Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 18(10): e0292579, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37816033

RESUMO

Pancreatic islet failure is a key characteristic of type 2 diabetes besides insulin resistance. To get molecular insights into the pathology of islets in type 2 diabetes, we developed a computational approach to integrating expression profiles of Goto-Kakizaki and Wistar rat islets from a designed experiment with those of the human islets from an observational study. A principal gene-eigenvector in the expression profiles characterized by up-regulated angiogenesis and down-regulated oxidative phosphorylation was identified conserved across the two species. In the case of Goto-Kakizaki versus Wistar islets, such alteration in gene expression can be verified directly by the treatment-control tests over time, and corresponds to the alteration of α/ß-cell distribution obtained by quantifying the islet micrographs. Furthermore, the correspondence between the dual sample- and gene-eigenvectors unveils more delicate structures. In the case of rats, the up- and down-trend of insulin mRNA levels before and after week 8 correspond respectively to the top two principal eigenvectors. In the case of human, the top two principal eigenvectors correspond respectively to the late and early stages of diabetes. According to the aggregated expression signature, a large portion of genes involved in the hypoxia-inducible factor signaling pathway, which activates transcription of angiogenesis, were significantly up-regulated. Furthermore, top-ranked anti-angiogenic genes THBS1 and PEDF indicate the existence of a counteractive mechanism that is in line with thickened and fragmented capillaries found in the deteriorated islets. Overall, the integrative analysis unravels the principal transcriptional alterations underlying the islet deterioration of morphology and insulin secretion along type 2 diabetes progression.


Assuntos
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Ilhotas Pancreáticas , Ratos , Humanos , Animais , Diabetes Mellitus Tipo 2/patologia , Ratos Wistar , Ilhotas Pancreáticas/metabolismo , Células Secretoras de Insulina/metabolismo , Secreção de Insulina , Insulina/genética , Insulina/metabolismo
2.
Sci Rep ; 7(1): 5044, 2017 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-28698587

RESUMO

Type 2 diabetes (T2D) is a complex and polygenic disease yet in need of a complete picture of its development mechanisms. To better understand the mechanisms, we examined gene expression profiles of multi-tissues from outbred mice fed with a high-fat diet (HFD) or regular chow at weeks 1, 9, and 18. To analyze such complex data, we proposed a novel dual eigen-analysis, in which the sample- and gene-eigenvectors correspond respectively to the macro- and micro-biology information. The dual eigen-analysis identified the HFD eigenvectors as well as the endogenous eigenvectors for each tissue. The results imply that HFD influences the hepatic function or the pancreatic development as an exogenous factor, while in adipose HFD's impact roughly coincides with the endogenous eigenvector driven by aging. The enrichment analysis of the eigenvectors revealed diverse HFD impact on the three tissues over time. The diversity includes: inflammation, degradation of branched chain amino acids (BCAA), and regulation of peroxisome proliferator activated receptor gamma (PPARγ). We reported that in the pancreas remarkable up-regulation of angiogenesis as downstream of the HIF signaling pathway precedes hyperinsulinemia. The dual eigen-analysis and discoveries provide new evaluations/guidance in T2D prevention and therapy, and will also promote new thinking in biology and medicine.


Assuntos
Diabetes Mellitus Tipo 2/genética , Perfilação da Expressão Gênica , Especificidade de Órgãos/genética , Adiponectina/metabolismo , Tecido Adiposo/metabolismo , Aminoácidos de Cadeia Ramificada/metabolismo , Animais , Colesterol/biossíntese , Dieta Hiperlipídica , Regulação para Baixo/genética , Insulina/metabolismo , Fígado/metabolismo , Camundongos , PPAR gama/metabolismo , Pâncreas/metabolismo , Transdução de Sinais , Regulação para Cima/genética
3.
Cell Mol Life Sci ; 73(20): 3917-33, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27113546

RESUMO

Vascular permeability regulated by the vascular endothelial growth factor (VEGF) through endothelial-barrier junctions is essential for inflammation. Mechanisms regulating vascular permeability remain elusive. Although 'Akt' and 'Src' have been implicated in the endothelial-barrier regulation, it is puzzling how both agents that protect and disrupt the endothelial-barrier activate these kinases to reciprocally regulate vascular permeability. To delineate the role of Akt1 in endothelial-barrier regulation, we created endothelial-specific, tamoxifen-inducible Akt1 knockout mice and stable ShRNA-mediated Akt1 knockdown in human microvascular endothelial cells. Akt1 loss leads to decreased basal and angiopoietin1-induced endothelial-barrier resistance, and enhanced VEGF-induced endothelial-barrier breakdown. Endothelial Akt1 deficiency resulted in enhanced VEGF-induced vascular leakage in mice ears, which was rescued upon re-expression with Adeno-myrAkt1. Furthermore, co-treatment with angiopoietin1 reversed VEGF-induced vascular leakage in an Akt1-dependent manner. Mechanistically, our study revealed that while VEGF-induced short-term vascular permeability is independent of Akt1, its recovery is reliant on Akt1 and FoxO-mediated claudin expression. Pharmacological inhibition of FoxO transcription factors rescued the defective endothelial barrier due to Akt1 deficiency. Here we provide novel insights on the endothelial-barrier protective role of VEGF in the long term and the importance of Akt1-FoxO signaling on tight-junction stabilization and prevention of vascular leakage through claudin expression.


Assuntos
Células Endoteliais/metabolismo , Proteína Forkhead Box O3/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Junções Íntimas/metabolismo , Junções Aderentes/efeitos dos fármacos , Junções Aderentes/metabolismo , Angiopoietina-1/farmacologia , Animais , Permeabilidade Capilar/efeitos dos fármacos , Claudina-5/metabolismo , Células Endoteliais/efeitos dos fármacos , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Camundongos Transgênicos , Microvasos/citologia , Proteínas Proto-Oncogênicas c-akt/deficiência , Junções Íntimas/efeitos dos fármacos , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/farmacologia
4.
Cancer Cell ; 29(4): 523-535, 2016 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-26996309

RESUMO

Akt is frequently hyperactivated in human cancers and is targeted for cancer therapy. However, the physiological consequences of systemic Akt isoform inhibition were not fully explored. We showed that while combined Akt1 and Akt3 deletion in adult mice is tolerated, combined Akt1 and Akt2 deletion induced rapid mortality. Akt2(-/-) mice survived hepatic Akt1 deletion but all developed spontaneous hepatocellular carcinoma (HCC), which is associated with FoxO-dependent liver injury and inflammation. The gene expression signature of HCC-bearing livers is similar to aggressive human HCC. Consistently, neither Akt1(-/-) nor Akt2(-/-) mice are resistant to diethylnitrosamine-induced hepatocarcinogenesis, and Akt2(-/-) mice display a high incidence of lung metastasis. Thus, in contrast to other cancers, hepatic Akt inhibition induces liver injury that could promote HCC.


Assuntos
Neoplasias Hepáticas Experimentais/genética , Proteínas Proto-Oncogênicas c-akt/deficiência , Fatores Etários , Animais , Glicemia/análise , Metabolismo Energético/genética , Ácidos Graxos/metabolismo , Feminino , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/fisiologia , Transtornos do Crescimento/genética , Hepatite/genética , Hipoglicemia/genética , Insulina/sangue , Resistência à Insulina , Interleucina-6/sangue , Neoplasias Hepáticas Experimentais/induzido quimicamente , Neoplasias Pulmonares/secundário , Masculino , Camundongos , Camundongos Knockout , Regiões Promotoras Genéticas/efeitos dos fármacos , Isoformas de Proteínas/fisiologia , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/fisiologia , Proteínas Recombinantes de Fusão/metabolismo
5.
Nat Commun ; 7: 10960, 2016 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-26971877

RESUMO

The signalling pathways operational in quiescent, post-development vasculature remain enigmatic. Here we show that unlike neovascularization, endothelial Akt signalling in established vasculature is crucial not for endothelial cell (EC) survival, but for sustained interactions with pericytes and vascular smooth muscle cells (VSMCs) regulating vascular stability and function. Inducible endothelial-specific Akt1 deletion in adult global Akt2KO mice triggers progressive VSMC apoptosis. In hearts, this causes a loss of arteries and arterioles and, despite a high capillary density, diminished vascular patency and severe cardiac dysfunction. Similarly, endothelial Akt deletion induces retinal VSMC loss and basement membrane deterioration resulting in vascular regression and retinal atrophy. Mechanistically, the Akt/mTOR axis controls endothelial Jagged1 expression and, thereby, Notch signalling regulating VSMC maintenance. Jagged1 peptide treatment of Akt1ΔEC;Akt2KO mice and Jagged1 re-expression in Akt-deficient endothelium restores VSMC coverage. Thus, sustained endothelial Akt1/2 signalling is critical in maintaining vascular stability and homeostasis, thereby preserving tissue and organ function.


Assuntos
Vasos Sanguíneos/metabolismo , Proteínas de Ligação ao Cálcio/genética , Células Endoteliais/metabolismo , Endotélio/metabolismo , Homeostase/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteínas de Membrana/genética , Proteínas Proto-Oncogênicas c-akt/genética , Angiografia , Animais , Materiais Biocompatíveis , Barreira Hematoencefálica/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Colágeno , Vasos Coronários/metabolismo , Combinação de Medicamentos , Ecocardiografia , Olho/irrigação sanguínea , Imunofluorescência , Regulação da Expressão Gênica , Coração , Células Endoteliais da Veia Umbilical Humana , Humanos , Immunoblotting , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteína Jagged-1 , Laminina , Pulmão/irrigação sanguínea , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso , Pericitos , Proteoglicanas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Retina , Vasos Retinianos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Serrate-Jagged , Transdução de Sinais/genética , Microtomografia por Raio-X
6.
EMBO J ; 31(5): 1134-46, 2012 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-22307088

RESUMO

The requirement of Akt for cell proliferation and oncogenesis is mammalian target of rapamycin complex 1 (mTORC1) dependent. SV40 large T expression in Akt-deficient cells restores cell proliferation rate, but is insufficient for exiting contact inhibition and oncogene-induced anchorage-independent growth, because of a failure to promote Skp2 mRNA translation. Skp2 mRNA and protein are induced upon exiting contact inhibition, which enables entry into mitosis. While Skp2 mRNA is induced in Akt-deficient cells, it is not translated, preventing entry into mitosis. Restoring Skp2 expression in Akt-deficient cells is sufficient to restore exit from contact inhibition and oncogenesis. Skp2 mRNA translation is dependent on mTORC1 and the eukaryotic translation initiation factor 4E (eIF4E). Thus, the requirement of Akt for exiting contact inhibition is mediated by the induction of Skp2 mRNA translation in eIF4E-dependent mechanism. These results provide a new insight into the role of the Akt/mTORC1/eIF4E axis in tumourigenesis. Akt-dependent Skp2 mRNA translation is also required for mitotic clonal expansion (MCE)--the earliest event in adipogenesis. Skp2 re-expression in Akt-deficient preadipocytes, which are impaired in adipogenesis, is sufficient to restore adipogenesis. These results uncover the mechanism by which Akt mediates adipogenesis.


Assuntos
Adipogenia , Transformação Celular Neoplásica , Inibição de Contato , Fator de Iniciação 4E em Eucariotos/metabolismo , Proteína Oncogênica v-akt/metabolismo , Proteínas/metabolismo , Proteínas Quinases Associadas a Fase S/biossíntese , Animais , Proliferação de Células , Células Cultivadas , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Knockout , Complexos Multiproteicos , Biossíntese de Proteínas , Serina-Treonina Quinases TOR
7.
Endocrinology ; 150(7): 3177-85, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19342460

RESUMO

Hyperactivation of the GHRH receptor or downstream signaling components is associated with hyperplasia of the pituitary somatotrope population, in which adenomas form relatively late in life, with less than 100% penetrance. Hyperplastic and adenomatous pituitaries of metallothionein promoter-human GHRH transgenic (Tg) mice (4 and > 10 months, respectively) were used to identify mechanisms that may prevent or delay adenoma formation in the presence of excess GHRH. In hyperplastic pituitaries, expression of the late G(1)/G(2) marker Ki67 increased, whereas the proportion of 5-bromo-2'-deoxyuridine-labeled cells (S phase marker) did not differ from age-matched controls. These results indicate cell cycle progression is blocked, with further evidence suggesting that enhanced p27 activity may contribute to this process. For adenomas, formation was associated with loss of p27 activity (nuclear localization and mRNA). Increased endogenous somatostatin (SST) tone may also slow the conversion from hyperplastic to adenomatous state because mRNA levels for SST receptors, sst2 and sst5, were elevated in hyperplastic pituitaries, whereas adenomas were associated with a decline in sst1 and sst5 mRNA. Also, SST-knockout Tg pituitaries were larger and adenomas formed earlier compared with those of SST-intact Tg mice. Unexpectedly, these changes were independent of changes in proliferation rate within the hyperplastic tissue, suggesting that endogenous SST controls GHRH-induced adenoma formation primarily via modulation of apoptotic and/or cellular senescence pathways, consistent with the predicted function of some of the most differentially expressed genes (Casp1, MAP2K1, TNFR2) identified by membrane arrays and confirmed by quantitative real-time RT-PCR.


Assuntos
Adenoma/fisiopatologia , Hormônio Liberador de Hormônio do Crescimento/genética , Metalotioneína/genética , Neoplasias Hipofisárias/fisiopatologia , Receptores de Neuropeptídeos/fisiologia , Receptores de Hormônios Reguladores de Hormônio Hipofisário/fisiologia , Envelhecimento , Animais , Proliferação de Células , Feminino , Hormônio Liberador de Hormônio do Crescimento/metabolismo , Hormônio do Crescimento Humano/genética , Humanos , Masculino , Camundongos , Camundongos Knockout , Tamanho do Órgão , Hipófise/anatomia & histologia , Antígeno Nuclear de Célula em Proliferação/biossíntese , Proteínas Proto-Oncogênicas/biossíntese , RNA Mensageiro/metabolismo , Somatostatina/fisiologia , Somatotrofos/metabolismo
8.
Mol Cell Biol ; 29(11): 3151-62, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19289493

RESUMO

Phenotypic analyses of mice null for the individual Akt isoforms suggested that they are functionally distinct and that only Akt2 plays a role in diabetes. We show here that Akt isoforms play compensatory and complementary roles in glucose homeostasis and diabetes. Insulin resistance in Akt2(-/-) mice was inhibited by haplodeficiency of Pten, suggesting that other Akt isoforms can compensate for Akt2 function. Haplodeficiency of Akt1 in Akt2(-/-) mice, however, converts prediabetes to overt type 2 diabetes, which is also reversed by haplodeficiency of Pten. Akt3 does not appear to contribute significantly to diabetes. Overt type 2 diabetes in Akt1(+/-) Akt2(-/-) mice is manifested by hyperglycemia due to beta-cell dysfunction combined with impaired glucose homeostasis due to markedly decreased leptin levels. Restoring leptin levels was sufficient to restore normal blood glucose and insulin levels in Akt1(+/-) Akt2(-/-) and Akt2(-/-) mice, suggesting that leptin-deficiency is the predominant cause of diabetes in these mice. These results uncover a new mechanism linking Akt to diabetes, provide a therapeutic strategy, and show that diabetes induced as a consequence of cancer therapy, via Akt inhibition, could be reversed by leptin therapy.


Assuntos
Diabetes Mellitus Tipo 2/enzimologia , Diabetes Mellitus Tipo 2/fisiopatologia , Células Secretoras de Insulina/enzimologia , Leptina/deficiência , Proteínas Proto-Oncogênicas c-akt/deficiência , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Glicemia/metabolismo , Diabetes Mellitus Tipo 2/sangue , Comportamento Alimentar , Homeostase , Técnicas In Vitro , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Isoenzimas/deficiência , Isoenzimas/metabolismo , Leptina/sangue , Camundongos , Camundongos Knockout
9.
Clin Chim Acta ; 390(1-2): 44-8, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18230350

RESUMO

BACKGROUND: Recently, membrane type matrix metalloproteinase-1 (MT1-MMP) was found to participate in bone metabolism. We investigated the relationship between serum MT1-MMP and bone mineral density (BMD) as well as bone metabolic markers in 206 Chinese postmenopausal women aged 43-80 years. METHODS: Western analysis and ELISA were performed to detect serum soluble MT1-MMP levels. BMD was measured by dual energy X-ray absorptiometry (DXA). Serum alkaline phosphatase (BAP) and N-telopeptides of type I collagen (NTX) were assayed using ELISA. RESULTS: We found that soluble MT1-MMP abundantly existed in human serum as protein lack of transmembrane domain. Serum MT1-MMP levels were detectable in all participants and the range of value was 221.2-863.0 ng/ml (435.6+/-98.2 ng/ml). We found a significant negative weaker correlation between MT1-MMP and BMD at lumbar spine, total hip (Thip), and femoral neck (FN) (all P<0.05). After adjustment for age and BMI, the correlation with BMD at FN and Thip disappeared (all P>0.05). Multiple linear stepwise regression analysis showed that MT1-MMP was not a determinant factor for BMD. The significant positive correlations between MT1-MMP and BAP, NTX were found, and remained significant after adjustment for age and BMI (all P<0.05). Moreover, serum MT1-MMP, BAP, and NTX decreased in response to alendronate therapy. CONCLUSION: Circulating MT1-MMP and bone turnover markers are correlated, and serum MT1-MMP levels may rise with increase in bone turnover.


Assuntos
Biomarcadores/sangue , Densidade Óssea , Metaloproteinase 14 da Matriz/sangue , Pós-Menopausa , Absorciometria de Fóton , Adulto , Idoso , Idoso de 80 Anos ou mais , Western Blotting , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Pessoa de Meia-Idade , Solubilidade
10.
Clin Chim Acta ; 387(1-2): 31-5, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17884030

RESUMO

BACKGROUND: Adiponectin, leptin, resistin, and visfatin, as the main circulating peptides secreted by adipose tissue, are potential contributors to bone metabolism. We investigated whether these serum adipocytokines levels are associated with BMD and bone turnover biochemical markers in 232 Chinese men (20-80 y). METHODS: Serum adiponectin, leptin, resistin, and visfatin levels were determined by ELISA. RESULTS: Leptin had a positively correlation with fat mass, and remained significant after adjustment for age and BMI. There was a significant negative weak correlation between adiponectin and fat mass, and disappear after adjustment for age and BMI. Resistin and visfatin were not significantly correlated with fat mass. In the multiple linear stepwise regression analysis, lean mass and adiponectin, but not leptin, resistin and visfatin, were independent predictors of BMD. The significant positive correlations between adiponectin and bone-specific alkaline phosphatase (BAP), bone cross-linked N-telopeptides of type collagen (NTX) were found, and remained significant after adjustment for age and fat mass. CONCLUSIONS: Adiponectin was an independent predictor of BMD in Chinese men, and positively correlated with bone turnover biochemical markers. It suggested that adiponectin exert a negative effect on bone mass in men.


Assuntos
Adiponectina/sangue , Biomarcadores/sangue , Densidade Óssea , Osso e Ossos/metabolismo , Leptina/sangue , Nicotinamida Fosforribosiltransferase/sangue , Resistina/sangue , Adulto , Idoso , Idoso de 80 Anos ou mais , Índice de Massa Corporal , Ensaio de Imunoadsorção Enzimática , Humanos , Masculino , Pessoa de Meia-Idade
11.
Cancer Cell ; 10(4): 269-80, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17045205

RESUMO

Akt contributes to tumorigenesis by inhibiting apoptosis. Here we establish that Akt is required for normal cell proliferation and susceptibility to oncogenesis independently of its antiapoptotic activity. Partial ablation of Akt activity by deleting Akt1 inhibits cell proliferation and oncogenesis. These effects are compounded by deleting both Akt1 and Akt2. In vivo, Akt1 null mice are resistant to MMTV-v-H-Ras-induced tumors and to skin carcinogenesis. Thus, partial ablation of Akt activity is sufficient to suppress tumorigenesis in vitro and in vivo. The effect of Akt deficiency on cell proliferation and oncogenesis is p53 independent but mTORC1 dependent. Surprisingly, upon mTORC1 hyperactivation, the reduction in Akt activity does not impair cell proliferation and susceptibility to oncogenic transformation; thus, Akt may mediate these processes exclusively via mTORC1.


Assuntos
Proliferação de Células , Neoplasias/etiologia , Proteínas Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/deficiência , Transativadores/metabolismo , Animais , Linhagem Celular Transformada , Transformação Celular Viral , Cruzamentos Genéticos , Embrião de Mamíferos , Fibroblastos/metabolismo , Cinética , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Complexos Multiproteicos , Neoplasias/patologia , Proteínas Quinases/genética , Proteínas , Proteínas Proto-Oncogênicas c-akt/genética , Retroviridae/genética , Serina-Treonina Quinases TOR , Transativadores/genética , Fatores de Transcrição , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
12.
Genes Dev ; 20(12): 1569-74, 2006 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-16778075

RESUMO

The tumor suppressor PTEN is frequently inactivated in human cancers. A major downstream effector of PTEN is Akt, which is hyperactivated via PTEN inactivation. It is not known, however, whether diminished Akt activity is sufficient to inhibit tumorigenesis initiated by Pten deficiency. Here we showed that the deficiency of Akt1 is sufficient to dramatically inhibit tumor development in Pten+/- mice. Akt1 deficiency had a profound effect on endometrium and prostate neoplasia, two types of human cancer, in which PTEN is frequently mutated, and also affected thyroid and adrenal medulla tumors and intestinal polyps. Even haplodeficiency of Akt1 was sufficient to markedly attenuate the development of high-grade prostate intraepithelial neoplasia (PIN) and endometrial carcinoma. These results have significant implications for cancer therapy.


Assuntos
Neoplasias/enzimologia , Neoplasias/patologia , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Proteínas Proto-Oncogênicas c-akt/deficiência , Neoplasias das Glândulas Suprarrenais/enzimologia , Neoplasias das Glândulas Suprarrenais/genética , Neoplasias das Glândulas Suprarrenais/patologia , Animais , Neoplasias do Endométrio/enzimologia , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/patologia , Feminino , Heterozigoto , Pólipos Intestinais/enzimologia , Pólipos Intestinais/patologia , Masculino , Camundongos , Neoplasias/genética , Neoplasia Prostática Intraepitelial/enzimologia , Neoplasia Prostática Intraepitelial/genética , Neoplasia Prostática Intraepitelial/patologia , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-akt/genética , Pseudolinfoma/enzimologia , Pseudolinfoma/genética , Pseudolinfoma/patologia , Neoplasias da Glândula Tireoide/enzimologia , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/patologia
13.
Neuroendocrinology ; 81(6): 360-71, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16244497

RESUMO

In the fasted and the streptozotocin (STZ)-induced diabetic male rat, hypothalamic growth hormone (GH)-releasing hormone (GHRH) mRNA levels, and pulsatile GH release are decreased. These changes are believed to be due to a rise in hypothalamic neuropeptide Y (NPY) that inhibits GHRH expression. To directly test if NPY is required for metabolic regulation of hypothalamic neuropeptides important in GH secretion, NPY, GHRH and somatostatin (SRIH) mRNA levels were determined in fasted (48 h) and STZ-treated wild-type (NPY(+/+)) and NPY-knockout (NPY(-/-)) mice by ribonuclease protection assay. In addition, pituitary receptor mRNA levels for GHRH (GHRH-R), ghrelin (GHS-R) and SRIH (sst2) were assessed by RT-PCR. Under fed conditions the GH axis of NPY(+/+) and NPY(-/-) did not differ. In the NPY(+/+) mouse, fasting resulted in a 23% weight loss and >250% increase in NPY mRNA accompanied by a significant reduction in both GHRH and SRIH mRNA. These changes were associated with increases in pituitary expression of GHRH-R and GHS-R and a concomitant suppression of sst2. In the NPY(-/-) mouse, fasting also resulted in a 23% weight loss and comparable changes in GHRH-R and sst2, but failed to alter GHRH, SRIH and GHS-R mRNA levels. Fasting resulted in an overall increase in circulating GH, which reached significance in the fasted NPY(-/-) mouse. Induction of diabetes in NPY(+/+) mice, using a single, high-dose, STZ injection (150 mg/kg), resulted in modest weight loss (5%), and a 158% increase NPY expression which was associated with reciprocal changes in pituitary GHS-R and sst2 expression, similar to that observed in the fasted state, but no change in hypothalamic GHRH or SRIF expression was observed. Induction of diabetes in NPY(+/+) and NPY(-/-) mice, using a multiple, low-dose, STZ paradigm (5 consecutive daily injections of 40 mg/kg), did not alter body weight, hypothalamic neuropeptide expression or pituitary receptor expression, with the exception that sst2 mRNA levels were suppressed and GH levels did rise in the NPY(-/-) mouse. These observations demonstrate that NPY is not required for basal regulation of the GH axis, but is required for fasting-induced suppression of GHRH and SRIH expression, as well as fasting-induced augmentation of pituitary GHS-R mRNA. In contrast to the rat, fasting clearly did not suppress circulating GH levels in mice, but resulted in an overall rise in mean GH levels, similar to that observed in other mammalian species. The fact that many of the fasting-induced changes in the GH axis were observed in the high-dose STZ-treated mice, but were not observed in the multiple, low-dose paradigm, suggests STZ-mediated modulation of GH axis function is dependent on the severity of the catabolic state and not hyperglycemia.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Jejum/fisiologia , Hormônio do Crescimento/biossíntese , Hipotálamo/metabolismo , Neuropeptídeo Y/fisiologia , Hipófise/metabolismo , Animais , Northern Blotting , Privação de Alimentos/fisiologia , Insulina/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuropeptídeo Y/genética , Ensaios de Proteção de Nucleases , RNA/biossíntese , RNA/isolamento & purificação , Receptores Acoplados a Proteínas G/biossíntese , Receptores de Grelina , Receptores de Neuropeptídeos/biossíntese , Receptores de Hormônios Reguladores de Hormônio Hipofisário/biossíntese , Somatostatina/biossíntese , Somatostatina/genética
14.
Endocrinology ; 145(7): 3182-9, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15044357

RESUMO

Secretion of GH by pituitary somatotropes is primarily stimulated by the hypothalamic GHRH through the activation of a specific G protein-coupled receptor, GHRH receptor (GHRH-R). GH is also released in response to ghrelin, a peptide produced in the stomach, hypothalamus, and pituitary that activates somatotropes via a distinct G protein-coupled receptor, referred to as the GH secretagogue receptor (GHS-R). Here, we have analyzed the expression of both GHRH-R and GHS-R (by multiplex RT-PCR) in porcine pituitary cell cultures, after acute (4 h) treatment with GHRH or ghrelin as well as with other regulators of somatotropes (somatostatin, dexamethasone). Exposure of cultures to GHRH decreased GHRH-R mRNA content and also diminished GHS-R transcript levels. Likewise, ghrelin down-regulated both GHS-R and GHRH-R expression. Interestingly, administration of the activator of adenylate cyclase, forskolin, decreased GHRH-R mRNA levels but had no effect on GHS-R, thus suggesting a distinct contribution of the various intracellular signals operating in somatotropes to the regulation of the expression of these receptors. Accordingly, an atypical activator of adenylate cyclase in the pig somatotrope is low-dose (10(-13) m) somatostatin, which also suppressed GHRH-R mRNA levels without altering GHS-R expression. Finally, dexamethasone did not modify GHRH-R or GHS-R expression. In summary, our data show for the first time that ghrelin, as well as GHRH, mediates homologous and heterologous down-regulation of their own receptor synthesis. However, our results also indicate that the expression of porcine GHRH-R and GHS-R is regulated by distinct signals that may differ from those reported in other mammalian species.


Assuntos
Hipófise/fisiologia , Receptores Acoplados a Proteínas G/genética , Receptores de Neuropeptídeos/genética , Receptores de Hormônios Reguladores de Hormônio Hipofisário/genética , Animais , Células Cultivadas , Colforsina/farmacologia , Dexametasona/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Grelina , Glucocorticoides/farmacologia , Hormônio Liberador de Hormônio do Crescimento/farmacologia , Hormônios Peptídicos/farmacologia , Hipófise/citologia , RNA Mensageiro/análise , Receptores de Grelina , Reprodutibilidade dos Testes , Reação em Cadeia da Polimerase Via Transcriptase Reversa/normas , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Somatostatina/farmacologia , Sus scrofa
15.
Genes Dev ; 17(11): 1352-65, 2003 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-12782654

RESUMO

To elucidate the functions of the serine/threonine kinase Akt/PKB in vivo, we generated mice lacking both akt1 and akt2 genes. Akt1/Akt2 double-knockout (DKO) mice exhibit severe growth deficiency and die shortly after birth. These mice display impaired skin development because of a proliferation defect, severe skeletal muscle atrophy because of a marked decrease in individual muscle cell size, and impaired bone development. These defects are strikingly similar to the phenotypes of IGF-1 receptor-deficient mice and suggest that Akt may serve as the most critical downstream effector of the IGF-1 receptor during development. In addition, Akt1/Akt2 DKO mice display impeded adipogenesis. Specifically, Akt1 and Akt2 are required for the induced expression of PPARgamma, the master regulator of adipogenesis, establishing a new essential role for Akt in adipocyte differentiation. Overall, the combined deletion of Akt1 and Akt2 establishes in vivo roles for Akt in cell proliferation, growth, and differentiation. These functions of Akt were uncovered despite the observed lower level of Akt activity mediated by Akt3 in Akt1/Akt2 DKO cells, suggesting that a critical threshold level of Akt activity is required to maintain normal cell proliferation, growth, and differentiation.


Assuntos
Desenvolvimento Embrionário e Fetal/genética , Regulação da Expressão Gênica no Desenvolvimento , Músculo Esquelético/patologia , Osteogênese/genética , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Anormalidades Múltiplas/embriologia , Anormalidades Múltiplas/genética , Adipócitos/citologia , Adipócitos/patologia , Adipócitos/fisiologia , Tecido Adiposo/crescimento & desenvolvimento , Animais , Atrofia , Cruzamentos Genéticos , Nanismo/genética , Feminino , Genótipo , Heterozigoto , Masculino , Camundongos , Camundongos Knockout , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas , Proteínas Proto-Oncogênicas c-akt , Pele/crescimento & desenvolvimento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA