Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Cell ; 164(3): 353-64, 2016 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-26824653

RESUMO

More than one-half billion people are obese, and despite progress in genetic research, much of the heritability of obesity remains enigmatic. Here, we identify a Trim28-dependent network capable of triggering obesity in a non-Mendelian, "on/off" manner. Trim28(+/D9) mutant mice exhibit a bi-modal body-weight distribution, with isogenic animals randomly emerging as either normal or obese and few intermediates. We find that the obese-"on" state is characterized by reduced expression of an imprinted gene network including Nnat, Peg3, Cdkn1c, and Plagl1 and that independent targeting of these alleles recapitulates the stochastic bi-stable disease phenotype. Adipose tissue transcriptome analyses in children indicate that humans too cluster into distinct sub-populations, stratifying according to Trim28 expression, transcriptome organization, and obesity-associated imprinted gene dysregulation. These data provide evidence of discrete polyphenism in mouse and man and thus carry important implications for complex trait genetics, evolution, and medicine.


Assuntos
Epigênese Genética , Haploinsuficiência , Proteínas Nucleares/genética , Obesidade/genética , Proteínas Repressoras/genética , Magreza/genética , Adolescente , Animais , Índice de Massa Corporal , Criança , Pré-Escolar , Humanos , Camundongos , Inquéritos Nutricionais , Polimorfismo Genético , Proteína 28 com Motivo Tripartido
2.
Proc Natl Acad Sci U S A ; 114(35): 9421-9426, 2017 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-28811369

RESUMO

An intergenic region of human chromosome 2 (2p25.3) harbors genetic variants which are among those most strongly and reproducibly associated with obesity. The gene closest to these variants is TMEM18, although the molecular mechanisms mediating these effects remain entirely unknown. Tmem18 expression in the murine hypothalamic paraventricular nucleus (PVN) was altered by changes in nutritional state. Germline loss of Tmem18 in mice resulted in increased body weight, which was exacerbated by high fat diet and driven by increased food intake. Selective overexpression of Tmem18 in the PVN of wild-type mice reduced food intake and also increased energy expenditure. We provide evidence that TMEM18 has four, not three, transmembrane domains and that it physically interacts with key components of the nuclear pore complex. Our data support the hypothesis that TMEM18 itself, acting within the central nervous system, is a plausible mediator of the impact of adjacent genetic variation on human adiposity.


Assuntos
Apetite/genética , Peso Corporal/genética , Proteínas de Membrana/metabolismo , Obesidade/genética , Animais , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Núcleo Hipotalâmico Paraventricular/metabolismo , Proteínas de Transporte Vesicular
3.
Neuroendocrinology ; 109(3): 200-207, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30261489

RESUMO

Haploinsufficiency occurs when loss of one copy of a diploid gene (hemizygosity) causes a phenotype. It is relatively rare, in that most genes can produce sufficient mRNA and protein from a single copy to prevent any loss of normal activity and function. Reproduction is a complex process relying on migration of GnRH neurons from the olfactory placode to the hypothalamus during development. We have studied 3 different homeodomain genes Otx2, Vax1, and Six3 and found that the deletion of one allele for any of these genes in mice produces subfertility or infertility in one or both sexes, despite the presence of one intact allele. All 3 heterozygous mice have reduced numbers of GnRH neurons, but the mechanisms of subfertility differ significantly. This review compares the subfertility phenotypes and their mechanisms.


Assuntos
Proteínas do Olho/genética , Haploinsuficiência , Proteínas de Homeodomínio/genética , Infertilidade/genética , Proteínas do Tecido Nervoso/genética , Neuropeptídeos/genética , Fatores de Transcrição Otx/genética , Animais , Proteínas do Olho/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Proteínas de Homeodomínio/metabolismo , Infertilidade/metabolismo , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Fatores de Transcrição Otx/metabolismo , Fenótipo , Proteína Homeobox SIX3
4.
Neuroendocrinology ; 108(4): 328-342, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30739114

RESUMO

There is an increasing trend in studies utilizing cell-specific deletion of genes through conditional gene deletion by CRE recombination. Despite numerous advantages, this strategy also has limitations such as ectopic CRE-expression and germline recombination. Two commonly used gonadotropin-releasing hormone (Gnrh)-driven CRE-expressing mice both target GnRH neurons. However, a direct comparison of the cells targeted and their phenotypic outcome have not yet been presented. To compare where recombination takes place, we crossed the Gnrh-cre and Lhrh-cre lines with the Rosa26-LacZ reporter mouse. Lhrh-cre allowed recombination of the Rosa26-LacZ gene in ∼700 cells, which is comparable to the GnRH neuronal population. Surprisingly, there were > 20 times more LacZ expressing cells in the adult Gnrh-cre:Rosa26-LacZ than the Lhrh-cre:Rosa26-LacZ brain. The greatest differences in targeting of the Gnrh-cre and Lhrh-cre lines were found in the septum, the suprachiasmatic nucleus, and the septohypothalamic area. This difference in cells targeted was present from embryonic day 12. A prior study using the Gnrh-cre to delete the transcription factor Otx2 found fewer GnRH neurons, leading to male and female subfertility. To recapitulate this study, we performed a fertility assay in Otx2:Lhrh-cre mice. We confirmed the requirement for Otx2 in GnRH neuron development, fertility and correct gonadotropin hormone release in Otx2:Lhrh-cre males, but the subfertility was more modest than in Otx2:Gnrh-cre and absent in female Otx2:Lhrh-cre. This suggests that ectopic expression of Gnrh-cre contributes to the reproductive phenotype observed. Finally, the Cre alleles caused germline recombination of the flox allele when transmitted from either parent, generating embryonic lethal knock-out offspring, producing smaller live litters.


Assuntos
Hormônio Liberador de Gonadotropina/genética , Infertilidade/genética , Fatores de Transcrição Otx/genética , Alelos , Animais , Encéfalo/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Camundongos Transgênicos , Neurônios/metabolismo , Regiões Promotoras Genéticas/genética , RNA Mensageiro/metabolismo
5.
Proc Natl Acad Sci U S A ; 111(30): 11133-8, 2014 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-25028498

RESUMO

The gut endocrine system is emerging as a central player in the control of appetite and glucose homeostasis, and as a rich source of peptides with therapeutic potential in the field of diabetes and obesity. In this study we have explored the physiology of insulin-like peptide 5 (Insl5), which we identified as a product of colonic enteroendocrine L-cells, better known for their secretion of glucagon-like peptide-1 and peptideYY. i.p. Insl5 increased food intake in wild-type mice but not mice lacking the cognate receptor Rxfp4. Plasma Insl5 levels were elevated by fasting or prolonged calorie restriction, and declined with feeding. We conclude that Insl5 is an orexigenic hormone released from colonic L-cells, which promotes appetite during conditions of energy deprivation.


Assuntos
Colo/metabolismo , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Células Enteroendócrinas/metabolismo , Hormônios Peptídicos/metabolismo , Hormônios Peptídicos/farmacologia , Animais , Feminino , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Humanos , Masculino , Camundongos , Camundongos Knockout , Peptídeo YY/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Peptídeos/metabolismo
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.
J Neurosci ; 31(2): 426-38, 2011 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-21228153

RESUMO

The hypothalamus, pituitary, and gonads coordinate to direct the development and regulation of reproductive function in mammals. Control of the hypothalamic-pituitary-gonadal axis is dependent on correct migration of gonadotropin-releasing hormone (GnRH) neurons from the nasal placode to the hypothalamus, followed by proper synthesis and pulsatile secretion of GnRH, functions absent in patients with hypogonadal hypogonadism. In this study, we identify sine oculis-related homeobox 6 (Six6) as a novel factor necessary for proper targeting of GnRH expression to the limited population of GnRH neurons within the adult mouse hypothalamus and demonstrate that it is required for proper reproductive function in both male and female mice. Female Six6-null mice exhibit a striking decrease in fertility, failing to progress through the estrous cycle normally, show any signs of successful ovulation, or produce litters. Although basal gonadotropin production in these mice is relatively normal, analysis of GnRH expression reveals a dramatic decrease in total GnRH neuron numbers. We show that expression of Six6 is dramatically increased during GnRH neuronal maturation and that overexpression of Six6 induces GnRH transcription in neuronal cells. Finally, we demonstrate that this induction in GnRH expression is mediated via binding of Six6 to evolutionarily conserved ATTA sites located within the GnRH proximal promoter. Together, these data indicate that Six6 plays an important role in the regulation of GnRH expression and hypothalamic control of fertility.


Assuntos
Fertilidade/fisiologia , Hormônio Liberador de Gonadotropina/biossíntese , Proteínas de Homeodomínio/fisiologia , Hipotálamo/metabolismo , Reprodução/fisiologia , Transativadores/fisiologia , Animais , Linhagem Celular , Ciclo Estral/fisiologia , Feminino , Hormônio Liberador de Gonadotropina/genética , Proteínas de Homeodomínio/genética , Hipotálamo/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/citologia , Neurônios/metabolismo , Ovulação/fisiologia , Gravidez , Regiões Promotoras Genéticas , Fatores Sexuais , Transativadores/genética , Transcrição Gênica
8.
J Biol Chem ; 284(25): 16966-16978, 2009 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-19401468

RESUMO

Hormonal communication between the hypothalamus, pituitary, and gonads orchestrates the development and regulation of mammalian reproductive function. In mice, gonadotropin-releasing hormone (GnRH) expression is limited to approximately 1000 neurons that originate in the olfactory placode then migrate to specific positions scattered throughout the hypothalamus. Coordination of the hypothalamic-pituitary-gonadal axis is dependent upon correct migration of GnRH neurons into the hypothalamus followed by the appropriate synthesis and pulsatile secretion of GnRH. Defects in any one of these processes can cause infertility. Recently, substantial progress has been made in identifying transcription factors, and their cofactors, that regulate not only adult expression of GnRH, but also the maturation of GnRH neurons. Here, we show that expression of Otx2, a homeodomain protein required for the formation of the forebrain, is dramatically up-regulated during GnRH neuronal maturation and that overexpression of Otx2 increases GnRH promoter activity in GnRH neuronal cell lines. Furthermore, Otx2 transcriptional activity is modulated by Grg4, a member of the Groucho-related-gene (Grg) family. Using mutational analysis, we show that a WRPW peptide motif within the Otx2 protein is required for physical interaction between Otx2 and Grg4. Without this physical interaction, Grg4 cannot repress Otx2-dependent activation of GnRH gene transcription. Taken together, these data show that Otx2 is important for GnRH expression and that direct interaction between Otx2 and Grg co-repressors regulates GnRH gene expression in hypothalamic neurons.


Assuntos
Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Fatores de Transcrição Otx/genética , Fatores de Transcrição Otx/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Motivos de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , Linhagem Celular , Proteínas Correpressoras , Primers do DNA/genética , Camundongos , Dados de Sequência Molecular , Mutação , Células NIH 3T3 , Neurônios/metabolismo , Fatores de Transcrição Otx/química , Regiões Promotoras Genéticas , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Homologia de Sequência do Ácido Nucleico , Ativação Transcricional
9.
Mol Endocrinol ; 21(5): 1216-33, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17327421

RESUMO

G protein-coupled receptor regulation of gene transcription primarily occurs through the phosphorylation of transcription factors by MAPKs. This requires transduction of an activating signal via scaffold proteins that can ultimately determine the outcome by binding signaling kinases and adapter proteins with effects on the target transcription factor and locus of activation. By investigating these mechanisms, we have elucidated how pituitary gonadotrope cells decode an input GnRH signal into coherent transcriptional output from the LH beta-subunit gene promoter. We show that GnRH activates c-Src and multiple members of the MAPK family, c-Jun NH2-terminal kinase 1/2, p38MAPK, and ERK1/2. Using dominant-negative point mutations and chemical inhibitors, we identified that calcium-dependent proline-rich tyrosine kinase 2 specifically acts as a scaffold for a focal adhesion/cytoskeleton-dependent complex comprised of c-Src, Grb2, and mSos that translocates an ERK-activating signal to the nucleus. The locus of action of ERK was specifically mapped to early growth response-1 (Egr-1) DNA binding sites within the LH beta-subunit gene proximal promoter, which was also activated by p38MAPK, but not c-Jun NH2-terminal kinase 1/2. Egr-1 was confirmed as the transcription factor target of ERK and p38MAPK by blockade of protein expression, transcriptional activity, and DNA binding. We have identified a novel GnRH-activated proline-rich tyrosine kinase 2-dependent ERK-mediated signal transduction pathway that specifically regulates Egr-1 activation of the LH beta-subunit proximal gene promoter, and thus provide insight into the molecular mechanisms required for differential regulation of gonadotropin gene expression.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação da Expressão Gênica , Hormônio Liberador de Gonadotropina/fisiologia , Hormônio Luteinizante Subunidade beta/genética , Proteínas Tirosina Quinases/fisiologia , Transcrição Gênica , Animais , Proteína 1 de Resposta de Crescimento Precoce/fisiologia , Amplificação de Genes , Humanos , Camundongos , Fosforilação , Regiões Promotoras Genéticas , Ovinos , Transdução de Sinais , Transfecção
10.
J Clin Invest ; 128(3): 960-969, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29376887

RESUMO

Profound hyperphagia is a major disabling feature of Prader-Willi syndrome (PWS). Characterization of the mechanisms that underlie PWS-associated hyperphagia has been slowed by the paucity of animal models with increased food intake or obesity. Mice with a microdeletion encompassing the Snord116 cluster of noncoding RNAs encoded within the Prader-Willi minimal deletion critical region have previously been reported to show growth retardation and hyperphagia. Here, consistent with previous reports, we observed growth retardation in Snord116+/-P mice with a congenital paternal Snord116 deletion. However, these mice neither displayed increased food intake nor had reduced hypothalamic expression of the proprotein convertase 1 gene PCSK1 or its upstream regulator NHLH2, which have recently been suggested to be key mediators of PWS pathogenesis. Specifically, we disrupted Snord116 expression in the mediobasal hypothalamus in Snord116fl mice via bilateral stereotaxic injections of a Cre-expressing adeno-associated virus (AAV). While the Cre-injected mice had no change in measured energy expenditure, they became hyperphagic between 9 and 10 weeks after injection, with a subset of animals developing marked obesity. In conclusion, we show that selective disruption of Snord116 expression in the mediobasal hypothalamus models the hyperphagia of PWS.


Assuntos
Hiperfagia/metabolismo , Hipotálamo/metabolismo , Síndrome de Prader-Willi/genética , RNA Nucleolar Pequeno/genética , Animais , Composição Corporal , Dependovirus , Modelos Animais de Doenças , Deleção de Genes , Genótipo , Hiperfagia/genética , Masculino , Camundongos , Camundongos Transgênicos , Obesidade/metabolismo , Síndrome de Prader-Willi/metabolismo
11.
Endocrinology ; 147(12): 5676-89, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16946016

RESUMO

GnRH binds its cognate G protein-coupled GnRH receptor (GnRHR) located on pituitary gonadotropes and drives expression of gonadotropin hormones. There are two gonadotropin hormones, comprised of a common alpha- and hormone-specific beta-subunit, which are required for gonadal function. Recently we identified that Fanconi anemia a (Fanca), a DNA damage repair gene, is differentially expressed within the LbetaT2 gonadotrope cell line in response to stimulation with GnRH. FANCA is mutated in more than 60% of cases of Fanconi anemia (FA), a rare genetically heterogeneous autosomal recessive disorder characterized by bone marrow failure, endocrine tissue cancer susceptibility, and infertility. Here we show that induction of FANCA protein is mediated by the GnRHR and that the protein constitutively adopts a nucleocytoplasmic intracellular distribution pattern. Using inhibitors to block nuclear import and export and a GnRHR antagonist, we demonstrated that GnRH induces nuclear accumulation of FANCA and green fluorescent protein (GFP)-FANCA before exporting back to the cytoplasm using the nuclear export receptor CRM1. Using FANCA point mutations that locate GFP-FANCA to the cytoplasm (H1110P) or functionally uncouple GFP-FANCA (Q1128E) from the wild-type nucleocytoplasmic distribution pattern, we demonstrated that wild-type FANCA was required for GnRH-induced activation of gonadotrope cell markers. Cotransfection of H1110P and Q1128E blocked GnRH activation of the alphaGsu and GnRHR but not the beta-subunit gene promoters. We conclude that nucleocytoplasmic shuttling of FANCA is required for GnRH transduction of the alphaGSU and GnRHR gene promoters and propose that FANCA functions as a GnRH-induced signal transducer.


Assuntos
Proteína do Grupo de Complementação A da Anemia de Fanconi/fisiologia , Hormônio Liberador de Gonadotropina/metabolismo , Proteínas de Transporte Nucleocitoplasmático/fisiologia , Receptores LHRH/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Células Cultivadas , Proteína do Grupo de Complementação A da Anemia de Fanconi/metabolismo , Subunidade beta do Hormônio Folículoestimulante/genética , Subunidade alfa de Hormônios Glicoproteicos/genética , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Carioferinas/fisiologia , Hormônio Luteinizante Subunidade beta/genética , Proteínas Mutantes Quiméricas/metabolismo , Regiões Promotoras Genéticas , Receptores Citoplasmáticos e Nucleares/fisiologia , Receptores LHRH/genética , Distribuição Tecidual , Proteína Exportina 1
12.
Mol Endocrinol ; 29(6): 842-55, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25915183

RESUMO

Sine oculis-related homeobox 3 (SIX3) and SIX6, 2 closely related homeodomain transcription factors, are involved in development of the mammalian neuroendocrine system and mutations of Six6 adversely affect fertility in mice. We show that both small interfering RNA knockdown in gonadotrope cell lines and knockout of Six6 in both embryonic and adult male mice (Six6 knockout) support roles for SIX3 and SIX6 in transcriptional regulation in gonadotrope gene expression and that SIX3 and SIX6 can functionally compensate for each other. Six3 and Six6 expression patterns in gonadotrope cell lines reflect the timing of the expression of pituitary markers they regulate. Six3 is expressed in an immature gonadotrope cell line and represses transcription of the early lineage-specific pituitary genes, GnRH receptor (GnRHR) and the common α-subunit (Cga), whereas Six6 is expressed in a mature gonadotrope cell line and represses the specific ß-subunits of LH and FSH (LHb and FSHb) that are expressed later in development. We show that SIX6 repression requires interaction with transducin-like enhancer of split corepressor proteins and competition for DNA-binding sites with the transcriptional activator pituitary homeobox 1. Our studies also suggest that estradiol and circadian rhythm regulate pituitary expression of Six6 and Six3 in adult females but not in males. In summary, SIX3 and SIX6 play distinct but compensatory roles in regulating transcription of gonadotrope-specific genes as gonadotrope cells differentiate.


Assuntos
Proteínas do Olho/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Gonadotrofos/metabolismo , Proteínas de Homeodomínio/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Transativadores/metabolismo , Animais , Células COS , Chlorocebus aethiops , Proteínas do Olho/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Gonadotrofos/efeitos dos fármacos , Proteínas de Homeodomínio/genética , Masculino , Proteínas do Tecido Nervoso/genética , Especificidade de Órgãos/efeitos dos fármacos , Especificidade de Órgãos/genética , Fatores de Transcrição Box Pareados/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Esteroides/farmacologia , Transativadores/genética , Transcrição Gênica/efeitos dos fármacos , Ativação Transcricional/efeitos dos fármacos , Ativação Transcricional/genética , Proteína Homeobox SIX3
13.
Handb Clin Neurol ; 124: 93-106, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25248581

RESUMO

Obesity and its related metabolic consequences represent a major public health problem. Huge changes within the environment have undoubtedly contributed to the increased prevalence of obesity but genetic factors are also critical in determining an individual's predisposition to gain weight. The last two decades have seen a huge increase in the understanding of the mechanisms controlling appetitive behavior, body composition, and energy expenditure. Many regions throughout the central nervous system play critical roles in these processes but the hypothalamus, in particular, receives and orchestrates a variety of signals to bring about coordinated changes in energy balance. Reviewing data from human genetic and model organism studies, we consider how disruptions of hypothalamic pathways evolved to maintain energy homeostasis and go on to cause obesity. We highlight ongoing technological developments which continue to lead to novel insights and discuss how this increased knowledge may lead to effective therapeutic interventions in the future.


Assuntos
Hipotálamo/fisiologia , Obesidade/genética , Obesidade/metabolismo , Animais , Metabolismo Energético/fisiologia , Predisposição Genética para Doença/genética , Homeostase/fisiologia , Humanos , Obesidade/diagnóstico , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo
14.
Mol Cell Endocrinol ; 377(1-2): 16-22, 2013 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-23811236

RESUMO

Together, the hypothalamus, pituitary and gonads direct the development and regulation of reproductive function in mammals. Gonadotropin-releasing hormone (GnRH) expression is limited to ∼800 neurons that originate in the olfactory placode then migrate to the hypothalamus. Coordination of the hypothalamic-pituitary-gonadal (HPG) axis is dependent upon correct neuronal migration of GnRH neurons into the hypothalamus followed by proper synthesis and pulsatile secretion of GnRH. Defects in any one of these processes causes infertility. Otx2, the vertebrate homologue of Drosophila orthodenticle, is a transcription factor that has been shown to be critical for normal brain and eye development and is expressed in both the developing GnRH neurons and the pituitary, suggesting that this gene may play a critical role in development of the HPG axis. As Otx2-null mice are embryonic lethal, we have analyzed the reproductive capacity of heterozygous Otx2 mice to determine the contribution of Otx2 gene dosage to normal HPG axis function. Our data reveal that correct dosage of Otx2 is critical for normal fertility as loss of one allele of Otx2 leads to a discernible reproductive phenotype in male mice due to disruption of the migration of GnRH neurons during development.


Assuntos
Fertilidade/genética , Dosagem de Genes/genética , Fatores de Transcrição Otx/genética , Envelhecimento/metabolismo , Alelos , Animais , Axônios/metabolismo , Movimento Celular/genética , Regulação da Expressão Gênica , Gonadotrofos/metabolismo , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Gonadotropinas/genética , Heterozigoto , Hormônio Luteinizante/sangue , Masculino , Camundongos , Fatores de Transcrição Otx/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcrição Gênica
15.
J Exp Med ; 210(6): 1137-51, 2013 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-23712428

RESUMO

Fibroblast activation protein-α (FAP) identifies stromal cells of mesenchymal origin in human cancers and chronic inflammatory lesions. In mouse models of cancer, they have been shown to be immune suppressive, but studies of their occurrence and function in normal tissues have been limited. With a transgenic mouse line permitting the bioluminescent imaging of FAP(+) cells, we find that they reside in most tissues of the adult mouse. FAP(+) cells from three sites, skeletal muscle, adipose tissue, and pancreas, have highly similar transcriptomes, suggesting a shared lineage. FAP(+) cells of skeletal muscle are the major local source of follistatin, and in bone marrow they express Cxcl12 and KitL. Experimental ablation of these cells causes loss of muscle mass and a reduction of B-lymphopoiesis and erythropoiesis, revealing their essential functions in maintaining normal muscle mass and hematopoiesis, respectively. Remarkably, these cells are altered at these sites in transplantable and spontaneous mouse models of cancer-induced cachexia and anemia. Thus, the FAP(+) stromal cell may have roles in two adverse consequences of cancer: their acquisition by tumors may cause failure of immunosurveillance, and their alteration in normal tissues contributes to the paraneoplastic syndromes of cachexia and anemia.


Assuntos
Anemia/metabolismo , Medula Óssea/metabolismo , Caquexia/metabolismo , Gelatinases/genética , Gelatinases/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Músculo Esquelético/metabolismo , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Células Estromais/metabolismo , Tecido Adiposo/metabolismo , Anemia/genética , Anemia/patologia , Animais , Caquexia/genética , Caquexia/patologia , Linhagem da Célula/genética , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Endopeptidases , Eritropoese/genética , Folistatina/genética , Folistatina/metabolismo , Hematopoese/genética , Linfopoese/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos/genética , Camundongos Transgênicos/metabolismo , Músculo Esquelético/citologia , Transtornos Musculares Atróficos/genética , Transtornos Musculares Atróficos/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Pâncreas/metabolismo , Células Estromais/citologia , Transcriptoma/genética
16.
Nat Med ; 18(5): 666-7, 2012 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-22561823

RESUMO

Losing weight can pose a challenge, but how to avoid putting those pounds back on can be a real struggle. A major health problem for obese people is that diseases linked to obesity, such as type 2 diabetes and cardiovascular disease, put their lives at risk, even in young individuals. Although bariatric surgery-a surgical method to reduce or modify the gastrointestinal tract-was originally envisioned for the most severe cases of obesity, evidence suggests that the benefit of this procedure may not be limited to the staggering weight loss it causes. Endogenous factors released from the gut, and modified after surgery, may explain why bariatric surgery can be beneficial for obesity-related diseases and why operated individuals successfully maintain the weight loss. In 'Bedside to Bench,' Rachel Larder and Stephen O'Rahilly peruse a human study with dieters who regained weight despite a successful diet. Appetite-regulating hormones in the gut may be responsible for this relapse in the long term. In 'Bench to Bedside,' Keval Chandarana and Rachel Batterham examine how two different methods of bariatric surgery highlight the relevance of gut-derived hormones not only in inducing sustained weight loss but also in improving glucose homeostasis. These insights may open new avenues to bypass the surgery and obtain the same results with targeted drugs.


Assuntos
Cirurgia Bariátrica , Obesidade/cirurgia , Colecistocinina/fisiologia , Peptídeo 1 Semelhante ao Glucagon/fisiologia , Glucose/metabolismo , Humanos , Obesidade/metabolismo , Peptídeo YY/fisiologia , Redução de Peso
17.
Trends Endocrinol Metab ; 22(2): 53-9, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21131211

RESUMO

An understanding of the mechanisms underlying body-weight regulation is crucial to tackle the growing problem of obesity. Recent technological advances in the analysis of genetic variation have given novel insights into the molecular basis of common disease. In particular, genomic variants in the fat mass and obesity-associated (FTO) gene have been consistently associated with human adiposity and metabolic disorders. Studies of the product of this previously mysterious gene have formed a vanguard in the quest to turn statistical association into hard biology. In this review, we examine data from human genetic and murine studies that explore the potential role of FTO, a member of the Fe(II)- and 2-oxoglutarate-dependent oxygenase superfamily, in the regulation of energy homeostasis and metabolism.


Assuntos
Obesidade/genética , Proteínas/genética , Dioxigenase FTO Dependente de alfa-Cetoglutarato , Animais , Índice de Massa Corporal , Metilação de DNA , Ingestão de Energia , Metabolismo Energético/genética , Predisposição Genética para Doença , Homeostase/genética , Humanos , Camundongos , Mutação , Fenótipo , Polimorfismo de Nucleotídeo Único/genética , Proteínas/química , Proteínas/fisiologia
18.
Biol Reprod ; 71(3): 828-36, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15128600

RESUMO

Gonadal function is critically dependant on regulated secretion of the gonadotropin hormones from anterior pituitary gonadotroph cells. Gonadotropin biosynthesis and release is triggered by the binding of hypothalamic GnRH to GnRH receptor expressed on the gonadotroph cell surface. The repertoire of regulatory molecules involved in this process are still being defined. We used the mouse L beta T2 gonadotroph cell line, which expresses both gonadotropin hormones, as a model to investigate GnRH regulation of gene expression and differential display reverse transcription-polymerase chain reaction (RT-PCR) to identify and isolate hormonally induced changes. This approach identified Fanconi anemia a (Fanca), a gene implicated in DNA damage repair, as a differentially expressed transcript. Mutations in Fanca account for the majority of cases of Fanconi anemia (FA), a recessively inherited disease identified by congenital defects, bone marrow failure, infertility, and cancer susceptibility. We confirmed expression and hormonal regulation of Fanca mRNA by quantitative RT-PCR, which showed that GnRH induced a rapid, transient increase in Fanca mRNA. Fanca protein was also acutely upregulated after GnRH treatment of L beta T2 cells. In addition, Fanca gene expression was confined to mature pituitary gonadotrophs and adult mouse pituitary and was not expressed in the immature alpha T3-1 gonadotroph cell line. Thus, this study extends the expression profile of Fanca into a highly specialized endocrine cell and demonstrates hormonal regulation of expression of the Fanca locus. We suggest that this regulatory mechanism may have a crucial role in the GnRH-response mechanism of mature gonadotrophs and perhaps the etiology of FA.


Assuntos
Proteínas de Ligação a DNA/genética , Hormônio Liberador de Gonadotropina/farmacologia , Adeno-Hipófise/fisiologia , Animais , Sequência de Bases , Linhagem Celular , Dano ao DNA , Proteína do Grupo de Complementação A da Anemia de Fanconi , Expressão Gênica/efeitos dos fármacos , Camundongos , Dados de Sequência Molecular , Adeno-Hipófise/citologia , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA