Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 120
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
FASEB J ; 37(11): e23211, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37773757

RESUMO

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.


Assuntos
Fissura Palatina , Masculino , Feminino , Camundongos , Animais , Técnicas de Inativação de Genes , Fissura Palatina/genética , Fissura Palatina/metabolismo , Fibroblastos/metabolismo , Dieta Hiperlipídica , Tecido Adiposo Marrom/metabolismo , Adiponectina/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout
2.
Diabetes Obes Metab ; 25(11): 3161-3170, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37435697

RESUMO

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.


Assuntos
Cisteína , Mesna , Humanos , Masculino , Mesna/farmacologia , Camundongos Endogâmicos C3H , Obesidade/tratamento farmacológico , Sobrepeso/complicações , Sobrepeso/tratamento farmacológico , Animais , Camundongos , Ensaios Clínicos Fase I como Assunto
3.
Am J Physiol Gastrointest Liver Physiol ; 319(3): G345-G360, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32755310

RESUMO

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.


Assuntos
Dieta Ocidental/efeitos adversos , Fígado Gorduroso/genética , Estilo de Vida , Hepatopatia Gordurosa não Alcoólica/genética , Obesidade/metabolismo , Ração Animal , Animais , Composição Corporal , Fígado Gorduroso/metabolismo , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/genética , Teste de Tolerância a Glucose , Resistência à Insulina , Lipídeos/sangue , Cirrose Hepática/genética , Cirrose Hepática/patologia , Testes de Função Hepática , Neoplasias Hepáticas/epidemiologia , Neoplasias Hepáticas/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/metabolismo , Síndrome
4.
Mamm Genome ; 31(1-2): 17-29, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31974728

RESUMO

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.


Assuntos
Diabetes Mellitus/genética , Obesidade/genética , Pró-Proteína Convertase 1/genética , Alelos , Animais , Linhagem Celular , Diarreia/genética , Retículo Endoplasmático/metabolismo , Éxons , Feminino , Intolerância à Glucose/genética , Homozigoto , Hiperfagia/genética , Masculino , Camundongos , Mutação , Pró-Proteína Convertase 1/metabolismo , Splicing de RNA
5.
Proc Natl Acad Sci U S A ; 110(7): 2557-62, 2013 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-23359686

RESUMO

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.


Assuntos
Aminoácidos/metabolismo , Composição Corporal/genética , Obesidade/genética , Proteínas/genética , Proteínas/metabolismo , Transdução de Sinais/genética , Serina-Treonina Quinases TOR/metabolismo , Dioxigenase FTO Dependente de alfa-Cetoglutarato , Animais , Fracionamento Celular , Cromatografia Líquida , Eletroforese em Gel de Poliacrilamida , Fibroblastos , Células HEK293 , Humanos , Imunoprecipitação , Camundongos , Polimorfismo de Nucleotídeo Único/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espectrometria de Massas em Tandem
6.
PLoS Genet ; 9(1): e1003166, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23300482

RESUMO

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.


Assuntos
Composição Corporal/genética , Ingestão de Alimentos/genética , Metabolismo Energético/genética , Oxigenases de Função Mista/genética , Obesidade , Oxo-Ácido-Liases/genética , Dioxigenase FTO Dependente de alfa-Cetoglutarato , Animais , Peso Corporal/genética , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/metabolismo , Homeostase , Humanos , Masculino , Camundongos , Obesidade/genética , Obesidade/metabolismo
7.
Liver Int ; 35(3): 764-73, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25244648

RESUMO

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.


Assuntos
Acetaminofen/toxicidade , Doença Hepática Induzida por Substâncias e Drogas/fisiopatologia , Intestinos/microbiologia , Fígado/patologia , Microbiota , Alanina Transaminase/sangue , Animais , Doença Hepática Induzida por Substâncias e Drogas/microbiologia , Citocromo P-450 CYP2E1/sangue , Citocinas/sangue , Modelos Animais de Doenças , Glutationa/sangue , Masculino , Camundongos , Camundongos Endogâmicos C3H , Espectroscopia de Prótons por Ressonância Magnética
8.
Brain ; 137(Pt 12): 3171-85, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25348630

RESUMO

Mutations in the skeletal muscle channel (SCN4A), encoding the Nav1.4 voltage-gated sodium channel, are causative of a variety of muscle channelopathies, including non-dystrophic myotonias and periodic paralysis. The effects of many of these mutations on channel function have been characterized both in vitro and in vivo. However, little is known about the consequences of SCN4A mutations downstream from their impact on the electrophysiology of the Nav1.4 channel. Here we report the discovery of a novel SCN4A mutation (c.1762A>G; p.I588V) in a patient with myotonia and periodic paralysis, located within the S1 segment of the second domain of the Nav1.4 channel. Using N-ethyl-N-nitrosourea mutagenesis, we generated and characterized a mouse model (named draggen), carrying the equivalent point mutation (c.1744A>G; p.I582V) to that found in the patient with periodic paralysis and myotonia. Draggen mice have myotonia and suffer from intermittent hind-limb immobility attacks. In-depth characterization of draggen mice uncovered novel systemic metabolic abnormalities in Scn4a mouse models and provided novel insights into disease mechanisms. We discovered metabolic alterations leading to lean mice, as well as abnormal AMP-activated protein kinase activation, which were associated with the immobility attacks and may provide a novel potential therapeutic target.


Assuntos
Proteínas Quinases Ativadas por AMP/genética , Canalopatias/genética , Mutação/genética , Miotonia/genética , Transtornos Miotônicos/genética , Canal de Sódio Disparado por Voltagem NAV1.4/genética , Paralisias Periódicas Familiares/genética , Animais , Humanos , Camundongos , Linhagem
9.
Am J Pathol ; 183(3): 692-708, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23845568

RESUMO

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.


Assuntos
Progressão da Doença , Pâncreas/patologia , Pancreatite Crônica/induzido quimicamente , Pancreatite Crônica/patologia , Animais , Separação Celular , Ceruletídeo , Quimiocina CCL8/genética , Quimiocina CCL8/metabolismo , Quimiocinas CC/genética , Quimiocinas CC/metabolismo , Colágeno/metabolismo , Fibrose/patologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Estudos de Associação Genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , NADP Trans-Hidrogenases/deficiência , NADP Trans-Hidrogenases/metabolismo , Pâncreas/metabolismo , Células Estreladas do Pâncreas/metabolismo , Células Estreladas do Pâncreas/patologia , Pancreatite Crônica/enzimologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Risco , Tripsinogênio/genética , Tripsinogênio/metabolismo
10.
Nat Cell Biol ; 9(4): 453-60, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17369816

RESUMO

Pancreatic islets have a central role in blood glucose homeostasis. In addition to insulin-producing beta-cells and glucagon-secreting alpha-cells, the islets contain somatostatin-releasing delta-cells. Somatostatin is a powerful inhibitor of insulin and glucagon secretion. It is normally secreted in response to glucose and there is evidence suggesting its release becomes perturbed in diabetes. Little is known about the control of somatostatin release. Closure of ATP-regulated K(+)-channels (K(ATP)-channels) and a depolarization-evoked increase in cytoplasmic free Ca(2+) concentration ([Ca(2+)](i)) have been proposed to be essential. Here, we report that somatostatin release evoked by high glucose (>or=10 mM) is unaffected by the K(ATP)-channel activator diazoxide and proceeds normally in K(ATP)-channel-deficient islets. Glucose-induced somatostatin secretion is instead primarily dependent on Ca(2+)-induced Ca(2+)-release (CICR). This constitutes a novel mechanism for K(ATP)-channel-independent metabolic control of pancreatic hormone secretion.


Assuntos
Canais de Cálcio Tipo R/fisiologia , Cálcio/metabolismo , Glucose/farmacologia , Somatostatina/metabolismo , Animais , Cálcio/farmacologia , Canais de Cálcio Tipo R/genética , Citofotometria , Diazóxido/farmacologia , Relação Dose-Resposta a Droga , Eletrofisiologia , Imuno-Histoquímica , Técnicas In Vitro , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Isradipino/farmacologia , Compostos Macrocíclicos/farmacologia , Manoeptulose/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Microscopia Confocal , Oxazóis/farmacologia , Potássio/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/fisiologia , Rianodina/farmacologia , Células Secretoras de Somatostatina/efeitos dos fármacos , Células Secretoras de Somatostatina/metabolismo
11.
PLoS Genet ; 7(4): e1001372, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21533022

RESUMO

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.


Assuntos
Densidade Óssea , Fraturas Ósseas/genética , Estudo de Associação Genômica Ampla , N-Acetilgalactosaminiltransferases/genética , Osteoporose Pós-Menopausa/genética , Trombospondinas/genética , Idoso , Idoso de 80 Anos ou mais , Animais , Estudos de Casos e Controles , Canais de Cloreto/genética , Cromossomos Humanos/genética , Estudos de Coortes , Modelos Animais de Doenças , Feminino , Genótipo , Humanos , Sialoproteína de Ligação à Integrina/genética , Proteínas de Ligação a TGF-beta Latente/genética , Masculino , Camundongos , Pessoa de Meia-Idade , Modelos Animais , Mutação , Polimorfismo de Nucleotídeo Único , Proteoglicanas/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Fatores de Transcrição SOXC/genética , Polipeptídeo N-Acetilgalactosaminiltransferase
12.
bioRxiv ; 2024 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-39131393

RESUMO

There are multiple independent genetic signals at the Ras-responsive element binding protein 1 (RREB1) locus associated with type 2 diabetes risk, fasting glucose, ectopic fat, height, and bone mineral density. We have previously shown that loss of RREB1 in pancreatic beta cells reduces insulin content and impairs islet cell development and function. However, RREB1 is a widely expressed transcription factor and the metabolic impact of RREB1 loss in vivo remains unknown. Here, we show that male and female global heterozygous knockout (Rreb1 +/-) mice have reduced body length, weight, and fat mass on high-fat diet. Rreb1+/- mice have sex- and diet-specific decreases in adipose tissue and adipocyte size; male mice on high-fat diet had larger gonadal adipocytes, while males on standard chow and females on high-fat diet had smaller, more insulin sensitive subcutaneous adipocytes. Mouse and human precursor cells lacking RREB1 have decreased adipogenic gene expression and activated transcription of genes associated with osteoblast differentiation, which was associated with Rreb1 +/- mice having increased bone mineral density in vivo. Finally, human carriers of RREB1 T2D protective alleles have smaller adipocytes, consistent with RREB1 loss-of-function reducing diabetes risk.

14.
FASEB J ; 26(8): 3550-62, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22593545

RESUMO

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.


Assuntos
Inflamação/fisiopatologia , Macrófagos/imunologia , NADP Trans-Hidrogenases/fisiologia , Animais , Linhagem Celular , Sistema Imunitário/enzimologia , Pulmão/patologia , Macrófagos/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fagocitose/fisiologia , Infecções Pneumocócicas/patologia , Infecções Pneumocócicas/fisiopatologia , Pneumonia Bacteriana/etiologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia
15.
Nat Genet ; 30(3): 255-6, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11850622

RESUMO

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.


Assuntos
Etilnitrosoureia/farmacologia , Mutagênicos/farmacologia , Mutação , Alelos , Animais , Feminino , Heterozigoto , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Mutantes
16.
Nat Genet ; 34(2): 181-6, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12754510

RESUMO

Atopic or immunoglobulin E (IgE)-mediated diseases include the common disorders of asthma, atopic dermatitis and allergic rhinitis. Chromosome 13q14 shows consistent linkage to atopy and the total serum IgE concentration. We previously identified association between total serum IgE levels and a novel 13q14 microsatellite (USAT24G1; ref. 7) and have now localized the underlying quantitative-trait locus (QTL) in a comprehensive single-nucleotide polymorphism (SNP) map. We found replicated association to IgE levels that was attributed to several alleles in a single gene, PHF11. We also found association with these variants to severe clinical asthma. The gene product (PHF11) contains two PHD zinc fingers and probably regulates transcription. Distinctive splice variants were expressed in immune tissues and cells.


Assuntos
Asma/genética , Cromossomos Humanos Par 13/genética , Imunoglobulina E/sangue , Locos de Características Quantitativas , Adulto , Alelos , Processamento Alternativo , Estudos de Casos e Controles , Criança , Feminino , Haplótipos , Humanos , Masculino , Dados de Sequência Molecular , Polimorfismo de Nucleotídeo Único , Distribuição Tecidual , Dedos de Zinco/genética
17.
Nat Genet ; 36(9): 925-7, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15340424

RESUMO

The European Mouse Mutagenesis Consortium is the European initiative contributing to the international effort on functional annotation of the mouse genome. Its objectives are to establish and integrate mutagenesis platforms, gene expression resources, phenotyping units, storage and distribution centers and bioinformatics resources. The combined efforts will accelerate our understanding of gene function and of human health and disease.


Assuntos
Pesquisa em Genética , Genoma , Camundongos/genética , Mutagênese , Animais , Biologia Computacional , Europa (Continente) , Fenótipo
18.
Nat Genet ; 36(11): 1133-7, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15514660

RESUMO

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.


Assuntos
Cruzamento , Recursos em Saúde , Camundongos Endogâmicos , Animais , Redes Comunitárias , Cruzamentos Genéticos , Bases de Dados Genéticas , Pesquisa sobre Serviços de Saúde , Humanos , Camundongos , Recombinação Genética
19.
JBMR Plus ; 7(6): e10739, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37283649

RESUMO

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.

20.
J Clin Invest ; 119(1): 80-90, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19065048

RESUMO

Neonatal diabetes is a rare monogenic form of diabetes that usually presents within the first six months of life. It is commonly caused by gain-of-function mutations in the genes encoding the Kir6.2 and SUR1 subunits of the plasmalemmal ATP-sensitive K+ (KATP) channel. To better understand this disease, we generated a mouse expressing a Kir6.2 mutation (V59M) that causes neonatal diabetes in humans and we used Cre-lox technology to express the mutation specifically in pancreatic beta cells. These beta-V59M mice developed severe diabetes soon after birth, and by 5 weeks of age, blood glucose levels were markedly increased and insulin was undetectable. Islets isolated from beta-V59M mice secreted substantially less insulin and showed a smaller increase in intracellular calcium in response to glucose. This was due to a reduced sensitivity of KATP channels in pancreatic beta cells to inhibition by ATP or glucose. In contrast, the sulfonylurea tolbutamide, a specific blocker of KATP channels, closed KATP channels, elevated intracellular calcium levels, and stimulated insulin release in beta-V59M beta cells, indicating that events downstream of KATP channel closure remained intact. Expression of the V59M Kir6.2 mutation in pancreatic beta cells alone is thus sufficient to recapitulate the neonatal diabetes observed in humans. beta-V59M islets also displayed a reduced percentage of beta cells, abnormal morphology, lower insulin content, and decreased expression of Kir6.2, SUR1, and insulin mRNA. All these changes are expected to contribute to the diabetes of beta-V59M mice. Their cause requires further investigation.


Assuntos
Diabetes Mellitus/genética , Modelos Animais de Doenças , Células Secretoras de Insulina/metabolismo , Mutação , Canais de Potássio Corretores do Fluxo de Internalização/genética , Subunidades Proteicas/genética , Trifosfato de Adenosina/metabolismo , Animais , Cálcio/metabolismo , Diabetes Mellitus/metabolismo , Feminino , Humanos , Hipoglicemiantes/farmacologia , Recém-Nascido , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Masculino , Camundongos , Camundongos Transgênicos , Técnicas de Patch-Clamp , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Subunidades Proteicas/metabolismo , Tolbutamida/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA