Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 150(6): 1287-99, 2012 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-22939713

RESUMO

Metabolic homeostasis is achieved by complex molecular and cellular networks that differ significantly among individuals and are difficult to model with genetically engineered lines of mice optimized to study single gene function. Here, we systematically acquired metabolic phenotypes by using the EUMODIC EMPReSS protocols across a large panel of isogenic but diverse strains of mice (BXD type) to study the genetic control of metabolism. We generated and analyzed 140 classical phenotypes and deposited these in an open-access web service for systems genetics (www.genenetwork.org). Heritability, influence of sex, and genetic modifiers of traits were examined singly and jointly by using quantitative-trait locus (QTL) and expression QTL-mapping methods. Traits and networks were linked to loci encompassing both known variants and novel candidate genes, including alkaline phosphatase (ALPL), here linked to hypophosphatasia. The assembled and curated phenotypes provide key resources and exemplars that can be used to dissect complex metabolic traits and disorders.


Assuntos
Modelos Animais de Doenças , Doenças Metabólicas/genética , Camundongos/genética , Fosfatase Alcalina/química , Fosfatase Alcalina/genética , Animais , Cruzamentos Genéticos , Feminino , Homeostase , Humanos , Hipofosfatasia/genética , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Polimorfismo Genético , Locos de Características Quantitativas , Padrões de Referência , Vitamina B 6/metabolismo
2.
PLoS Genet ; 16(1): e1008577, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31929527

RESUMO

Circadian systems provide a fitness advantage to organisms by allowing them to adapt to daily changes of environmental cues, such as light/dark cycles. The molecular mechanism underlying the circadian clock has been well characterized. However, how internal circadian clocks are entrained with regular daily light/dark cycles remains unclear. By collecting and analyzing indirect calorimetry (IC) data from more than 2000 wild-type mice available from the International Mouse Phenotyping Consortium (IMPC), we show that the onset time and peak phase of activity and food intake rhythms are reliable parameters for screening defects of circadian misalignment. We developed a machine learning algorithm to quantify these two parameters in our misalignment screen (SyncScreener) with existing datasets and used it to screen 750 mutant mouse lines from five IMPC phenotyping centres. Mutants of five genes (Slc7a11, Rhbdl1, Spop, Ctc1 and Oxtr) were found to be associated with altered patterns of activity or food intake. By further studying the Slc7a11tm1a/tm1a mice, we confirmed its advanced activity phase phenotype in response to a simulated jetlag and skeleton photoperiod stimuli. Disruption of Slc7a11 affected the intercellular communication in the suprachiasmatic nucleus, suggesting a defect in synchronization of clock neurons. Our study has established a systematic phenotype analysis approach that can be used to uncover the mechanism of circadian entrainment in mice.


Assuntos
Ritmo Circadiano/genética , Sistema y+ de Transporte de Aminoácidos/genética , Animais , Aprendizado de Máquina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Receptores de Ocitocina/genética , Proteínas Repressoras/genética , Serina Endopeptidases/genética , Proteínas de Ligação a Telômeros/genética , Complexos Ubiquitina-Proteína Ligase/genética
3.
PLoS Genet ; 16(12): e1009190, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33370286

RESUMO

The genetic landscape of diseases associated with changes in bone mineral density (BMD), such as osteoporosis, is only partially understood. Here, we explored data from 3,823 mutant mouse strains for BMD, a measure that is frequently altered in a range of bone pathologies, including osteoporosis. A total of 200 genes were found to significantly affect BMD. This pool of BMD genes comprised 141 genes with previously unknown functions in bone biology and was complementary to pools derived from recent human studies. Nineteen of the 141 genes also caused skeletal abnormalities. Examination of the BMD genes in osteoclasts and osteoblasts underscored BMD pathways, including vesicle transport, in these cells and together with in silico bone turnover studies resulted in the prioritization of candidate genes for further investigation. Overall, the results add novel pathophysiological and molecular insight into bone health and disease.


Assuntos
Densidade Óssea/genética , Regulação da Expressão Gênica/genética , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteoporose/genética , Animais , Feminino , Ontologia Genética , Pleiotropia Genética , Estudo de Associação Genômica Ampla , Genótipo , Masculino , Camundongos , Camundongos Transgênicos , Mutação , Osteoblastos/patologia , Osteoclastos/patologia , Osteoporose/metabolismo , Fenótipo , Regiões Promotoras Genéticas , Mapas de Interação de Proteínas , Caracteres Sexuais , Transcriptoma
4.
J Neurosci ; 41(22): 4910-4936, 2021 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-33888607

RESUMO

Spinocerebellar ataxia type 7 (SCA7) is an inherited neurodegenerative disease mainly characterized by motor incoordination because of progressive cerebellar degeneration. SCA7 is caused by polyglutamine expansion in ATXN7, a subunit of the transcriptional coactivator SAGA, which harbors histone modification activities. Polyglutamine expansions in specific proteins are also responsible for SCA1-SCA3, SCA6, and SCA17; however, the converging and diverging pathomechanisms remain poorly understood. Using a new SCA7 knock-in mouse, SCA7140Q/5Q, we analyzed gene expression in the cerebellum and assigned gene deregulation to specific cell types using published datasets. Gene deregulation affects all cerebellar cell types, although at variable degree, and correlates with alterations of SAGA-dependent epigenetic marks. Purkinje cells (PCs) are by far the most affected neurons and show reduced expression of 83 cell-type identity genes, including these critical for their spontaneous firing activity and synaptic functions. PC gene downregulation precedes morphologic alterations, pacemaker dysfunction, and motor incoordination. Strikingly, most PC genes downregulated in SCA7 have also decreased expression in SCA1 and SCA2 mice, revealing converging pathomechanisms and a common disease signature involving cGMP-PKG and phosphatidylinositol signaling pathways and LTD. Our study thus points out molecular targets for therapeutic development, which may prove beneficial for several SCAs. Furthermore, we show that SCA7140Q/5Q males and females exhibit the major disease features observed in patients, including cerebellar damage, cerebral atrophy, peripheral nerves pathology, and photoreceptor dystrophy, which account for progressive impairment of behavior, motor, and visual functions. SCA7140Q/5Q mice represent an accurate model for the investigation of different aspects of SCA7 pathogenesis.SIGNIFICANCE STATEMENT Spinocerebellar ataxia 7 (SCA7) is one of the several forms of inherited SCAs characterized by cerebellar degeneration because of polyglutamine expansion in specific proteins. The ATXN7 involved in SCA7 is a subunit of SAGA transcriptional coactivator complex. To understand the pathomechanisms of SCA7, we determined the cell type-specific gene deregulation in SCA7 mouse cerebellum. We found that the Purkinje cells are the most affected cerebellar cell type and show downregulation of a large subset of neuronal identity genes, critical for their spontaneous firing and synaptic functions. Strikingly, the same Purkinje cell genes are downregulated in mouse models of two other SCAs. Thus, our work reveals a disease signature shared among several SCAs and uncovers potential molecular targets for their treatment.


Assuntos
Cerebelo/patologia , Células de Purkinje/patologia , Ataxias Espinocerebelares/patologia , Animais , Regulação para Baixo , Feminino , Técnicas de Introdução de Genes , Masculino , Camundongos , Transcriptoma
5.
Am J Physiol Endocrinol Metab ; 322(6): E517-E527, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35403438

RESUMO

Insulin resistance is a major public health burden that often results in other comorbidities including type 2 diabetes, nonalcoholic fatty liver disease (NAFLD), and cardiovascular disease. An insulin sensitizer has the potential to become a disease-modifying therapy. It remains an unmet medical need to identify therapeutics that target the insulin signaling pathway to treat insulin resistance. Low-molecular-weight protein tyrosine phosphatase (LMPTP) negatively regulates insulin signaling and has emerged as a potential therapeutic target for insulin sensitization. Genetic studies have demonstrated that LMPTP is positively associated with obesity in humans and promotes insulin resistance in rodents. A recent study showed that pharmacological inhibition or genetic deletion of LMPTP protects mice from high-fat diet-induced insulin resistance and diabetes. Here, we show that loss of LMPTP by genetic deletion has no significant effects on improving glucose tolerance in lean or diet-induced obese mice. Furthermore, our data demonstrate that LMPTP deficiency potentiates cardiac hypertrophy that leads to mild cardiac dysfunction. Our findings suggest that the development of LMPTP inhibitors for the treatment of insulin resistance and type 2 diabetes should be reevaluated, and further studies are needed to characterize the molecular and pathophysiological role of LMPTP.NEW & NOTEWORTHY Inhibition of LMPTP with a small-molecule inhibitor, Cmpd23, improves glucose tolerance in mice as reported earlier. However, genetic deficiency of the LMPTP-encoding gene, Acp1, has limited effects on glucose metabolism but leads to mild cardiac hypertrophy in mice. The findings suggest the potential off-target effects of Cmpd23 and call for reevaluation of LMPTP as a therapeutic target for the treatment of insulin resistance and type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2 , Resistência à Insulina , Animais , Cardiomegalia/genética , Cardiomegalia/metabolismo , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/genética , Dieta Hiperlipídica , Glucose/metabolismo , Insulina/metabolismo , Resistência à Insulina/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Tirosina Fosfatases/uso terapêutico , Magreza
6.
Mol Psychiatry ; 26(11): 6336-6349, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34050326

RESUMO

Microglia play a critical role in maintaining neural function. While microglial activity follows a circadian rhythm, it is not clear how this intrinsic clock relates to their function, especially in stimulated conditions such as in the control of systemic energy homeostasis or memory formation. In this study, we found that microglia-specific knock-down of the core clock gene, Bmal1, resulted in increased microglial phagocytosis in mice subjected to high-fat diet (HFD)-induced metabolic stress and likewise among mice engaged in critical cognitive processes. Enhanced microglial phagocytosis was associated with significant retention of pro-opiomelanocortin (POMC)-immunoreactivity in the mediobasal hypothalamus in mice on a HFD as well as the formation of mature spines in the hippocampus during the learning process. This response ultimately protected mice from HFD-induced obesity and resulted in improved performance on memory tests. We conclude that loss of the rigorous control implemented by the intrinsic clock machinery increases the extent to which microglial phagocytosis can be triggered by neighboring neurons under metabolic stress or during memory formation. Taken together, microglial responses associated with loss of Bmal1 serve to ensure a healthier microenvironment for neighboring neurons in the setting of an adaptive response. Thus, microglial Bmal1 may be an important therapeutic target for metabolic and cognitive disorders with relevance to psychiatric disease.


Assuntos
Fatores de Transcrição ARNTL , Dieta Hiperlipídica , Memória , Microglia , Obesidade , Fatores de Transcrição ARNTL/genética , Fatores de Transcrição ARNTL/metabolismo , Animais , Ritmo Circadiano/fisiologia , Dieta Hiperlipídica/efeitos adversos , Técnicas de Silenciamento de Genes , Hipocampo/metabolismo , Hipocampo/fisiologia , Aprendizagem/fisiologia , Memória/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Obesidade/etiologia , Obesidade/genética , Obesidade/metabolismo , Obesidade/prevenção & controle , Fagocitose/fisiologia , Pró-Opiomelanocortina/metabolismo , Estresse Fisiológico/fisiologia
7.
Bioinformatics ; 36(5): 1492-1500, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31591642

RESUMO

MOTIVATION: High-throughput phenomic projects generate complex data from small treatment and large control groups that increase the power of the analyses but introduce variation over time. A method is needed to utlize a set of temporally local controls that maximizes analytic power while minimizing noise from unspecified environmental factors. RESULTS: Here we introduce 'soft windowing', a methodological approach that selects a window of time that includes the most appropriate controls for analysis. Using phenotype data from the International Mouse Phenotyping Consortium (IMPC), adaptive windows were applied such that control data collected proximally to mutants were assigned the maximal weight, while data collected earlier or later had less weight. We applied this method to IMPC data and compared the results with those obtained from a standard non-windowed approach. Validation was performed using a resampling approach in which we demonstrate a 10% reduction of false positives from 2.5 million analyses. We applied the method to our production analysis pipeline that establishes genotype-phenotype associations by comparing mutant versus control data. We report an increase of 30% in significant P-values, as well as linkage to 106 versus 99 disease models via phenotype overlap with the soft-windowed and non-windowed approaches, respectively, from a set of 2082 mutant mouse lines. Our method is generalizable and can benefit large-scale human phenomic projects such as the UK Biobank and the All of Us resources. AVAILABILITY AND IMPLEMENTATION: The method is freely available in the R package SmoothWin, available on CRAN http://CRAN.R-project.org/package=SmoothWin. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Saúde da População , Software , Animais , Estudos de Associação Genética , Humanos , Camundongos , Fenótipo
8.
PLoS Genet ; 12(2): e1005709, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26872257

RESUMO

The 16p11.2 600 kb BP4-BP5 deletion and duplication syndromes have been associated with developmental delay; autism spectrum disorders; and reciprocal effects on the body mass index, head circumference and brain volumes. Here, we explored these relationships using novel engineered mouse models carrying a deletion (Del/+) or a duplication (Dup/+) of the Sult1a1-Spn region homologous to the human 16p11.2 BP4-BP5 locus. On a C57BL/6N inbred genetic background, Del/+ mice exhibited reduced weight and impaired adipogenesis, hyperactivity, repetitive behaviors, and recognition memory deficits. In contrast, Dup/+ mice showed largely opposite phenotypes. On a F1 C57BL/6N × C3B hybrid genetic background, we also observed alterations in social interaction in the Del/+ and the Dup/+ animals, with other robust phenotypes affecting recognition memory and weight. To explore the dosage effect of the 16p11.2 genes on metabolism, Del/+ and Dup/+ models were challenged with high fat and high sugar diet, which revealed opposite energy imbalance. Transcriptomic analysis revealed that the majority of the genes located in the Sult1a1-Spn region were sensitive to dosage with a major effect on several pathways associated with neurocognitive and metabolic phenotypes. Whereas the behavioral consequence of the 16p11 region genetic dosage was similar in mice and humans with activity and memory alterations, the metabolic defects were opposite: adult Del/+ mice are lean in comparison to the human obese phenotype and the Dup/+ mice are overweight in comparison to the human underweight phenotype. Together, these data indicate that the dosage imbalance at the 16p11.2 locus perturbs the expression of modifiers outside the CNV that can modulate the penetrance, expressivity and direction of effects in both humans and mice.


Assuntos
Deleção Cromossômica , Duplicação Cromossômica/genética , Cognição , Adiposidade , Alelos , Animais , Arilsulfotransferase/genética , Arilsulfotransferase/metabolismo , Comportamento Animal , Peso Corporal , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Cromossomos de Mamíferos/genética , Anormalidades Craniofaciais/genética , Dieta Hiperlipídica , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Rearranjo Gênico/genética , Hipocampo/fisiopatologia , Memória , Camundongos Endogâmicos C57BL , Atividade Motora , Fenótipo , Transmissão Sináptica/genética , Síndrome , Desmame
9.
PLoS Genet ; 12(3): e1005898, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26938916

RESUMO

BAHD1 is a vertebrate protein that promotes heterochromatin formation and gene repression in association with several epigenetic regulators. However, its physiological roles remain unknown. Here, we demonstrate that ablation of the Bahd1 gene results in hypocholesterolemia, hypoglycemia and decreased body fat in mice. It also causes placental growth restriction with a drop of trophoblast glycogen cells, a reduction of fetal weight and a high neonatal mortality rate. By intersecting transcriptome data from murine Bahd1 knockout (KO) placentas at stages E16.5 and E18.5 of gestation, Bahd1-KO embryonic fibroblasts, and human cells stably expressing BAHD1, we also show that changes in BAHD1 levels alter expression of steroid/lipid metabolism genes. Biochemical analysis of the BAHD1-associated multiprotein complex identifies MIER proteins as novel partners of BAHD1 and suggests that BAHD1-MIER interaction forms a hub for histone deacetylases and methyltransferases, chromatin readers and transcription factors. We further show that overexpression of BAHD1 leads to an increase of MIER1 enrichment on the inactive X chromosome (Xi). In addition, BAHD1 and MIER1/3 repress expression of the steroid hormone receptor genes ESR1 and PGR, both playing important roles in placental development and energy metabolism. Moreover, modulation of BAHD1 expression in HEK293 cells triggers epigenetic changes at the ESR1 locus. Together, these results identify BAHD1 as a core component of a chromatin-repressive complex regulating placental morphogenesis and body fat storage and suggest that its dysfunction may contribute to several human diseases.


Assuntos
Proteínas Cromossômicas não Histona/genética , Proteínas Nucleares/genética , Placentação/genética , Esteroides/metabolismo , Fatores de Transcrição/genética , Animais , Cromatina/genética , Proteínas Cromossômicas não Histona/biossíntese , Proteínas de Ligação a DNA , Receptor alfa de Estrogênio/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Células HEK293 , Humanos , Camundongos , Camundongos Knockout , Proteínas Nucleares/biossíntese , Placenta/metabolismo , Gravidez , Fatores de Transcrição/biossíntese , Transcriptoma/genética
10.
Proc Natl Acad Sci U S A ; 112(48): E6691-8, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26627260

RESUMO

The light-entrained master central circadian clock (CC) located in the suprachiasmatic nucleus (SCN) not only controls the diurnal alternance of the active phase (the light period of the human light-dark cycle, but the mouse dark period) and the rest phase (the human dark period, but the mouse light period), but also synchronizes the ubiquitous peripheral CCs (PCCs) with these phases to maintain homeostasis. We recently elucidated in mice the molecular signals through which metabolic alterations induced on an unusual feeding schedule, taking place during the rest phase [i.e., restricted feeding (RF)], creates a 12-h PCC shift. Importantly, a previous study showed that the SCN CC is unaltered during RF, which creates a misalignment between the RF-shifted PCCs and the SCN CC-controlled phases of activity and rest. However, the molecular basis of SCN CC insensitivity to RF and its possible pathological consequences are mostly unknown. Here we deciphered, at the molecular level, how RF creates this misalignment. We demonstrate that the PPARα and glucagon receptors, the two instrumental transducers in the RF-induced shift of PCCs, are not expressed in the SCN, thereby preventing on RF a shift of the master SCN CC and creating the misalignment. Most importantly, this RF-induced misalignment leads to a misexpression (with respect to their normal physiological phase of expression) of numerous CC-controlled homeostatic genes, which in the long term generates in RF mice a number of metabolic pathologies including diabetes, obesity, and metabolic syndrome, which have been reported in humans engaged in shift work schedules.


Assuntos
Ritmo Circadiano , Comportamento Alimentar , Síndrome Metabólica/metabolismo , Proteínas Circadianas Period/metabolismo , Animais , Relógios Circadianos/fisiologia , Diabetes Mellitus/metabolismo , Modelos Animais de Doenças , Ingestão de Alimentos/fisiologia , Regulação da Expressão Gênica , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Hipercolesterolemia/metabolismo , Hipertrigliceridemia/metabolismo , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Fotoperíodo , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Núcleo Supraquiasmático/fisiologia , Fatores de Tempo , Tolerância ao Trabalho Programado
11.
J Biol Chem ; 291(45): 23428-23439, 2016 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-27621313

RESUMO

Mutations of the AMP-activated kinase gamma 2 subunit (AMPKγ2), N488I (AMPKγ2NI) and R531G (AMPKγ2RG), are associated with Wolff-Parkinson-White (WPW) syndrome, a cardiac disorder characterized by ventricular pre-excitation in humans. Cardiac-specific transgenic overexpression of human AMPKγ2NI or AMPKγ2RG leads to constitutive AMPK activation and the WPW phenotype in mice. However, overexpression of these mutant proteins also caused profound, non-physiological increase in cardiac glycogen, which might abnormally alter the true phenotype. To investigate whether physiological levels of AMPKγ2NI or AMPKγ2RG mutation cause WPW syndrome and metabolic changes in other organs, we generated two knock-in mouse lines on the C57BL/6N background harboring mutations of human AMPKγ2NI and AMPKγ2RG, respectively. Similar to the reported phenotypes of mice overexpressing AMPKγ2NI or AMPKγ2RG in the heart, both lines developed WPW syndrome and cardiac hypertrophy; however, these effects were independent of cardiac glycogen accumulation. Compared with AMPKγ2WT mice, AMPKγ2NI and AMPKγ2RG mice exhibited reduced body weight, fat mass, and liver steatosis when fed with a high fat diet (HFD). Surprisingly, AMPKγ2RG but not AMPKγ2NI mice fed with an HFD exhibited severe kidney injury characterized by glycogen accumulation, inflammation, apoptosis, cyst formation, and impaired renal function. These results demonstrate that expression of AMPKγ2NI and AMPKγ2RG mutations at physiological levels can induce beneficial metabolic effects but that this is accompanied by WPW syndrome. Our data also reveal an unexpected effect of AMPKγ2RG in the kidney, linking lifelong constitutive activation of AMPK to a potential risk for kidney dysfunction in the context of an HFD.


Assuntos
Proteínas Quinases Ativadas por AMP/genética , Mutação , Insuficiência Renal/genética , Síndrome de Wolff-Parkinson-White/genética , Animais , Apoptose , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Inflamação/genética , Inflamação/patologia , Rim/metabolismo , Rim/patologia , Masculino , Camundongos Endogâmicos C57BL , Insuficiência Renal/patologia , Síndrome de Wolff-Parkinson-White/patologia
12.
Nature ; 452(7186): 429-35, 2008 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-18344982

RESUMO

Identifying variations in DNA that increase susceptibility to disease is one of the primary aims of genetic studies using a forward genetics approach. However, identification of disease-susceptibility genes by means of such studies provides limited functional information on how genes lead to disease. In fact, in most cases there is an absence of functional information altogether, preventing a definitive identification of the susceptibility gene or genes. Here we develop an alternative to the classic forward genetics approach for dissecting complex disease traits where, instead of identifying susceptibility genes directly affected by variations in DNA, we identify gene networks that are perturbed by susceptibility loci and that in turn lead to disease. Application of this method to liver and adipose gene expression data generated from a segregating mouse population results in the identification of a macrophage-enriched network supported as having a causal relationship with disease traits associated with metabolic syndrome. Three genes in this network, lipoprotein lipase (Lpl), lactamase beta (Lactb) and protein phosphatase 1-like (Ppm1l), are validated as previously unknown obesity genes, strengthening the association between this network and metabolic disease traits. Our analysis provides direct experimental support that complex traits such as obesity are emergent properties of molecular networks that are modulated by complex genetic loci and environmental factors.


Assuntos
Redes Reguladoras de Genes/genética , Predisposição Genética para Doença/genética , Variação Genética/genética , Síndrome Metabólica/genética , Obesidade/genética , Tecido Adiposo/metabolismo , Animais , Apolipoproteína A-II/genética , Cromossomos de Mamíferos/genética , Feminino , Desequilíbrio de Ligação , Lipase Lipoproteica/genética , Fígado/metabolismo , Escore Lod , Macrófagos/metabolismo , Masculino , Proteínas de Membrana/genética , Síndrome Metabólica/enzimologia , Síndrome Metabólica/metabolismo , Camundongos , Obesidade/enzimologia , Obesidade/metabolismo , Fenótipo , Fosfoproteínas Fosfatases/deficiência , Fosfoproteínas Fosfatases/genética , Fosfoproteínas Fosfatases/metabolismo , Locos de Características Quantitativas , Reprodutibilidade dos Testes , Proteínas Ribossômicas/genética
13.
J Neurosci ; 32(6): 1962-8, 2012 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-22323709

RESUMO

Vesicular (v)- and target (t)-SNARE proteins assemble in SNARE complex to mediate membrane fusion. Tetanus neurotoxin-insensitive vesicular-associated membrane protein (TI-VAMP/VAMP7), a vesicular SNARE expressed in several cell types including neurons, was previously shown to play a major role in exocytosis involved in neurite growth in cultured neurons. Here we generated a complete constitutive knock-out by deleting the exon 3 of Vamp7. Loss of TI-VAMP expression did not lead to any striking developmental or neurological defect. Knock-out mice displayed decreased brain weight and increased third ventricle volume. Axon growth appeared normal in cultured knock-out neurons. Behavioral characterization unraveled that TI-VAMP knock-out was associated with increased anxiety. Our results thus suggest compensatory mechanisms allowing the TI-VAMP knock-out mice to fulfill major developmental processes. The phenotypic traits unraveled here further indicate an unexpected role of TI-VAMP-mediated vesicular traffic in anxiety and suggest a role for TI-VAMP in higher brain functions.


Assuntos
Ansiedade/genética , Metaloendopeptidases , Proteínas R-SNARE/deficiência , Proteínas R-SNARE/genética , Toxina Tetânica , Animais , Ansiedade/etiologia , Ansiedade/psicologia , Células COS , Células Cultivadas , Chlorocebus aethiops , Masculino , Metaloendopeptidases/administração & dosagem , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Coelhos , Toxina Tetânica/administração & dosagem
14.
Cell Metab ; 3(3): 211-22, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16517408

RESUMO

Signaling cascades that control adipogenesis are essential in the regulation of body weight and obesity. The adaptor p62 controls pathways that modulate cell differentiation. We report here that p62(-/-) mice develop mature-onset obesity, leptin resistance, as well as impaired glucose and insulin intolerance. The metabolic rate was significantly reduced in p62(-/-) nonobese mice, which displayed increased mRNA levels of PPAR-gamma and reduced levels of UCP-1 in adipose tissue. Basal activity of ERK was enhanced in fat from nonobese mutant mice. Embryo fibroblasts from p62(-/-) mice differentiated better than the wild-type controls into adipocytes, which was abrogated by pharmacological inhibition of the ERK pathway. p62 is induced during adipocyte differentiation and inhibits ERK activation by direct interaction. We propose that p62 normally antagonizes basal ERK activity and adipocyte differentiation and that its loss leads to the hyperactivation of ERK that favors adipogenesis and obesity.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Resistência à Insulina , Transdução de Sinais , Fatores de Transcrição/deficiência , Fatores de Transcrição/metabolismo , Células 3T3-L1 , Adipócitos/citologia , Adipogenia , Tecido Adiposo/citologia , Animais , Embrião de Mamíferos/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Fibroblastos/metabolismo , Células HeLa , Humanos , Camundongos , Camundongos Knockout , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Transcrição TFIIH , Fatores de Transcrição/genética
15.
Biochim Biophys Acta ; 1801(12): 1349-60, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20817122

RESUMO

The purinergic receptor P2Y(13) has been shown to play a role in the uptake of holo-HDL particles in in vitro hepatocyte experiments. In order to determine the role of P2Y(13) in lipoprotein metabolism in vivo, we ablated the expression of this gene in mice. Here we show that P2Y(13) knockout mice have lower fecal concentrations of neutral sterols (-27%±2.1% in males) as well as small decreases in plasma HDL (-13.1%±3.2% in males; -17.5%±4.0% in females) levels. In addition, significant decreases were detected in serum levels of fatty acids and glycerol in female P2Y(13) knockout mice. Hepatic mRNA profiling analyses showed increased expression of SREBP-regulated cholesterol and fatty acid biosynthesis genes, while fatty acid ß-oxidation genes were significantly decreased. Liver gene signatures also identified changes in PPARα-regulated transcript levels. With the exception of a small increase in bone area, P2Y(13) knockout mice do not show any additional major abnormalities, and display normal body weight, fat mass and lean body mass. No changes in insulin sensitivity and oral glucose tolerance could be detected. Taken together, our experiments assess a role for the purinergic receptor P2Y(13) in the regulation of lipoprotein metabolism and demonstrate that modulating its activity could be of benefit to the treatment of dyslipidemia in people.


Assuntos
Lipoproteínas/metabolismo , Receptores Purinérgicos P2/fisiologia , Animais , Feminino , Perfilação da Expressão Gênica , Fígado/metabolismo , Masculino , Camundongos , Camundongos Knockout , RNA Mensageiro/genética , Receptores Purinérgicos P2/genética
16.
Arch Cardiovasc Dis ; 111(1): 41-52, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29113787

RESUMO

BACKGROUND: Metabolic syndrome is a combination of symptoms including obesity, dyslipidaemia, glucose intolerance and hypertension. Oxidative stress appears to be a pathophysiological factor that links these signs and encourages progression towards heart failure and diabetes. Nox4 is a hydrogen peroxide nicotinamide adenine dinucleotide phosphate (NADPH) oxidase isoform - found in various cardiovascular cells and tissues, but also in tissues such as the liver - which is involved in glucose and lipid homeostasis. AIMS: To test whether inhibition of the Nox4 enzyme could improve blood pressure and metabolic parameters in mice receiving either angiotensin II or a high-fat diet. METHODS: Systolic and diastolic arterial pressures, pulse rate and heart rate were obtained in 24 male mice (12 wild-type [WT] and 12 Nox4-/-) before and during 14 days of angiotensin II infusion. After angiotensin II infusion, cardiac histological remodeling was assessed. Weight and biochemical parameters were measured in 18 male and 18 female mice (nine WT and nine Nox4-/- per gender) after 10 weeks on a standard chow diet, then 15 weeks on a high-fat diet. Glucose tolerance and insulin sensitivity were tested at age 25 weeks. RESULTS: Knock-out animals did not demonstrate a baseline blood pressure phenotype, but blocking Nox4 protected against angiotensin II-mediated arterial and pulse pressure increases. No protection against angiotensin II-induced cardiac fibrosis was observed. From a metabolic point of view, Nox4 inhibition reduced plasma triglycerides in male and female mice under a chow diet. However, Nox4 deletion did not affect the metabolic profile under a high-fat diet in males or females, but increased glucose intolerance in females. CONCLUSION: Our data identify Nox4 as a key source of radical oxygen species involved in hypertension and some metabolic problems.


Assuntos
Pressão Sanguínea , Hipertensão/enzimologia , Síndrome Metabólica/enzimologia , NADPH Oxidase 4/deficiência , Angiotensina II , Animais , Biomarcadores/sangue , Glicemia/metabolismo , Pressão Sanguínea/genética , Cardiomegalia/induzido quimicamente , Cardiomegalia/enzimologia , Cardiomegalia/fisiopatologia , Dieta Hiperlipídica , Modelos Animais de Doenças , Feminino , Fibrose , Predisposição Genética para Doença , Frequência Cardíaca , Hipertensão/induzido quimicamente , Hipertensão/genética , Hipertensão/fisiopatologia , Masculino , Síndrome Metabólica/sangue , Síndrome Metabólica/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocárdio/enzimologia , Miocárdio/patologia , NADPH Oxidase 4/genética , Fenótipo , Espécies Reativas de Oxigênio/metabolismo , Fatores de Tempo , Triglicerídeos/sangue , Remodelação Ventricular
17.
Nat Commun ; 9(1): 288, 2018 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-29348434

RESUMO

Metabolic diseases are a worldwide problem but the underlying genetic factors and their relevance to metabolic disease remain incompletely understood. Genome-wide research is needed to characterize so-far unannotated mammalian metabolic genes. Here, we generate and analyze metabolic phenotypic data of 2016 knockout mouse strains under the aegis of the International Mouse Phenotyping Consortium (IMPC) and find 974 gene knockouts with strong metabolic phenotypes. 429 of those had no previous link to metabolism and 51 genes remain functionally completely unannotated. We compared human orthologues of these uncharacterized genes in five GWAS consortia and indeed 23 candidate genes are associated with metabolic disease. We further identify common regulatory elements in promoters of candidate genes. As each regulatory element is composed of several transcription factor binding sites, our data reveal an extensive metabolic phenotype-associated network of co-regulated genes. Our systematic mouse phenotype analysis thus paves the way for full functional annotation of the genome.


Assuntos
Metabolismo Basal/genética , Glicemia/metabolismo , Peso Corporal/genética , Diabetes Mellitus Tipo 2/genética , Obesidade/genética , Consumo de Oxigênio/genética , Triglicerídeos/metabolismo , Animais , Área Sob a Curva , Redes Reguladoras de Genes , Estudo de Associação Genômica Ampla , Ensaios de Triagem em Larga Escala , Humanos , Doenças Metabólicas/genética , Camundongos , Camundongos Knockout , Fenótipo
18.
Sci Rep ; 7(1): 9618, 2017 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-28851918

RESUMO

ATP6AP2 codes for the (pro)renin receptor and is an essential component of vacuolar H+ ATPase. Activating (pro)renin for conversion of Angiotensinogen to Angiotensin makes ATP6AP2 attractive for drug intervention. Tissue-specific ATP6AP2 inactivation in mouse suggested a strong impact on various organs. Consistent with this, we found that embryonic ablation of Atp6ap2 resulted in both male hemizygous lethality and female haploinsufficiency. Next, we examined the phenotype of an induced inactivation in the adult animal, most akin to detect potential effect of functional interference of ATP6AP2 through drug therapy. Induced ablation of Atp6ap2, even without equal efficiency in all tissues (aorta, brain and kidney), resulted in rapid lethality marked by weight loss, changes in nutritional as well as blood parameters, leukocyte depletion, and bone marrow hypoplasia. Upon Atp6ap2 ablation, the colon demonstrated a rapid disruption of crypt morphology, aberrant proliferation, cell-death activation, as well as generation of microadenomas. Consequently, disruption of ATP6AP2 is extremely poorly tolerated in the adult, and severely affects various organ systems demonstrating that ATP6AP2 is an essential gene implicated in basic cellular mechanisms and necessary for multiple organ function. Accordingly, any potential drug targeting of this gene product must be strictly assessed for safety.


Assuntos
Insuficiência de Múltiplos Órgãos/mortalidade , Insuficiência de Múltiplos Órgãos/patologia , ATPases Translocadoras de Prótons/deficiência , Receptores de Superfície Celular/deficiência , Animais , Técnicas de Inativação de Genes , Camundongos , Análise de Sobrevida
19.
Neuron ; 93(2): 331-347, 2017 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-28065649

RESUMO

Fragile X-associated tremor/ataxia syndrome (FXTAS) is a neurodegenerative disorder caused by a limited expansion of CGG repeats in the 5' UTR of FMR1. Two mechanisms are proposed to cause FXTAS: RNA gain-of-function, where CGG RNA sequesters specific proteins, and translation of CGG repeats into a polyglycine-containing protein, FMRpolyG. Here we developed transgenic mice expressing CGG repeat RNA with or without FMRpolyG. Expression of FMRpolyG is pathogenic, while the sole expression of CGG RNA is not. FMRpolyG interacts with the nuclear lamina protein LAP2ß and disorganizes the nuclear lamina architecture in neurons differentiated from FXTAS iPS cells. Finally, expression of LAP2ß rescues neuronal death induced by FMRpolyG. Overall, these results suggest that translation of expanded CGG repeats into FMRpolyG alters nuclear lamina architecture and drives pathogenesis in FXTAS.


Assuntos
Ataxia/genética , Proteínas de Ligação a DNA/metabolismo , Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/genética , Proteínas de Membrana/metabolismo , Lâmina Nuclear/metabolismo , Peptídeos/genética , Biossíntese de Proteínas , RNA Mensageiro/metabolismo , Tremor/genética , Expansão das Repetições de Trinucleotídeos/genética , Animais , Ataxia/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Síndrome do Cromossomo X Frágil/metabolismo , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Lâmina Nuclear/patologia , Peptídeos/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Tremor/metabolismo
20.
Neuropsychopharmacology ; 31(8): 1733-44, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16237385

RESUMO

Exposure to stress triggers hormonal and behavioral responses. It has been shown that the endogenous opioid system plays a role in some physiological reactions to stress. The opioid system was described to mediate analgesia induced by mild stressors and to modulate the activation of the hypothalamic-pituitary-adrenal axis. Our study assessed the contribution of opioid receptors in stress-induced analgesia and adrenocorticotropic hormone (ACTH) and corticosterone release by a genetic approach. We performed a parallel analysis of mice deficient in mu, delta, or kappa opioid receptors, as well as of triple opioid receptor knockout mice, following exposure to a mild stress (3-min swim at 32 degrees C). In wild-type mice, stress elicited an increase in jumping latency on the hot plate, which was influenced by gender and genetic background. This analgesic response was reversed both by naloxone and by the triple mutation, and decreased in mu and delta opioid receptor knockout females. In wild-type females, stress also delayed front- and hindpaw behaviors in the hot plate test and increased tail-flick latency in the tail immersion test. Opioid receptor deletion however did not affect these stress responses. In addition, stress produced an increase in ACTH and corticosterone plasma levels. This endocrine response remained unchanged in all mutant strains. Therefore our data indicate that, under our stress conditions, the endogenous opioid system is recruited to produce some analgesia whereas it does not influence hypothalamic-pituitary-adrenal axis activity. This implies that brain circuits mediating analgesic and hormonal responses to stress can be dissociated.


Assuntos
Hormônio Adrenocorticotrópico/metabolismo , Analgesia/métodos , Corticosterona/metabolismo , Receptores Opioides/deficiência , Receptores Opioides/metabolismo , Estresse Fisiológico/metabolismo , Natação , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Medição da Dor/métodos , Tempo de Reação/fisiologia , Estresse Fisiológico/psicologia , Natação/psicologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA