parallel the development of a robust hepatic phenotype, ALIOS-fed mice displayed many of the extrahepatic manifestations of NAFLD, including hyperlipidemia, increased fat mass, sarcopenia, and insulin resistance. The ALIOS diet in mice recapitulates many of the clinical features of NAFLD and, therefore, represents a robust and reproducible model for investigating the pathogenesis of NAFLD and its progression.NEW & NOTEWORTHY Nonalcoholic fatty liver disease (NAFLD) affects 30% of the general population and can progress to nonalcoholic steatohepatitis (NASH) and potentially hepatocellular carcinoma. Preclinical models rely on mouse models that often display hepatic characteristics of NAFLD but rarely progress to NASH and seldom depict the multisystem effects of the disease. We have conducted comprehensive metabolic analysis of both male and female mice consuming a Western diet of trans fats and sugar, focusing on both their hepatic phenotype and extrahepatic manifestations.


Asunto(s)
Dieta Occidental/efectos adversos , Hígado Graso/genética , Estilo de Vida , Enfermedad del Hígado Graso no Alcohólico/genética , Obesidad/metabolismo , Alimentación Animal , Animales , Composición Corporal , Hígado Graso/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/genética , Prueba de Tolerancia a la Glucosa , Resistencia a la Insulina , Lípidos/sangre , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Pruebas de Función Hepática , Neoplasias Hepáticas/epidemiología , Neoplasias Hepáticas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Síndrome
4.
Mamm Genome ; 31(1-2): 17-29, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31974728

RESUMEN

The proprotein convertase subtilisin/Kexin type 1 (PCSK1/PC1) protein processes inactive pro-hormone precursors into biologically active hormones in a number of neuroendocrine and endocrine cell types. Patients with recessive mutations in PCSK1 exhibit a complex spectrum of traits including obesity, diarrhoea and endocrine disorders. We describe here a new mouse model with a point mutation in the Pcsk1 gene that exhibits obesity, hyperphagia, transient diarrhoea and hyperproinsulinaemia, phenotypes consistent with human patient traits. The mutation results in a pV96L amino acid substitution and changes the first nucleotide of mouse exon 3 leading to skipping of that exon and in homozygotes very little full-length transcript. Overexpression of the exon 3 deleted protein or the 96L protein results in ER retention in Neuro2a cells. This is the second Pcsk1 mouse model to display obesity phenotypes, contrasting knockout mouse alleles. This model will be useful in investigating the basis of endocrine disease resulting from prohormone processing defects.


Asunto(s)
Diabetes Mellitus/genética , Obesidad/genética , Proproteína Convertasa 1/genética , Alelos , Animales , Línea Celular , Diarrea/genética , Retículo Endoplásmico/metabolismo , Exones , Femenino , Intolerancia a la Glucosa/genética , Homocigoto , Hiperfagia/genética , Masculino , Ratones , Mutación , Proproteína Convertasa 1/metabolismo , Empalme del ARN
5.
Proc Natl Acad Sci U S A ; 110(7): 2557-62, 2013 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-23359686

RESUMEN

SNPs in the first intron of FTO (fat mass and obesity associated) are strongly associated with human obesity. While it is not yet formally established that this effect is mediated through the actions of the FTO protein itself, loss of function mutations in FTO or its murine homologue Fto result in severe growth retardation, and mice globally overexpressing FTO are obese. The mechanisms through which FTO influences growth and body composition are unknown. We describe a role for FTO in the coupling of amino acid levels to mammalian target of rapamycin complex 1 signaling. These findings suggest that FTO may influence body composition through playing a role in cellular nutrient sensing.


Asunto(s)
Aminoácidos/metabolismo , Composición Corporal/genética , Obesidad/genética , Proteínas/genética , Proteínas/metabolismo , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/metabolismo , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato , Animales , Fraccionamiento Celular , Cromatografía Liquida , Electroforesis en Gel de Poliacrilamida , Fibroblastos , Células HEK293 , Humanos , Inmunoprecipitación , Ratones , Polimorfismo de Nucleótido Simple/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espectrometría de Masas en Tándem
6.
PLoS Genet ; 9(1): e1003166, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23300482

RESUMEN

The strongest BMI-associated GWAS locus in humans is the FTO gene. Rodent studies demonstrate a role for FTO in energy homeostasis and body composition. The phenotypes observed in loss of expression studies are complex with perinatal lethality, stunted growth from weaning, and significant alterations in body composition. Thus understanding how and where Fto regulates food intake, energy expenditure, and body composition is a challenge. To address this we generated a series of mice with distinct temporal and spatial loss of Fto expression. Global germline loss of Fto resulted in high perinatal lethality and a reduction in body length, fat mass, and lean mass. When ratio corrected for lean mass, mice had a significant increase in energy expenditure, but more appropriate multiple linear regression normalisation showed no difference in energy expenditure. Global deletion of Fto after the in utero and perinatal period, at 6 weeks of age, removed the high lethality of germline loss. However, there was a reduction in weight by 9 weeks, primarily as loss of lean mass. Over the subsequent 10 weeks, weight converged, driven by an increase in fat mass. There was a switch to a lower RER with no overall change in food intake or energy expenditure. To test if the phenotype can be explained by loss of Fto in the mediobasal hypothalamus, we sterotactically injected adeno-associated viral vectors encoding Cre recombinase to cause regional deletion. We observed a small reduction in food intake and weight gain with no effect on energy expenditure or body composition. Thus, although hypothalamic Fto can impact feeding, the effect of loss of Fto on body composition is brought about by its actions at sites elsewhere. Our data suggest that Fto may have a critical role in the control of lean mass, independent of its effect on food intake.


Asunto(s)
Composición Corporal/genética , Ingestión de Alimentos/genética , Metabolismo Energético/genética , Oxigenasas de Función Mixta/genética , Obesidad , Oxo-Ácido-Liasas/genética , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato , Animales , Peso Corporal/genética , Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica , Células Germinativas/metabolismo , Homeostasis , Humanos , Masculino , Ratones , Obesidad/genética , Obesidad/metabolismo
7.
Liver Int ; 35(3): 764-73, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25244648

RESUMEN

BACKGROUND & AIMS: Variations in intestinal microbiota may influence acetaminophen metabolism. This study aimed to determine whether intestinal microbiota are a source of differential susceptibility to acetaminophen-induced hepatotoxicity. METHODS: Conventionally housed C3H/HeH (CH) and C3H/HeH germ-free (GF) mice were administered a 200 mg/kg IP dose of acetaminophen. The severity of hepatotoxicity at 8 h was assessed by histology and biochemical indices. A urinary metabolic profile was obtained using (1) H-NMR. Baseline hepatic glutathione content and CYP2E1 expression were quantified. An additional group of C3H/HeJ (LPS-r) mice were assessed to determine the contribution of LPS/TLR4 signalling. RESULTS: Baseline glutathione levels were significantly reduced (P = 0.03) in GF mice. CYP2E1 mRNA expression and protein levels were not altered. Interindividual variability did not differ between GF and CH groups. No significant differences in the extent of hepatocellular injury (ALT or percentage necrosis) were demonstrated. However, a milder acute liver failure (ALF) phenotype was shown in GF compared with CH mice, with reduced plasma bilirubin and creatinine and increased blood glucose. Differential acetaminophen metabolism was demonstrated. GF mice displayed a higher urinary acetaminophen-sulphate:glucuronide ratio compared with CH (P = 0.01). Urinary analysis showed metabolic differentiation of GF and CH groups at baseline and 8 h (cross-validated anova P = 1 × 10(-22) ). Interruption of TLR4 signalling in LPS-r mice had additional protective effects. CONCLUSION: Variations in intestinal microbiota do not fully explain differential susceptibility to acetaminophen-induced hepatotoxicity. GF mice experienced some protection from secondary complications following acetaminophen overdose and this may be mediated through reduced TLR4/LPS signalling.


Asunto(s)
Acetaminofén/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/fisiopatología , Intestinos/microbiología , Hígado/patología , Microbiota , Alanina Transaminasa/sangre , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/microbiología , Citocromo P-450 CYP2E1/sangre , Citocinas/sangre , Modelos Animales de Enfermedad , Glutatión/sangre , Masculino , Ratones , Ratones Endogámicos C3H , Espectroscopía de Protones por Resonancia Magnética
8.
Am J Pathol ; 183(3): 692-708, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23845568

RESUMEN

A frequently used experimental model of chronic pancreatitis (CP) recapitulating human disease is repeated injection of cerulein into mice. C57BL/6 is the most commonly used inbred mouse strain for biomedical research, but widespread demand has led to generation of several substrains with subtly different phenotypes. In this study, two common substrains, C57BL/6J and C57BL/6NHsd, exhibited different degrees of CP, with C57BL/6J being more susceptible to repetitive cerulein-induced CP as assessed by pancreatic atrophy, pancreatic morphological changes, and fibrosis. We hypothesized that the deficiency of nicotinamide nucleotide transhydrogenase (NNT) protein in C57BL/6J is responsible for the more severe C57BL/6J phenotype but the parameters of CP in NNT-expressing transgenic mice generated on a C57BL6/J background do not differ with those of wild-type C57BL/6J. The highly similar genetic backgrounds but different CP phenotypes of these two substrains presents a unique opportunity to discover genes important in pathogenesis of CP. We therefore performed whole mouse genome Affymetrix microarray analysis of pancreatic gene expression of C57BL/6J and C57BL/6NHsd before and after induction of CP. Genes with differentially regulated expression between the two substrains that might be candidates in CP progression included Mmp7, Pcolce2, Itih4, Wdfy1, and Vtn. We also identified several genes associated with development of CP in both substrains, including RIKEN cDNA 1810009J06 gene (trypsinogen 5), Ccl8, and Ccl6.


Asunto(s)
Progresión de la Enfermedad , Páncreas/patología , Pancreatitis Crónica/inducido químicamente , Pancreatitis Crónica/patología , Animales , Separación Celular , Ceruletida , Quimiocina CCL8/genética , Quimiocina CCL8/metabolismo , Quimiocinas CC/genética , Quimiocinas CC/metabolismo , Colágeno/metabolismo , Fibrosis/patología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Estudios de Asociación Genética , Humanos , Ratones , Ratones Endogámicos C57BL , NADP Transhidrogenasas/deficiencia , NADP Transhidrogenasas/metabolismo , Páncreas/metabolismo , Células Estrelladas Pancreáticas/metabolismo , Células Estrelladas Pancreáticas/patología , Pancreatitis Crónica/enzimología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Riesgo , Tripsinógeno/genética , Tripsinógeno/metabolismo
9.
PLoS Genet ; 7(4): e1001372, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21533022

RESUMEN

Osteoporotic fracture is a major cause of morbidity and mortality worldwide. Low bone mineral density (BMD) is a major predisposing factor to fracture and is known to be highly heritable. Site-, gender-, and age-specific genetic effects on BMD are thought to be significant, but have largely not been considered in the design of genome-wide association studies (GWAS) of BMD to date. We report here a GWAS using a novel study design focusing on women of a specific age (postmenopausal women, age 55-85 years), with either extreme high or low hip BMD (age- and gender-adjusted BMD z-scores of +1.5 to +4.0, n = 1055, or -4.0 to -1.5, n = 900), with replication in cohorts of women drawn from the general population (n = 20,898). The study replicates 21 of 26 known BMD-associated genes. Additionally, we report suggestive association of a further six new genetic associations in or around the genes CLCN7, GALNT3, IBSP, LTBP3, RSPO3, and SOX4, with replication in two independent datasets. A novel mouse model with a loss-of-function mutation in GALNT3 is also reported, which has high bone mass, supporting the involvement of this gene in BMD determination. In addition to identifying further genes associated with BMD, this study confirms the efficiency of extreme-truncate selection designs for quantitative trait association studies.


Asunto(s)
Densidad Ósea , Fracturas Óseas/genética , Estudio de Asociación del Genoma Completo , N-Acetilgalactosaminiltransferasas/genética , Osteoporosis Posmenopáusica/genética , Trombospondinas/genética , Anciano , Anciano de 80 o más Años , Animales , Estudios de Casos y Controles , Canales de Cloruro/genética , Cromosomas Humanos/genética , Estudios de Cohortes , Modelos Animales de Enfermedad , Femenino , Genotipo , Humanos , Sialoproteína de Unión a Integrina/genética , Proteínas de Unión a TGF-beta Latente/genética , Masculino , Ratones , Persona de Mediana Edad , Modelos Animales , Mutación , Polimorfismo de Nucleótido Simple , Proteoglicanos/genética , Receptores de Factores de Crecimiento Transformadores beta/genética , Factores de Transcripción SOXC/genética , Polipéptido N-Acetilgalactosaminiltransferasa
11.
FASEB J ; 26(8): 3550-62, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22593545

RESUMEN

Nicotinamide nucleotide transhydrogenase (NNT) is a mitochondrial redox-driven proton pump that couples the production of NADPH to the mitochondrial metabolic rate. In this study, we demonstrated for the first time that NNT has a significant effect in the modulation of the immune response and host defense against pathogens. We found that NNT mRNA is enriched in immune system-related tissues and regulated during macrophage activation. Overexpression of NNT in a macrophage cell-line resulted in decreased levels of reactive oxygen species (ROS) and nitric oxide upon induction of the macrophage inflammatory responses. These cells failed to fully activate MAPK signaling pathways, resulting in defective secretion of proinflammatory cytokines in response to LPS, and were inefficient in clearance of intracellular bacteria. We have shown that C57BL/6J mice, which have a deletion in the Nnt gene, exhibited greater resistance to acute pulmonary infection with Streptococcus pneumoniae. Macrophages from these mice generated more ROS and established a stronger inflammatory response to this pathogen. Our results demonstrate a novel role for NNT as a regulator of macrophage-mediated inflammatory responses.


Asunto(s)
Inflamación/fisiopatología , Macrófagos/inmunología , NADP Transhidrogenasas/fisiología , Animales , Línea Celular , Sistema Inmunológico/enzimología , Pulmón/patología , Macrófagos/enzimología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fagocitosis/fisiología , Infecciones Neumocócicas/patología , Infecciones Neumocócicas/fisiopatología , Neumonía Bacteriana/etiología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología
12.
Nat Genet ; 30(3): 255-6, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11850622

RESUMEN

The construction of parallel archives of DNA and sperm from mice mutagenized with ethylnitrosurea (ENU) represents a potentially powerful and rapid approach for identifying point mutations in any gene in the mouse genome. We provide support for this approach and report the identification of mutations in the gene (Gjb2) encoding connexin 26, using archives established from the UK ENU mutagenesis program.


Asunto(s)
Etilnitrosourea/farmacología , Mutágenos/farmacología , Mutación , Alelos , Animales , Femenino , Heterocigoto , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Mutantes
13.
Nat Genet ; 36(11): 1133-7, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15514660

RESUMEN

The goal of the Complex Trait Consortium is to promote the development of resources that can be used to understand, treat and ultimately prevent pervasive human diseases. Existing and proposed mouse resources that are optimized to study the actions of isolated genetic loci on a fixed background are less effective for studying intact polygenic networks and interactions among genes, environments, pathogens and other factors. The Collaborative Cross will provide a common reference panel specifically designed for the integrative analysis of complex systems and will change the way we approach human health and disease.


Asunto(s)
Cruzamiento , Recursos en Salud , Ratones Endogámicos , Animales , Redes Comunitarias , Cruzamientos Genéticos , Bases de Datos Genéticas , Investigación sobre Servicios de Salud , Humanos , Ratones , Recombinación Genética
14.
JBMR Plus ; 7(6): e10739, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37283649

RESUMEN

The nuclear factor I/X (NFIX) gene encodes a ubiquitously expressed transcription factor whose mutations lead to two allelic disorders characterized by developmental, skeletal, and neural abnormalities, namely, Malan syndrome (MAL) and Marshall-Smith syndrome (MSS). NFIX mutations associated with MAL mainly cluster in exon 2 and are cleared by nonsense-mediated decay (NMD) leading to NFIX haploinsufficiency, whereas NFIX mutations associated with MSS are clustered in exons 6-10 and escape NMD and result in the production of dominant-negative mutant NFIX proteins. Thus, different NFIX mutations have distinct consequences on NFIX expression. To elucidate the in vivo effects of MSS-associated NFIX exon 7 mutations, we used CRISPR-Cas9 to generate mouse models with exon 7 deletions that comprised: a frameshift deletion of two nucleotides (Nfix Del2); in-frame deletion of 24 nucleotides (Nfix Del24); and deletion of 140 nucleotides (Nfix Del140). Nfix +/Del2, Nfix +/Del24, Nfix +/Del140, Nfix Del24/Del24, and Nfix Del140/Del140 mice were viable, normal, and fertile, with no skeletal abnormalities, but Nfix Del2/Del2 mice had significantly reduced viability (p < 0.002) and died at 2-3 weeks of age. Nfix Del2 was not cleared by NMD, and NfixDel2/Del2 mice, when compared to Nfix +/+ and Nfix +/Del2 mice, had: growth retardation; short stature with kyphosis; reduced skull length; marked porosity of the vertebrae with decreased vertebral and femoral bone mineral content; and reduced caudal vertebrae height and femur length. Plasma biochemistry analysis revealed Nfix Del2/Del2 mice to have increased total alkaline phosphatase activity but decreased C-terminal telopeptide and procollagen-type-1-N-terminal propeptide concentrations compared to Nfix +/+ and Nfix +/Del2 mice. Nfix Del2/Del2 mice were also found to have enlarged cerebral cortices and ventricular areas but smaller dentate gyrus compared to Nfix +/+ mice. Thus, Nfix Del2/Del2 mice provide a model for studying the in vivo effects of NFIX mutants that escape NMD and result in developmental abnormalities of the skeletal and neural tissues that are associated with MSS. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.

15.
Curr Diab Rep ; 12(6): 651-8, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22996130

RESUMEN

The genomes of many species have now been completely sequenced including human and mouse. Great progress has been made in understanding the complex genetics that underlie diabetes and obesity in human populations. One of the current challenges is the functional identification and characterization of the genes within loci that are being mapped. There are many approaches to this problem and this review outlines the valuable role that the mouse can play. We outline the mouse resources that are available to the research community, including knockouts with conditional potential for every gene, and the efforts of the International Mouse Phenotyping Consortium to attach phenotype information to these genes. We also briefly consider the potential of TALEN technology to tailor-make new mouse models of specific mutations discovered in humans. Finally, we consider the recent progress in characterizing the GWAS genes FTO, TCF7L2, CDKAL1, and SLC30A8 in engineered mouse models.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Modelos Animales de Enfermedad , Estudio de Asociación del Genoma Completo , Obesidad/genética , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato , Animales , Quinasa 5 Dependiente de la Ciclina/genética , Predisposición Genética a la Enfermedad , Humanos , Ratones , Ratones Noqueados , Oxigenasas de Función Mixta/genética , Proteínas del Tejido Nervioso/genética , Oxo-Ácido-Liasas/genética , Proteínas/genética , Proteína 2 Similar al Factor de Transcripción 7/genética , ARNt Metiltransferasas
16.
PLoS Genet ; 5(8): e1000599, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19680540

RESUMEN

Human FTO gene variants are associated with body mass index and type 2 diabetes. Because the obesity-associated SNPs are intronic, it is unclear whether changes in FTO expression or splicing are the cause of obesity or if regulatory elements within intron 1 influence upstream or downstream genes. We tested the idea that FTO itself is involved in obesity. We show that a dominant point mutation in the mouse Fto gene results in reduced fat mass, increased energy expenditure, and unchanged physical activity. Exposure to a high-fat diet enhances lean mass and lowers fat mass relative to control mice. Biochemical studies suggest the mutation occurs in a structurally novel domain and modifies FTO function, possibly by altering its dimerisation state. Gene expression profiling revealed increased expression of some fat and carbohydrate metabolism genes and an improved inflammatory profile in white adipose tissue of mutant mice. These data provide direct functional evidence that FTO is a causal gene underlying obesity. Compared to the reported mouse FTO knockout, our model more accurately reflects the effect of human FTO variants; we observe a heterozygous as well as homozygous phenotype, a smaller difference in weight and adiposity, and our mice do not show perinatal lethality or an age-related reduction in size and length. Our model suggests that a search for human coding mutations in FTO may be informative and that inhibition of FTO activity is a possible target for the treatment of morbid obesity.


Asunto(s)
Tejido Adiposo/metabolismo , Obesidad/genética , Obesidad/metabolismo , Oxo-Ácido-Liasas/genética , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato , Secuencia de Aminoácidos , Animales , Peso Corporal , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Oxigenasas de Función Mixta , Datos de Secuencia Molecular , Mutación Missense , Obesidad/fisiopatología , Oxo-Ácido-Liasas/química , Oxo-Ácido-Liasas/metabolismo , Estructura Terciaria de Proteína , Alineación de Secuencia
17.
Front Physiol ; 13: 953199, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36091365

RESUMEN

Background: Increased waist-to-hip ratio (WHR) is associated with increased mortality and risk of type 2 diabetes and cardiovascular disease. The TBX15-WARS2 locus has consistently been associated with increased WHR. Previous study of the hypomorphic Wars2 V117L/V117L mouse model found phenotypes including severely reduced fat mass, and white adipose tissue (WAT) browning, suggesting Wars2 could be a potential modulator of fat distribution and WAT browning. Methods: To test for differences in browning induction across different adipose depots of Wars2 V117L/V117L mice, we measured multiple browning markers of a 4-month old chow-fed cohort in subcutaneous and visceral WAT and brown adipose tissue (BAT). To explain previously observed fat mass loss, we also tested for the upregulation of plasma mitokines FGF21 and GDF15 and for differences in food intake in the same cohort. Finally, to test for diet-associated differences in fat distribution, we placed Wars2 V117L/V117L mice on low-fat or high-fat diet (LFD, HFD) and assessed their body composition by Echo-MRI and compared terminal adipose depot weights at 6 months of age. Results: The chow-fed Wars2 V117L/V117L mice showed more changes in WAT browning marker gene expression in the subcutaneous inguinal WAT depot (iWAT) than in the visceral gonadal WAT depot (gWAT). These mice also demonstrated reduced food intake and elevated plasma FGF21 and GDF15, and mRNA from heart and BAT. When exposed to HFD, the Wars2 V117L/V117L mice showed resistance to diet-induced obesity and a male and HFD-specific reduction of gWAT: iWAT ratio. Conclusion: Severe reduction of Wars2 gene function causes a systemic phenotype which leads to upregulation of FGF21 and GDF15, resulting in reduced food intake and depot-specific changes in browning and fat mass.

18.
J Endocrinol ; 253(3): 97-113, 2022 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-35318963

RESUMEN

Steroid 5ß-reductase (AKR1D1) plays important role in hepatic bile acid synthesis and glucocorticoid clearance. Bile acids and glucocorticoids are potent metabolic regulators, but whether AKR1D1 controls metabolic phenotype in vivo is unknown. Akr1d1-/- mice were generated on a C57BL/6 background. Liquid chromatography/mass spectrometry, metabolomic and transcriptomic approaches were used to determine effects on glucocorticoid and bile acid homeostasis. Metabolic phenotypes including body weight and composition, lipid homeostasis, glucose tolerance and insulin tolerance were evaluated. Molecular changes were assessed by RNA-Seq and Western blotting. Male Akr1d1-/- mice were challenged with a high fat diet (60% kcal from fat) for 20 weeks. Akr1d1-/- mice had a sex-specific metabolic phenotype. At 30 weeks of age, male, but not female, Akr1d1-/- mice were more insulin tolerant and had reduced lipid accumulation in the liver and adipose tissue yet had hypertriglyceridemia and increased intramuscular triacylglycerol. This phenotype was associated with sexually dimorphic changes in bile acid metabolism and composition but without overt effects on circulating glucocorticoid levels or glucocorticoid-regulated gene expression in the liver. Male Akr1d1-/- mice were not protected against diet-induced obesity and insulin resistance. In conclusion, this study shows that AKR1D1 controls bile acid homeostasis in vivo and that altering its activity can affect insulin tolerance and lipid homeostasis in a sex-dependent manner.


Asunto(s)
Glucocorticoides , Oxidorreductasas , Animales , Ácidos y Sales Biliares , Dieta Alta en Grasa , Femenino , Glucocorticoides/metabolismo , Insulina/metabolismo , Lípidos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidorreductasas/genética , Fenotipo
19.
Cell Metab ; 3(1): 35-45, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16399503

RESUMEN

The C57BL/6J mouse displays glucose intolerance and reduced insulin secretion. QTL mapping identified Nicotinamide Nucleotide Transhydrogenase (Nnt), a nuclear-encoded mitochondrial protein thought to be involved in free radical detoxification, as a candidate gene. To investigate its functional role, we used siRNA to knock down Nnt in insulin-secreting MIN6 cells. This produced a dramatic reduction in insulin secretion and the rise in [Ca2+]i evoked by glucose, but not tolbutamide. We identified two ENU-induced point mutations in Nnt (N68K, G745D). Nnt mutant mice were glucose intolerant and secreted less insulin during a glucose tolerance test. Isolated islets showed impaired insulin secretion in response to glucose, but not to tolbutamide, and glucose failed to enhance ATP levels. Glucose utilization and production of reactive oxygen species were increased in Nnt beta cells. We hypothesize that Nnt mutations/deletion uncouple beta cell mitochondrial metabolism leading to less ATP production, enhanced KATP channel activity, and consequently impaired insulin secretion.


Asunto(s)
Insulina/metabolismo , Proteínas Mitocondriales/fisiología , NADP Transhidrogenasas/fisiología , Adenosina Trifosfato/metabolismo , Animales , Calcio/metabolismo , Línea Celular , Femenino , Glucosa/metabolismo , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Obesos , Proteínas Mitocondriales/deficiencia , Proteínas Mitocondriales/genética , NADP Transhidrogenasas/deficiencia , NADP Transhidrogenasas/genética , Canales de Potasio/metabolismo , ARN Interferente Pequeño/farmacología , Especies Reactivas de Oxígeno/metabolismo
20.
Expert Rev Mol Med ; 13: e1, 2011 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-21211090

RESUMEN

The use of mouse models in medical research has greatly contributed to our understanding of the development of type 2 diabetes mellitus and the mechanisms of disease progression in the context of insulin resistance and ß-cell dysfunction. Maintenance of glucose homeostasis involves a complex interplay of many genes and their actions in response to exogenous stimuli. In recent years, the availability of large population-based cohorts and the capacity to genotype enormous numbers of common genetic variants have driven various large-scale genome-wide association studies, which has greatly accelerated the identification of novel genes likely to be involved in the development of type 2 diabetes. The increasing demand for verifying novel genes is met by the timely development of new mouse resources established as various collaborative projects involving major transgenic and phenotyping centres and laboratories worldwide. The surge of new data will ultimately enable translational research into potential improvement and refinement of current type 2 diabetes therapy options, and hopefully restore quality of life for patients.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/fisiopatología , Modelos Animales de Enfermedad , Ratones , Animales , Estudios de Cohortes , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/terapia , Estudio de Asociación del Genoma Completo , Glucosa/metabolismo , Humanos , Insulina/fisiología , Resistencia a la Insulina/genética , Células Secretoras de Insulina/fisiología , Sitios de Carácter Cuantitativo , Transducción de Señal , Investigación Biomédica Traslacional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA