Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
Physiol Res ; 70(2): 227-236, 2021 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-33676383

RESUMO

Mice are important models for biomedical research by providing the possibility of standardizing genetic background and environmental conditions, which both affect phenotypic variability. Use of both sexes in experiments is strongly recommended because of possible differences in the outcome. However, sex-specific phenotypic variability is discussed with regard to putative consequences on the group size which is necessary for achieving valid and reproducible results. Here, we retrospectively analyzed the sex-specific variability of 25 blood parameters of C3H inbred mice in two different mouse facilities withinthe long-term, high-throughput Munich ENU mouse mutagenesis project. Using the 95 % data range, data of4,780-20,706 mice per parameter were analyzed and resulted in ratios of the coefficient of variation (= female CV / (female CV + male CV)) from 0.44 to 0.58 for the 25 parameters, with an overall mean of 0.51 in both facilities. Together with data analyses of three additional, smaller studies with 72-247 animals per parameter examined and various genetic backgrounds (inbred strains, F1 hybrids) included, hints for reproducible sex-specific variability were observed for particular parameters. Thus, the overall analysis comprising all 25 clinical chemical and hematological parameters of the standardized, long-term analysis of a high number of group housed, young adult, twelve-week-old C3H inbred mice showed no evidence for substantial sex-specific variability. The results may provide a basis for the examination of sex-specific variability in particular blood parameters.


Assuntos
Biomarcadores/sangue , Ensaios de Triagem em Larga Escala , Animais , Genótipo , Camundongos Endogâmicos C3H , Fenótipo , Caracteres Sexuais , Fatores Sexuais , Fatores de Tempo
2.
Mol Metab ; 18: 42-50, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30309776

RESUMO

OBJECTIVE: Although debated, metabolic health characterizes 10-25% of obese individuals and reduces risk of developing life-threatening co-morbidities. Adipose tissue is a recognized endocrine organ important for the maintenance of whole-body metabolic health. Adipocyte transcriptional signatures of healthy and unhealthy obesity are largely unknown. METHODS: Here, we used a small cohort of highly characterized obese individuals discordant for metabolic health, characterized their adipocytes transcriptional signatures, and cross-referenced them to mouse phenotypic and human GWAs databases. RESULTS AND CONCLUSIONS: Our study showed that glucose intolerance and insulin resistance co-operate to remodel adipocyte transcriptome. We also identified the Nuclear Export Mediator Factor (NEMF) and the Ectoderm-Neural Cortex 1 (ENC1) as novel potential targets in the management of metabolic health in human obesity.


Assuntos
Adipócitos/metabolismo , Intolerância à Glucose , Resistência à Insulina , Obesidade/metabolismo , Transcriptoma , Adulto , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Células Cultivadas , Feminino , Humanos , Masculino , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Pessoa de Meia-Idade , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas de Transporte Nucleocitoplasmático/genética , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Obesidade/genética
3.
Int J Obes (Lond) ; 42(3): 507-517, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-28901330

RESUMO

BACKGROUND/OBJECTIVES: Dieting is a popular yet often ineffective way to lower body weight, as the majority of people regain most of their pre-dieting weights in a relatively short time. The underlying molecular mechanisms driving weight regain and the increased risk for metabolic disease are still incompletely understood. Here we investigate the molecular alterations inherited from a history of obesity. METHODS: In our model, male high-fat diet (HFD)-fed obese C57BL/6J mice were switched to a low caloric chow diet, resulting in a decline of body weight to that of lean mice. We measured body composition, as well as metrics of glucose, insulin and lipid homeostasis. This was accompanied by histological and gene expression analysis of adipose tissue and liver to assess adipose tissue inflammation and hepatosteatosis. Moreover, acute hypothalamic response to (re-) exposure to HFD was assessed by qPCR. RESULTS & CONCLUSIONS: Within 7 weeks after diet switch, most obesity-associated phenotypes, such as body mass, glucose intolerance and blood metabolite levels were reversed. However, hepatic inflammation, hepatic steatosis as well as hypertrophy and inflammation of perigonadal, but not subcutaneous, adipocytes persisted in formerly obese mice. Transcriptional profiling of liver and perigonadal fat revealed an upregulation of pathways associated with immune function and cellularity. Thus, we show that weight reduction leaves signs of inflammation in liver and perigonadal fat, indicating that persisting proinflammatory signals in liver and adipose tissue could contribute to an increased risk of formerly obese subjects to develop the metabolic syndrome upon recurring weight gain.


Assuntos
Tecido Adiposo/metabolismo , Inflamação/metabolismo , Fígado/metabolismo , Obesidade/metabolismo , Redução de Peso/fisiologia , Tecido Adiposo/química , Animais , Biomarcadores/análise , Restrição Calórica , Fígado Gorduroso/metabolismo , Fígado/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/dietoterapia
4.
Mol Psychiatry ; 23(5): 1345-1355, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-28373690

RESUMO

Dietary intake of methyl donors, such as folic acid and methionine, shows considerable intra-individual variation in human populations. While it is recognized that maternal departures from the optimum of dietary methyl donor intake can increase the risk for mental health issues and neurological disorders in offspring, it has not been explored whether paternal dietary methyl donor intake influences behavioral and cognitive functions in the next generation. Here, we report that elevated paternal dietary methyl donor intake in a mouse model, transiently applied prior to mating, resulted in offspring animals (methyl donor-rich diet (MD) F1 mice) with deficits in hippocampus-dependent learning and memory, impaired hippocampal synaptic plasticity and reduced hippocampal theta oscillations. Gene expression analyses revealed altered expression of the methionine adenosyltransferase Mat2a and BK channel subunit Kcnmb2, which was associated with changes in Kcnmb2 promoter methylation in MD F1 mice. Hippocampal overexpression of Kcnmb2 in MD F1 mice ameliorated altered spatial learning and memory, supporting a role of this BK channel subunit in the MD F1 behavioral phenotype. Behavioral and gene expression changes did not extend into the F2 offspring generation. Together, our data indicate that paternal dietary factors influence cognitive and neural functions in the offspring generation.


Assuntos
Cognição/fisiologia , Suplementos Nutricionais/efeitos adversos , Herança Paterna/fisiologia , Animais , Metilação de DNA , Dieta , Epigênese Genética , Pai , Ácido Fólico/metabolismo , Hipocampo/metabolismo , Subunidades beta do Canal de Potássio Ativado por Cálcio de Condutância Alta , Aprendizagem/efeitos dos fármacos , Masculino , Memória/efeitos dos fármacos , Metionina/metabolismo , Metionina Adenosiltransferase , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/fisiologia , Herança Paterna/genética , Regiões Promotoras Genéticas
5.
J Mol Endocrinol ; 58(2): 67-78, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27965370

RESUMO

In an attempt to define novel genetic loci involved in the pathophysiology of primary aldosteronism, a mutagenesis screen after treatment with the alkylating agent N-ethyl-N-nitrosourea was established for the parameter aldosterone. One of the generated mouse lines with hyperaldosteronism was phenotypically and genetically characterized. This mouse line had high aldosterone levels but normal creatinine and urea values. The steroidogenic enzyme expression levels in the adrenal gland did not differ significantly among phenotypically affected and unaffected mice. Upon exome sequencing, point mutations were identified in seven candidate genes (Sspo, Dguok, Hoxaas2, Clstn3, Atm, Tipin and Mapk6). Subsequently, animals were stratified into wild-type and mutated groups according to their genotype for each of these candidate genes. A correlation of their genotypes with the respective aldosterone, aldosterone-to-renin ratio (ARR), urea and creatinine values as well as steroidogenic enzyme expression levels was performed. Aldosterone values were significantly higher in animals carrying mutations in four different genes (Sspo, Dguok, Hoxaas2 and Clstn3) and associated statistically significant adrenal Cyp11b2 overexpression as well as increased ARR was present only in mice with Sspo mutation. In contrast, mutations of the remaining candidate genes (Atm, Tipin and Mapk6) were associated with lower aldosterone values and lower Hsd3b6 expression levels. In summary, these data demonstrate association between the genes Sspo, Dguok, Hoxaas2 and Clstn3 and hyperaldosteronism. Final proofs for the causative nature of the mutations have to come from knock-out and knock-in experiments.


Assuntos
Estudos de Associação Genética , Predisposição Genética para Doença , Hiperaldosteronismo/genética , Hiperaldosteronismo/metabolismo , Aldosterona/sangue , Aldosterona/metabolismo , Animais , Biomarcadores , Modelos Animais de Doenças , Exoma , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Masculino , Camundongos , Mutação , Linhagem , Polimorfismo de Nucleotídeo Único
6.
Mol Metab ; 4(1): 39-50, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25685688

RESUMO

OBJECTIVE: Excess lipid intake has been implicated in the pathophysiology of hepatosteatosis and hepatic insulin resistance. Lipids constitute approximately 50% of the cell membrane mass, define membrane properties, and create microenvironments for membrane-proteins. In this study we aimed to resolve temporal alterations in membrane metabolite and protein signatures during high-fat diet (HF)-mediated development of hepatic insulin resistance. METHODS: We induced hepatosteatosis by feeding C3HeB/FeJ male mice an HF enriched with long-chain polyunsaturated C18:2n6 fatty acids for 7, 14, or 21 days. Longitudinal changes in hepatic insulin sensitivity were assessed via the euglycemic-hyperinsulinemic clamp, in membrane lipids via t-metabolomics- and membrane proteins via quantitative proteomics-analyses, and in hepatocyte morphology via electron microscopy. Data were compared to those of age- and litter-matched controls maintained on a low-fat diet. RESULTS: Excess long-chain polyunsaturated C18:2n6 intake for 7 days did not compromise hepatic insulin sensitivity, however, induced hepatosteatosis and modified major membrane lipid constituent signatures in liver, e.g. increased total unsaturated, long-chain fatty acid-containing acyl-carnitine or membrane-associated diacylglycerol moieties and decreased total short-chain acyl-carnitines, glycerophosphocholines, lysophosphatidylcholines, or sphingolipids. Hepatic insulin sensitivity tended to decrease within 14 days HF-exposure. Overt hepatic insulin resistance developed until day 21 of HF-intervention and was accompanied by morphological mitochondrial abnormalities and indications for oxidative stress in liver. HF-feeding progressively decreased the abundance of protein-components of all mitochondrial respiratory chain complexes, inner and outer mitochondrial membrane substrate transporters independent from the hepatocellular mitochondrial volume in liver. CONCLUSIONS: We assume HF-induced modifications in membrane lipid- and protein-signatures prior to and during changes in hepatic insulin action in liver alter membrane properties - in particular those of mitochondria which are highly abundant in hepatocytes. In turn, a progressive decrease in the abundance of mitochondrial membrane proteins throughout HF-exposure likely impacts on mitochondrial energy metabolism, substrate exchange across mitochondrial membranes, contributes to oxidative stress, mitochondrial damage, and the development of insulin resistance in liver.

7.
J Breath Res ; 8(1): 016004, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24566092

RESUMO

Breath gas analysis in humans proved successful in identifying disease states and assessing metabolic functions in a non-invasive way. While many studies report diagnostic capability using volatile organic compounds (VOC) in breath, the inter-individual variability even in healthy human cohorts is rather large and not completely understood in its biochemical origin. Laboratory mice are the predominant animal model system for human disorders and are analysed under highly standardized and controlled conditions. We established a novel setup to monitor VOCs as biomarkers for disease in the breath gas of non-anesthetized, non-restrained mice using a proton transfer reaction mass spectrometer with time of flight detection. In this study, we implemented breath gas analysis in a dietary intervention study in C57BL/6J mice with the aim to assess the variability in VOC signatures due to a change in the diet matrix. Mice were fed a standard laboratory chow and then exposed to four semi-purified low- or high-fat diets for four weeks. Random forest (RF++) was used to identify VOCs that specifically respond to the diet matrix change. Interestingly, we found that the change from a chow diet to semi-purified diets resulted in a considerable drop of several VOC levels. Our results suggest that the diet matrix impacts VOC signatures and the underlying metabolic functions and may be one source of variability in exhaled volatiles.


Assuntos
Testes Respiratórios/métodos , Dieta , Compostos Orgânicos Voláteis/análise , Acetatos/análise , Animais , Biomarcadores/análise , Sistemas Computacionais , Dimetil Sulfóxido/análise , Expiração/fisiologia , Comportamento Alimentar , Humanos , Modelos Lineares , Masculino , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Propionatos/análise , Sulfonas/análise , Aumento de Peso
8.
Biochim Biophys Acta ; 1842(2): 304-17, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24275555

RESUMO

Vulnerability of the fetus upon maternal obesity can potentially occur during all developmental phases. We aimed at elaborating longer-term health outcomes of fetal overnutrition during the earliest stages of development. We utilized Naval Medical Research Institute (NMRI) mice to induce pre-conceptional and gestational obesity and followed offspring outcomes in the absence of any postnatal obesogenic influences. Male adult offspring developed overweight, insulin resistance, hyperleptinemia, hyperuricemia and hepatic steatosis; all these features were not observed in females. Instead, they showed impaired fasting glucose and a reduced fat mass and adipocyte size. Influences of the interaction of maternal diet∗sex concerned offspring genes involved in fatty liver disease, lipid droplet size regulation and fat mass expansion. These data suggest that a peri-conceptional obesogenic exposure is sufficient to shape offspring gene expression patterns and health outcomes in a sex- and organ-specific manner, indicating varying developmental vulnerabilities between sexes towards metabolic disease in response to maternal overnutrition.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Suscetibilidade a Doenças/fisiopatologia , Obesidade/fisiopatologia , Adipócitos/metabolismo , Adipócitos/patologia , Animais , Peso Corporal/fisiologia , Tamanho Celular , Suscetibilidade a Doenças/etiologia , Fígado Gorduroso/etiologia , Fígado Gorduroso/fisiopatologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Teste de Tolerância a Glucose , Hiperuricemia/etiologia , Hiperuricemia/fisiopatologia , Resistência à Insulina/fisiologia , Leptina/sangue , Masculino , Camundongos Endogâmicos , Obesidade/etiologia , Obesidade/genética , Sobrepeso/etiologia , Sobrepeso/fisiopatologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/etiologia , Efeitos Tardios da Exposição Pré-Natal/genética , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores Sexuais , Gordura Subcutânea/metabolismo , Fatores de Tempo
9.
Eur J Pain ; 18(2): 249-57, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23861142

RESUMO

BACKGROUND: Smad-interacting protein 1 (also named Zeb2 and Zfhx1b) is a transcription factor that plays an important role in neuronal development and, when mutated, causes Mowat-Wilson syndrome (MWS). A corresponding mouse model carrying a heterozygous Zeb2 deletion was comprehensively analysed in the German Mouse Clinic. The most prominent phenotype was the reduced pain sensitivity. In this study, we investigated the role of Zeb2 in inflammatory and neuropathic pain. METHODS: For this, we tested mutant Zeb2 animals in different models of inflammatory pain like abdominal constriction, formalin and carrageenan test. Furthermore, we studied the pain reactivity of the mice after peripheral nerve ligation. To examine the nociceptive transmission of primary sensory dorsal root ganglia (DRG) neurons, we determined the neuronal activity in the spinal dorsal horn after the formalin test using staining of c-Fos. Next, we characterized the neuronal cell population in the DRGs and in the sciatic nerve to study the effect of the Zeb2 mutation on peripheral nerve morphology. RESULTS: The present data show that Zeb2 is involved in the development of primary sensory DRG neurons, especially of C- and Aδ fibres. These alterations contribute to a hypoalgesic phenotype in inflammatory but not in neuropathic pain in these Zeb2(+/-) mice. CONCLUSION: Our data suggest that the under-reaction to pain observed in MWS patients results from a reduced responsivity to nociceptive stimulation rather than an inability to communicate discomfort.


Assuntos
Dor Aguda/genética , Gânglios Espinais/metabolismo , Doença de Hirschsprung/genética , Proteínas de Homeodomínio/genética , Deficiência Intelectual/genética , Microcefalia/genética , Neuralgia/genética , Proteínas Repressoras/genética , Fatores de Transcrição/metabolismo , Animais , Dor Crônica/genética , Dor Crônica/metabolismo , Modelos Animais de Doenças , Fácies , Feminino , Predisposição Genética para Doença , Masculino , Camundongos , Mutação/genética , Neuralgia/metabolismo , Medição da Dor/métodos , Medula Espinal/metabolismo , Homeobox 2 de Ligação a E-box com Dedos de Zinco
10.
J Intern Med ; 274(5): 425-39, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24127940

RESUMO

BACKGROUND: Metabolomics is a versatile unbiased method to search for biomarkers of human disease. In particular, one approach in cancer therapy is to promote apoptosis in tumour cells; this could be improved with specific biomarkers of apoptosis for monitoring treatment. We recently observed specific metabolic patterns in apoptotic cell lines; however, in that study, apoptosis was only induced with one pro-apoptotic agent, staurosporine. OBJECTIVE: The aim of this study was to find novel biomarkers of apoptosis by verifying our previous findings using two further pro-apoptotic agents, 5-fluorouracil and etoposide, that are commonly used in anticancer treatment. METHODS: Metabolic parameters were assessed in HepG2 and HEK293 cells using the newborn screening assay adapted for cell culture approaches, quantifying the levels of amino acids and acylcarnitines with mass spectrometry. RESULTS: We were able to identify apoptosis-specific changes in the metabolite profile. Moreover, the amino acids alanine and glutamate were both significantly up-regulated in apoptotic HepG2 and HEK293 cells irrespective of the apoptosis inducer. CONCLUSION: Our observations clearly indicate the potential of metabolomics in detecting metabolic biomarkers applicable in theranostics and for monitoring drug efficacy.


Assuntos
Apoptose/genética , Linhagem Celular Tumoral/metabolismo , Metabolômica , Medicina de Precisão/métodos , Alanina/análise , Aminoácidos/análise , Apoptose/efeitos dos fármacos , Biomarcadores/metabolismo , Carnitina/análogos & derivados , Carnitina/análise , Caspase 3/metabolismo , Caspase 7/metabolismo , Linhagem Celular Tumoral/química , Linhagem Celular Tumoral/efeitos dos fármacos , Etoposídeo/farmacologia , Análise de Injeção de Fluxo , Fluoruracila/farmacologia , Ácido Glutâmico/análise , Células HEK293/química , Células HEK293/efeitos dos fármacos , Células HEK293/metabolismo , Células Hep G2/química , Células Hep G2/efeitos dos fármacos , Células Hep G2/metabolismo , Humanos , Metabolômica/métodos
11.
Oncogene ; 32(20): 2586-91, 2013 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-22751129

RESUMO

AVEN has been identified as an inhibitor of apoptosis, which binds to the adaptor protein, APAF-1, and thereby prevents apoptosome formation and mitochondrial apoptosis. Recent data have demonstrated high expression levels of AVEN messenger RNA in acute leukemias as well as a positive correlation between AVEN mRNA overexpression and poor prognosis in childhood acute lymphoblastic leukemia. On the basis of these data, we investigated the potential involvement of AVEN in tumorigenesis. First, we confirmed the overexpression of AVEN in T-cell acute lymphoblastic leukemia/lymphoma (T-ALL) patient samples. We then established a transgenic mouse model with T-cell-specific overexpression of AVEN, with which we demonstrated the oncogenic cooperation of AVEN with heterozygous loss of p53. Finally, we used a subcutaneous xenograft mouse model to show that AVEN knockdown in the T-ALL cell lines, MOLT-4 and CCRF-CEM, and in the acute myeloblastic leukemia cell line, Kasumi-1, leads to a halt in tumor growth owing to the increased apoptosis and decreased proliferation of tumor cells. Collectively, our data demonstrate that the anti-apoptotic molecule, AVEN, functions as an oncoprotein in hematopoietic neoplasms.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Reguladoras de Apoptose/genética , Proteínas de Membrana/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Sequência de Bases , Linhagem Celular Tumoral , Regulação Leucêmica da Expressão Gênica , Técnicas de Silenciamento de Genes , Genes p53 , Humanos , Linfoma de Células T/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Dados de Sequência Molecular , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patologia , Timócitos/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Mol Cell Endocrinol ; 362(1-2): 139-48, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22698525

RESUMO

In the large-scale Munich N-ethyl-N-nitrosourea (ENU) mouse mutagenesis project murine models recapitulating human diseases were generated. In one strain, a novel missense mutation (D217V) in the glucokinase (Gck) gene was identified, resulting in decreased glucokinase activity. Heterozygous mutants display mild hyperglycaemia, disturbed glucose tolerance, and decreased glucose-induced insulin secretion. In contrast, homozygous mutants exhibit severe but not survival affecting hyperglycaemia, mild growth retardation, diminished oxidative capacity, and increased abundance of CHOP protein in the islets. Furthermore, the total islet and ß-cell volumes and the total volume of isolated ß-cells are significantly decreased in adult homozygous mutants, whereas in neonatal mice, ß-cell mass is not yet significantly decreased and islet neogenesis is unaltered. Therefore, reduced total islet and ß-cell volumes of adult homozygous mutants might predominantly emerge from disturbed postnatal islet neogenesis. Thus, we identified a novel Gck mutation in mice, with relevance in humans, leading to glycaemic disease.


Assuntos
Glucoquinase/genética , Hiperglicemia/genética , Ilhotas Pancreáticas/patologia , Mutação de Sentido Incorreto , Estresse Oxidativo , Animais , Sequência de Bases , Glicemia , Peso Corporal , Análise Mutacional de DNA , Chaperona BiP do Retículo Endoplasmático , Fator de Iniciação 2 em Eucariotos/metabolismo , Feminino , Estudos de Associação Genética , Ligação Genética , Genótipo , Proteínas de Choque Térmico/metabolismo , Hiperglicemia/enzimologia , Hiperglicemia/metabolismo , Ilhotas Pancreáticas/enzimologia , Peroxidação de Lipídeos , Masculino , Malondialdeído/sangue , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Mutagênese , Fator de Transcrição CHOP/metabolismo
13.
Diabetologia ; 54(8): 2132-42, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21562757

RESUMO

AIMS/HYPOTHESIS: Obesity is strongly associated with the development of non-alcoholic fatty liver disease (NAFLD). The cytokine osteopontin (OPN) was recently shown to be involved in obesity-induced adipose tissue inflammation and reduced insulin response. Accumulating evidence links OPN to the pathogenesis of NAFLD. Here we aimed to identify the role of OPN in obesity-associated hepatic steatosis and impaired hepatic glucose metabolism. METHODS: Wild-type (WT) and Opn (also known as Spp1) knockout (Opn (-/-)) mice were fed a high-fat or low-fat diet to study OPN effects in obesity-driven hepatic alterations. RESULTS: We show that genetic OPN deficiency protected from obesity-induced hepatic steatosis, at least in part, by downregulating hepatic triacylglycerol synthesis. Conversely, absence of OPN promoted fat storage in adipose tissue thereby preventing the obesity-induced shift to ectopic fat accumulation in the liver. Euglycaemic-hyperinsulinaemic clamp studies revealed that insulin resistance and excess hepatic glucose production in obesity were significantly attenuated in Opn (-/-) mice. OPN deficiency markedly improved hepatic insulin signalling as shown by enhanced insulin receptor substrate-2 phosphorylation and prevented upregulation of the major hepatic transcription factor Forkhead box O1 and its gluconeogenic target genes. In addition, obesity-driven hepatic inflammation and macrophage accumulation was blocked by OPN deficiency. CONCLUSIONS/INTERPRETATION: Our data strongly emphasise OPN as mediator of obesity-associated hepatic alterations including steatosis, inflammation, insulin resistance and excess gluconeogenesis. Targeting OPN action could therefore provide a novel therapeutic strategy to prevent obesity-related complications such as NAFLD and type 2 diabetes.


Assuntos
Fígado Gorduroso/etiologia , Fígado Gorduroso/genética , Fígado Gorduroso/metabolismo , Glucose/metabolismo , Obesidade/complicações , Obesidade/fisiopatologia , Osteopontina/deficiência , Animais , Técnica Clamp de Glucose , Immunoblotting , Imuno-Histoquímica , Imunoprecipitação , Masculino , Camundongos , Camundongos Knockout , Obesidade/genética , Obesidade/metabolismo , Osteopontina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Triglicerídeos/metabolismo
14.
Pathologe ; 31 Suppl 2: 147-52, 2010 Oct.
Artigo em Alemão | MEDLINE | ID: mdl-20835709

RESUMO

Following sequencing of the human genome there are new challenges to decipher the knowledge concerning gene function and the role of gene interactions and pathways leading to disease. Mouse models have proven to be an ideal tool for this purpose. Point mutations induced by chemical mutagenesis by N-ethyl-N-nitrosourea (ENU) offer possibilities for the analysis of the phenotypic outcome of a single base pair exchange on the entire organism. The Munich ENU mouse mutagenesis project is part of the worldwide efforts to obtain mutations for each gene. The generation of new alleles or allelic series offers relevant insights into the relevance of single gene sections. Various mouse models for human diseases have been generated by a systematic large-scale genome-wide phenotyping screen in the last decade. This work illustrates how the implementation of the ENU mouse mutagenesis project with gene identification and parallel high-throughput screening is taking advantage of local cooperation with experienced phenotyping groups at the Helmholtz Zentrum München, leading to major advances in the functional analysis of the mammalian genome.


Assuntos
Alquilantes/farmacologia , Modelos Animais de Doenças , Etilnitrosoureia/farmacologia , Camundongos Mutantes/genética , Mutação Puntual/genética , Alelos , Animais , Pareamento de Bases/genética , Doenças Genéticas Inatas , Genoma Humano , Estudo de Associação Genômica Ampla , Humanos , Camundongos , Mutagênese/genética , Fenótipo
15.
Am J Physiol Endocrinol Metab ; 298(3): E512-23, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19952346

RESUMO

Several mutant mouse models for human diseases such as diabetes mellitus have been generated in the large-scale Munich ENU (N-ethyl-N-nitrosourea) mouse mutagenesis project. The aim of this study was to identify the causal mutation of one of these strains and to characterize the resulting diabetic phenotype. Mutants exhibit a T to G transversion mutation at nt 629 in the glucokinase (Gck) gene, leading to an amino acid exchange from methionine to arginine at position 210. Adult Munich Gck(M210R) mutant mice demonstrated a significant reduction of hepatic glucokinase enzyme activity but equal glucokinase mRNA and protein abundances. While homozygous mutant mice exhibited growth retardation and died soon after birth in consequence of severe hyperglycemia, heterozygous mutant mice displayed only slightly elevated blood glucose levels, present from birth, with development of disturbed glucose tolerance and glucose-induced insulin secretion. Additionally, insulin sensitivity and fasting serum insulin levels were slightly reduced in male mutant mice from an age of 90 days onward. While beta-cell mass was unaltered in neonate heterozygous and homozygous mutant mice, the total islet and beta-cell volumes and the total volume of isolated beta-cells were significantly decreased in 210-day-old male, but not female heterozygous mutant mice despite undetectable apoptosis. These findings indicate that reduced total islet and beta-cell volumes of male mutants might emerge from disturbed postnatal islet neogenesis. Considering the lack of knowledge about the pathomorphology of maturity-onset diabetes of the young type 2 (MODY 2), this glucokinase mutant model of reduced total islet and total beta-cell volume provides the opportunity to elucidate the impact of a defective glucokinase on development and maintenance of beta-cell mass and its relevance in MODY 2 patients.


Assuntos
Diabetes Mellitus Tipo 2/patologia , Diabetes Mellitus Tipo 2/fisiopatologia , Modelos Animais de Doenças , Resistência à Insulina , Células Secretoras de Insulina/patologia , Animais , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Mutação
16.
Genes Brain Behav ; 9(3): 305-17, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20039949

RESUMO

Loss of function of DJ-1 (PARK7) is associated with autosomal recessive early-onset Parkinson's disease (PD), one of the major age-related neurological diseases. In this study, we extended former studies on DJ-1 knockout mice by identifying subtle morphological and behavioural phenotypes. The DJ-1 gene trap-induced null mutants exhibit less dopamine-producing neurons in the ventral tegmental area (VTA). They also exhibit slight changes in behaviour, i.e. diminished rearing behaviour and impairments in object recognition. Furthermore, we detected subtle phenotypes, which suggest that these animals compensate for the loss of DJ-1. First, we found a significant upregulation of mitochondrial respiratory enzyme activities, a mechanism known to protect against oxidative stress. Second, a close to significant increase in c-Jun N-terminal kinase 1 phosphorylation in old DJ-1-deficient mice hints at a differential activation of neuronal cell survival pathways. Third, as no change in the density of tyrosine hydroxylase (TH)-positive terminals in the striatum was observed, the remaining dopamine-producing neurons likely compensate by increasing axonal sprouting. In summary, the present data suggest that DJ-1 is implicated in major non-motor symptoms of PD appearing in the early phases of the disease-such as subtle impairments in motivated behaviour and cognition-and that under basal conditions the loss of DJ-1 is compensated.


Assuntos
Neurônios/metabolismo , Proteínas Oncogênicas/genética , Tirosina 3-Mono-Oxigenase/metabolismo , Área Tegmentar Ventral/metabolismo , Fatores Etários , Análise de Variância , Animais , Comportamento Animal/fisiologia , Western Blotting , Cromatografia Líquida de Alta Pressão , Dopamina/metabolismo , Feminino , Genótipo , Imuno-Histoquímica , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Masculino , Camundongos , Camundongos Knockout , Mitocôndrias/genética , Mitocôndrias/metabolismo , Proteína Quinase 8 Ativada por Mitógeno/genética , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Atividade Motora/genética , Proteínas Oncogênicas/metabolismo , Peroxirredoxinas , Fosforilação/genética , Proteína Desglicase DJ-1 , Reconhecimento Psicológico/fisiologia , Regulação para Cima/genética
17.
Br J Cancer ; 100(4): 656-62, 2009 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-19190631

RESUMO

Ras acts in signalling pathways regulating the activity of multiple cellular functions including cell proliferation, differentiation, and apoptosis. Amino-acid exchanges at position 12, 13, or 61 of the Kras gene convert the proto-oncogene into an activated oncogene. Until now, a direct comparison of genome-wide expression profiling studies of Kras overexpression and different Kras mutant forms in a single assay system has not been carried out. In our study, we focused on the direct comparison of global gene expression effects caused by mutations in codon 12 or 13 of the Kras gene and Kras overexpression in murine fibroblasts. We determined Kras cellular mRNA, Ras protein and activated Ras protein levels. Further, we compared our data to the proteome analysis of the same transfected cell lines. Both overexpression and mutations of Kras lead to common altered gene expression patterns. Only two genes, Lox and Col1a1, were reversely regulated in the Kras transfectants. They may contribute to the higher aggressiveness of the Kras codon 12 mutation in tumour progression. The functional annotation of differentially expressed genes revealed a high frequency of proteins involved in tumour growth and angiogenesis. These data further support the important role of these genes in tumour-associated angiogenesis.


Assuntos
Fibroblastos/metabolismo , Expressão Gênica , Mutação , Proteínas Proto-Oncogênicas/genética , Proteínas ras/genética , Animais , Linhagem Celular , Transformação Celular Neoplásica , Códon , Perfilação da Expressão Gênica , Humanos , Camundongos , Células NIH 3T3 , Fenótipo , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas p21(ras) , Proteínas ras/metabolismo
18.
Proc Natl Acad Sci U S A ; 106(9): 3354-9, 2009 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-19208810

RESUMO

Obesity is associated with increased risk for developing pancreatic cancer, and it is suggested that insulin resistance provides the missing link. Here we demonstrate that under the context of genetic susceptibility, a high fat diet (HFD) predisposes mice with oncogenic K-ras activation to accelerated pancreatic intraepithelial neoplasm (PanIN) development. Tumor promotion is closely associated with increased inflammation and abrogation of TNFR1 signaling significantly blocks this process underlining a central role for TNFalpha in obesity-mediated enhancement of PanIN lesions. Interestingly, however, despite increased TNFalpha levels, mice remain insulin sensitive. We show that, while aggravating tumor promotion, a HFD exerts dramatic changes in energy metabolism through enhancement of pancreatic exocrine insufficiency, metabolic rates, and expression of genes involved in mitochondrial fatty acid (FA) beta-oxidation that collectively contribute to improved glucose tolerance in these mice. While on one hand these findings provide significant evidence that obesity is linked to tumor promotion in the pancreas, on the other it suggests alterations in inflammatory responses and bioenergetic pathways as the potential underlying cause.


Assuntos
Ácidos Graxos/metabolismo , Mitocôndrias/metabolismo , Obesidade/metabolismo , Neoplasias Pancreáticas/metabolismo , Ração Animal , Animais , Progressão da Doença , Insuficiência Pancreática Exócrina/induzido quimicamente , Insuficiência Pancreática Exócrina/metabolismo , Insuficiência Pancreática Exócrina/patologia , Ácidos Graxos/farmacologia , Inflamação/genética , Inflamação/metabolismo , Resistência à Insulina , Camundongos , Mitocôndrias/efeitos dos fármacos , Oxirredução , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Fatores de Tempo , Proteínas ras/metabolismo
19.
Behav Genet ; 38(4): 396-406, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18461434

RESUMO

Transport of glucose into neuronal cells is predominantly mediated by the glucose transporters GLUT1 and GLUT3. In addition, GLUT8 is expressed in some regions of the brain. By in situ hybridization we detected GLUT8-mRNA in hippocampus, thalamus, and cortex. However, its cellular and physiological function is still unknown. Thus, GLUT8 knockout (Slc2a8 -/-) mice were used for a screening approach in the modified hole board (mHB) behavioral test to analyze the role of GLUT8 in the central nervous system. Slc2a8 -/- mice showed increased mean velocity, total distance traveled and performed more turns in the mHB test. This hyperactivity of Slc2a8 -/- mice was confirmed by monitoring locomotor activity in the home cage and voluntary activity in a running wheel. In addition, Slc2a8 -/- mice showed increased arousal as indicated by elevated defecation, reduced latency to the first defecation and a tendency to altered grooming. Furthermore, the mHB test gave evidence that Slc2a8 -/- mice exhibit a reduced risk assessment because they performed less rearings in an unprotected area and showed significantly reduced latency to stretched body posture. Our data suggest that behavioral alterations of Slc2a8 -/- mice are due to dysfunctions in neuronal processes presumably as a consequence of defects in the glucose metabolism.


Assuntos
Proteínas Facilitadoras de Transporte de Glucose/deficiência , Atividade Motora/genética , RNA Mensageiro/genética , Animais , Encéfalo/metabolismo , Encéfalo/fisiologia , Deleção de Genes , Glucose/metabolismo , Hibridização In Situ , Camundongos , Camundongos Knockout
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...