Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Endocrinology ; 157(8): 3157-66, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27254000

RESUMO

Pheochromocytomas (PCCs) are tumors arising from neural crest-derived chromaffin cells. There are currently few animal models of PCC that recapitulate the key features of human tumors. Because such models may be useful for investigations of molecular pathomechanisms and development of novel therapeutic interventions, we characterized a spontaneous animal model (multiple endocrine neoplasia [MENX] rats) that develops endogenous PCCs with complete penetrance. Urine was longitudinally collected from wild-type (wt) and MENX-affected (mutant) rats and outputs of catecholamines and their O-methylated metabolites determined by mass spectrometry. Adrenal catecholamine contents, cellular ultrastructure, and expression of phenylethanolamine N-methyltransferase, which converts norepinephrine to epinephrine, were also determined in wt and mutant rats. Blood pressure was longitudinally measured and end-organ pathology assessed. Compared with wt rats, mutant animals showed age-dependent increases in urinary outputs of norepinephrine (P = .0079) and normetanephrine (P = .0014) that correlated in time with development of tumor nodules, increases in blood pressure, and development of hypertension-related end-organ pathology. Development of tumor nodules, which lacked expression of N-methyltransferase, occurred on a background of adrenal medullary morphological and biochemical changes occurring as early as 1 month of age and involving increased adrenal medullary concentrations of dense cored vesicles, tissue contents of both norepinephrine and epinephrine, and urinary outputs of metanephrine, the metabolite of epinephrine. Taken together, MENX-affected rats share several biochemical and pathophysiological features with PCC patients. This model thus provides a suitable platform to study the pathogenesis of PCC for preclinical translational studies aimed at the development of novel therapies for aggressive forms of human tumors.


Assuntos
Neoplasias das Glândulas Suprarrenais/metabolismo , Neoplasias das Glândulas Suprarrenais/patologia , Neoplasia Endócrina Múltipla/metabolismo , Neoplasia Endócrina Múltipla/patologia , Feocromocitoma/metabolismo , Feocromocitoma/patologia , Neoplasias das Glândulas Suprarrenais/fisiopatologia , Medula Suprarrenal/metabolismo , Medula Suprarrenal/patologia , Animais , Pressão Sanguínea/fisiologia , Modelos Animais de Doenças , Epinefrina/sangue , Feminino , Masculino , Metanefrina/sangue , Neoplasia Endócrina Múltipla/fisiopatologia , Norepinefrina/sangue , Normetanefrina/sangue , Feocromocitoma/fisiopatologia , Ratos , Ratos Mutantes , Ratos Sprague-Dawley
2.
Theranostics ; 6(5): 650-65, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27022413

RESUMO

Pheochromocytomas and extra-adrenal paragangliomas (PHEO/PGLs) are rare catecholamine-producing chromaffin cell tumors. For metastatic disease, no effective therapy is available. Overexpression of somatostatin type 2 receptors (SSTR2) in PHEO/PGLs promotes interest in applying therapies using somatostatin analogs linked to radionuclides and/or cytotoxic compounds, such as [(177)Lu]Lu-DOTA-(Tyr(3))octreotate (DOTATATE) and AN-238. Systematic evaluation of such therapies for the treatment of PHEO/PGLs requires sophisticated animal models. In this study, the mouse pheochromocytoma (MPC)-mCherry allograft model showed high tumor densities of murine SSTR2 (mSSTR2) and high tumor uptake of [(64)Cu]Cu-DOTATATE. Using tumor sections, we assessed mSSTR2-specific binding of DOTATATE, AN-238, and somatostatin-14. Therapeutic studies showed substantial reduction of tumor growth and tumor-related renal monoamine excretion in tumor-bearing mice after treatment with [(177)Lu]Lu-DOTATATE compared to AN-238 and doxorubicin. Analyses did not show agonist-dependent receptor downregulation after single mSSTR2-targeting therapies. This study demonstrates that the MPC-mCherry model is a uniquely powerful tool for the preclinical evaluation of SSTR2-targeting theranostic applications in vivo. Our findings highlight the therapeutic potential of somatostatin analogs, especially of [(177)Lu]Lu-DOTATATE, for the treatment of metastatic PHEO/PGLs. Repeated treatment cycles, fractionated combinations of SSTR2-targeting radionuclide and cytotoxic therapies, and other adjuvant compounds addressing additional mechanisms may further enhance therapeutic outcome.


Assuntos
Antineoplásicos/administração & dosagem , Doxorrubicina/análogos & derivados , Octreotida/análogos & derivados , Feocromocitoma/tratamento farmacológico , Compostos Radiofarmacêuticos/administração & dosagem , Nanomedicina Teranóstica/métodos , Animais , Modelos Animais de Doenças , Doxorrubicina/administração & dosagem , Camundongos , Octreotida/administração & dosagem , Peptídeos Cíclicos , Pirróis/administração & dosagem , Receptores de Somatostatina/metabolismo , Resultado do Tratamento
3.
Mol Cell Biol ; 36(3): 376-93, 2016 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-26572826

RESUMO

Angiogenesis is a central regulator for white (WAT) and brown (BAT) adipose tissue adaptation in the course of obesity. Here we show that deletion of hypoxia-inducible factor 2α (HIF2α) in adipocytes (by using Fabp4-Cre transgenic mice) but not in myeloid or endothelial cells negatively impacted WAT angiogenesis and promoted WAT inflammation, WAT dysfunction, hepatosteatosis, and systemic insulin resistance in obesity. Importantly, adipocyte HIF2α regulated vascular endothelial growth factor (VEGF) expression and angiogenesis of obese BAT as well as its thermogenic function. Consistently, obese adipocyte-specific HIF2α-deficient mice displayed BAT dysregulation, associated with reduced levels of uncoupling protein 1 (UCP1) and a dysfunctional thermogenic response to cold exposure. VEGF administration reversed WAT and BAT inflammation and BAT dysfunction in adipocyte HIF2α-deficient mice. Together, our findings show that adipocyte HIF2α is protective against maladaptation to obesity and metabolic dysregulation by promoting angiogenesis in both WAT and BAT and by counteracting obesity-mediated BAT dysfunction.


Assuntos
Adipócitos/patologia , Tecido Adiposo Marrom/fisiopatologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Deleção de Genes , Obesidade/genética , Obesidade/fisiopatologia , Adipócitos/metabolismo , Tecido Adiposo Marrom/irrigação sanguínea , Tecido Adiposo Marrom/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Inflamação/complicações , Canais Iônicos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Proteínas Mitocondriais/metabolismo , Neovascularização Fisiológica , Obesidade/complicações , Obesidade/metabolismo , Termogênese , Proteína Desacopladora 1 , Fator A de Crescimento do Endotélio Vascular/metabolismo
4.
Mol Cell Endocrinol ; 408: 178-84, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25575455

RESUMO

The adrenal is a highly plastic organ with the ability to adjust to physiological needs by adapting hormone production but also by generating and regenerating both adrenocortical and adrenomedullary tissue. It is now apparent that many adult tissues maintain stem and progenitor cells that contribute to their maintenance and adaptation. Research from the last years has proven the existence of stem and progenitor cells also in the adult adrenal medulla throughout life. These cells maintain some neural crest properties and have the potential to differentiate to the endocrine and neural lineages. In this article, we discuss the evidence for the existence of adrenomedullary multi potent progenitor cells, their isolation and characterization, their differentiation potential as well as their clinical potential in transplantation therapies but also in pathophysiology.


Assuntos
Medula Suprarrenal/citologia , Separação Celular/métodos , Células-Tronco Multipotentes/citologia , Medula Suprarrenal/transplante , Animais , Carcinogênese/patologia , Humanos , Modelos Biológicos , Transplante de Células-Tronco
5.
Endocrinology ; 155(11): 4149-56, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25137029

RESUMO

Pheochromocytoma (PHEO) is a rare but potentially lethal neuroendocrine tumor arising from catecholamine-producing chromaffin cells. Especially for metastatic PHEO, the availability of animal models is essential for developing novel therapies. For evaluating therapeutic outcome in rodent PHEO models, reliable quantification of multiple organ lesions depends on dedicated small-animal in vivo imaging, which is still challenging and only available at specialized research facilities. Here, we investigated whether whole-body fluorescence imaging and monitoring of urinary free monoamines provide suitable parameters for measuring tumor progression in a murine allograft model of PHEO. We generated an mCherry-expressing mouse PHEO cell line by lentiviral gene transfer. These cells were injected subcutaneously into nude mice to perform whole-body fluorescence imaging of tumor development. Urinary free monoamines were measured by liquid chromatography with tandem mass spectrometry. Tumor fluorescence intensity and urinary outputs of monoamines showed tumor growth-dependent increases (P < .001) over the 30 days of monitoring post-tumor engraftment. Concomitantly, systolic blood pressure was increased significantly during tumor growth. Tumor volume correlated significantly (P < .001) and strongly with tumor fluorescence intensity (rs = 0.946), and urinary outputs of dopamine (rs = 0.952), methoxytyramine (rs = 0.947), norepinephrine (rs = 0.756), and normetanephrine (rs = 0.949). Dopamine and methoxytyramine outputs allowed for detection of lesions at diameters below 2.3 mm. Our results demonstrate that mouse pheochromocytoma (MPC)-mCherry cell tumors are functionally similar to human PHEO. Both tumor fluorescence intensity and urinary outputs of free monoamines provide precise parameters of tumor progression in this sc mouse model of PHEO. This animal model will allow for testing new treatment strategies for chromaffin cell tumors.


Assuntos
Neoplasias das Glândulas Suprarrenais/diagnóstico , Aminas/urina , Biomarcadores Tumorais/urina , Imagem Óptica/métodos , Feocromocitoma/diagnóstico , Neoplasias das Glândulas Suprarrenais/patologia , Neoplasias das Glândulas Suprarrenais/urina , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Progressão da Doença , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Nus , Feocromocitoma/patologia , Feocromocitoma/urina , Urinálise
6.
Int J Radiat Biol ; 90(8): 700-9, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24512568

RESUMO

PURPOSE: In this work we examined the presence of the neural stem cell biomarker Hairy and Enhancer of Split 3 (Hes3) in the anterior eye segment and in the aberrant growth condition of the conjunctiva pterygium. Further, we studied the response of Hes3 to irradiation. MATERIALS AND METHODS: Adult mouse and human corneoscleral junction and conjunctiva, as well as human pterygium were prepared for immunohistochemical detection of Hes3 and other markers. Total body irradiation was used to study the changes in the pattern of Hes3 expression. RESULTS: The adult rodent and human eye as well as pterygium, contain a population of cells expressing Hes3. In the human eye, Hes3-expressing (Hes3+) cells are found predominantly in the subconjunctival space spanning over the limbus where they physically associate with blood vessels. The cytoarchitecture of Hes3 + cells is similar to those previously observed in the adult central nervous system. Furthermore, irradiation reduces the number of Hes3 + cells in the subconjunctival space. In contrast, irradiation strongly promotes the nuclear localization of Hes3 in the ciliary body epithelium. CONCLUSIONS: Our results suggest that a recently identified signal transduction pathway that regulates neural stem cells and glioblastoma cancer stem cells also operates in the ocular surface, ciliary body, and in pterygium.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas de Ligação a DNA/metabolismo , Olho/metabolismo , Regulação da Expressão Gênica , Proteínas do Tecido Nervoso/metabolismo , Pterígio/metabolismo , Fatores de Transcrição/metabolismo , Animais , Túnica Conjuntiva/efeitos dos fármacos , Túnica Conjuntiva/metabolismo , Túnica Conjuntiva/efeitos da radiação , Olho/irrigação sanguínea , Olho/efeitos dos fármacos , Olho/efeitos da radiação , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos da radiação , Humanos , Camundongos , Terapia de Alvo Molecular , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/efeitos da radiação , Pterígio/tratamento farmacológico , Pterígio/fisiopatologia , Proteínas Repressoras
7.
J Immunol ; 191(8): 4367-74, 2013 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-24043887

RESUMO

Obese adipose tissue (AT) inflammation contributes critically to development of insulin resistance. The complement anaphylatoxin C5a receptor (C5aR) has been implicated in inflammatory processes and as regulator of macrophage activation and polarization. However, the role of C5aR in obesity and AT inflammation has not been addressed. We engaged the model of diet-induced obesity and found that expression of C5aR was significantly upregulated in the obese AT, compared with lean AT. In addition, C5a was present in obese AT in the proximity of macrophage-rich crownlike structures. C5aR-sufficient and -deficient mice were fed a high-fat diet (HFD) or a normal diet (ND). C5aR deficiency was associated with increased AT weight upon ND feeding in males, but not in females, and with increased adipocyte size upon ND and HFD conditions in males. However, obese C5aR(-/-) mice displayed improved systemic and AT insulin sensitivity. Improved AT insulin sensitivity in C5aR(-/-) mice was associated with reduced accumulation of total and proinflammatory M1 macrophages in the obese AT, increased expression of IL-10, and decreased AT fibrosis. In contrast, no difference in ß cell mass was observed owing to C5aR deficiency under an HFD. These results suggest that C5aR contributes to macrophage accumulation and M1 polarization in the obese AT and thereby to AT dysfunction and development of AT insulin resistance.


Assuntos
Tecido Adiposo/imunologia , Tecido Adiposo/metabolismo , Resistência à Insulina/imunologia , Macrófagos/imunologia , Receptor da Anafilatoxina C5a/metabolismo , Adipócitos/imunologia , Adipócitos/metabolismo , Animais , Complemento C5a/metabolismo , Gorduras na Dieta/imunologia , Gorduras na Dieta/metabolismo , Feminino , Fibrose/imunologia , Inflamação/imunologia , Células Secretoras de Insulina/metabolismo , Interleucina-10/biossíntese , Ativação de Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/imunologia , Obesidade/metabolismo , Receptor da Anafilatoxina C5a/biossíntese , Receptor da Anafilatoxina C5a/imunologia , Regulação para Cima
8.
J Mol Endocrinol ; 51(1): 167-79, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23718963

RESUMO

UNLABELLED: Non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis are components of the metabolic syndrome. Serum leptin levels are elevated in obesity, but the role of leptin in the pathophysiology of the liver involvement is still unclear. To identify the effects and mechanisms by which leptin influences the pathogenesis of NAFLD, we performed epididymal white adipose tissue (eWAT) transplantation from congenic wild-type mice into the subcutaneous dorsal area of Lep(ob/ob) recipient mice and compared the results with those of the Lep(ob/ob) sham-operated mice. The mice were followed for 102-216 days. During killing, the transplanted mice had significantly lost body weight and exhibited significantly higher leptin levels, improved glucose tolerance, and lower liver injury scores than the sham-operated mice. Liver microarray analysis showed that novel pathways related to GA-binding protein (GABP) transcription factor targets, pheromone binding, and olfactory signaling were differentially expressed in the transplanted mice. Our data also replicate pathways known to be involved in NAFLD, such as those involved in the regulation of microRNAs, lipid, glucose, and glutathione metabolism, peroxisome proliferator-activated receptor signaling, cellular regulation, carboxylic acid processes, iron, heme, and tetrapyrrole binding, immunity and inflammation, insulin signaling, cytochrome P450 function, and cancer. CONCLUSION: wild-type eWAT transplantation into Lep(ob/ob) mice led to improvements in metabolism, body weight, and liver injury, possibly attributed to the production of leptin by the transplanted eWAT. These improvements were accompanied by the differential expression of novel pathways. The causal relationship between GABP downregulation and NAFLD improvement remains to be determined.


Assuntos
Fígado Gorduroso/genética , Fígado Gorduroso/metabolismo , Transdução de Sinais , Tecido Adiposo Branco/metabolismo , Tecido Adiposo Branco/transplante , Animais , Ácidos Graxos/metabolismo , Fígado Gorduroso/imunologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Teste de Tolerância a Glucose , Hormônios/sangue , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Insulina/metabolismo , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos Congênicos , Camundongos Transgênicos , Hepatopatia Gordurosa não Alcoólica , Esteroides/metabolismo
9.
PLoS Biol ; 11(3): e1001505, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23554573

RESUMO

Hedgehog (Hh) proteins control animal development and tissue homeostasis. They activate gene expression by regulating processing, stability, and activation of Gli/Cubitus interruptus (Ci) transcription factors. Hh proteins are secreted and spread through tissue, despite becoming covalently linked to sterol during processing. Multiple mechanisms have been proposed to release Hh proteins in distinct forms; in Drosophila, lipoproteins facilitate long-range Hh mobilization but also contain lipids that repress the pathway. Here, we show that mammalian lipoproteins have conserved roles in Sonic Hedgehog (Shh) release and pathway repression. We demonstrate that lipoprotein-associated forms of Hh and Shh specifically block lipoprotein-mediated pathway inhibition. We also identify a second conserved release form that is not sterol-modified and can be released independently of lipoproteins (Hh-N*/Shh-N*). Lipoprotein-associated Hh/Shh and Hh-N*/Shh-N* have complementary and synergistic functions. In Drosophila wing imaginal discs, lipoprotein-associated Hh increases the amount of full-length Ci, but is insufficient for target gene activation. However, small amounts of non-sterol-modified Hh synergize with lipoprotein-associated Hh to fully activate the pathway and allow target gene expression. The existence of Hh secretion forms with distinct signaling activities suggests a novel mechanism for generating a diversity of Hh responses.


Assuntos
Proteínas de Drosophila/metabolismo , Proteínas Hedgehog/metabolismo , Lipoproteínas/metabolismo , Animais , Drosophila , Células HeLa , Humanos , Imunoprecipitação , Mamíferos , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo , Proteína GLI1 em Dedos de Zinco
10.
Cell Transplant ; 21(11): 2471-86, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22507143

RESUMO

The differentiation of dopamine-producing neurons from chromaffin progenitors might represent a new valuable source for replacement therapies in Parkinson's disease. However, characterization of their differentiation potential is an important prerequisite for efficient engraftment. Based on our previous studies on isolation and characterization of chromaffin progenitors from adult adrenals, this study investigates their potential to produce dopaminergic neurons and means to enhance their dopaminergic differentiation. Chromaffin progenitors grown in sphere culture showed an increased expression of nestin and Mash1, indicating an increase of the progenitor subset. Proneurogenic culture conditions induced the differentiation into neurons positive for neural markers ß-III-tubulin, MAP2, and TH accompanied by a decrease of Mash1 and nestin. Furthermore, Notch2 expression decreased concomitantly with a downregulation of downstream effectors Hes1 and Hes5 responsible for self-renewal and proliferation maintenance of progenitor cells. Chromaffin progenitor-derived neurons secreted dopamine upon stimulation by potassium. Strikingly, treatment of differentiating cells with retinoic and ascorbic acid resulted in a twofold increase of dopamine secretion while norepinephrine and epinephrine were decreased. Initiation of dopamine synthesis and neural maturation is controlled by Pitx3 and Nurr1. Both Pitx3 and Nurr1 were identified in differentiating chromaffin progenitors. Along with the gained dopaminergic function, electrophysiology revealed features of mature neurons, such as sodium channels and the capability to fire multiple action potentials. In summary, this study elucidates the capacity of chromaffin progenitor cells to generate functional dopaminergic neurons, indicating their potential use in cell replacement therapies.


Assuntos
Células Cromafins/citologia , Neurônios Dopaminérgicos/citologia , Células-Tronco/citologia , Animais , Bovinos , Diferenciação Celular/fisiologia , Proliferação de Células , Células Cultivadas , Células Cromafins/metabolismo , Neurônios Dopaminérgicos/metabolismo , Células-Tronco/metabolismo , Tretinoína/metabolismo
11.
Proc Natl Acad Sci U S A ; 108(33): 13722-7, 2011 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-21825133

RESUMO

Corticotropin-releasing hormone (CRH) and growth hormone-releasing hormone (GHRH), primarily characterized as neuroregulators of the hypothalamic-pituitary-adrenal axis, directly influence tissue-specific receptor-systems for CRH and GHRH in the endocrine pancreas. Here, we demonstrate the expression of mRNA for CRH and CRH-receptor type 1 (CRHR1) and of protein for CRHR1 in rat and human pancreatic islets and rat insulinoma cells. Activation of CRHR1 and GHRH-receptor significantly increased cell proliferation and reduced cell apoptosis. CRH stimulated both cellular content and release of insulin in rat islet and insulinoma cells. At the ultrastructural level, CRHR1 stimulation revealed a more active metabolic state with enlarged mitochondria. Moreover, glucocorticoids that promote glucose production are balanced by both 11b-hydroxysteroid dehydrogenase (11ß-HSD) isoforms; 11ß-HSD-type-1 and 11ß-HSD-type-2. We demonstrated expression of mRNA for 11ß-HSD-1 and 11ß-HSD-2 and protein for 11ß-HSD-1 in rat and human pancreatic islets and insulinoma cells. Quantitative real-time PCR revealed that stimulation of CRHR1 and GHRH-receptor affects the metabolism of insulinoma cells by down-regulating 11ß-HSD-1 and up-regulating 11ß-HSD-2. The 11ß-HSD enzyme activity was analyzed by measuring the production of cortisol from cortisone. Similarly, activation of CRHR1 resulted in reduced cortisol levels, indicating either decreased 11ß-HSD-1 enzyme activity or increased 11ß-HSD-2 enzyme activity; thus, activation of CRHR1 alters the glucocorticoid balance toward the inactive form. These data indicate that functional receptor systems for hypothalamic-releasing hormone agonists exist within the endocrine pancreas and influence synthesis of insulin and the pancreatic glucocorticoid shuttle. Agonists of CRHR1 and GHRH-receptor, therefore, may play an important role as novel therapeutic tools in the treatment of diabetes mellitus.


Assuntos
11-beta-Hidroxiesteroide Desidrogenases/fisiologia , Sistema Hipotálamo-Hipofisário/metabolismo , Ilhotas Pancreáticas/metabolismo , Hormônios Liberadores de Hormônios Hipofisários/fisiologia , Sistema Hipófise-Suprarrenal/metabolismo , Animais , Hormônio Liberador da Corticotropina , Humanos , Insulina/biossíntese , Insulinoma/patologia , RNA Mensageiro , Ratos , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Receptores de Neuropeptídeos/metabolismo , Receptores de Hormônios Reguladores de Hormônio Hipofisário/metabolismo
12.
Mol Cell Endocrinol ; 336(1-2): 149-55, 2011 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-21195131

RESUMO

Dehydroepiandrosterone sulfate is classically seen as an inactive reservoir for the production of dehydroepiandrosterone. Steroid sulfatase is the enzyme that catalyzes the hydrolysis of dehydroepiandrosterone sulfate to dehydroepiandrosterone, which can then be further metabolized to other steroid hormones. Recent studies, however, indicate that dehydroepiandrosterone sulfate can mediate biological effects without being converted to dehydroepiandrosterone. This study aims to evaluate whether dehydroepiandrosterone sulfate itself influences the differentiation of PC-12 cells or if its desulfation to dehydroepiandrosterone is required. dehydroepiandrosterone and dehydroepiandrosterone sulfate both influence the differentiation of chromaffin PC-12 cells. Blocking steroid sulfatase activity and thereby the conversion of dehydroepiandrosterone sulfate to dehydroepiandrosterone by the enzyme blocker estrone sulfamate showed that the effect of dehydroepiandrosterone sulfate is independent of its conversion to dehydroepiandrosterone. Dehydroepiandrosterone sulfate, similar to dehydroepiandrosterone, reduced nerve growth factor-induced neurite outgrowth of PC-12 cells and the expression of synaptosomal-associated membrane protein of 25 kDa, increased the expression of chromogranin A and significantly increased dopamine release of PC-12 cells. In addition, dehydroepiandrosterone sulfate, dehydroepiandrosterone and membrane impermeable dehydroepiandrosterone-BSA all significantly reduced NGF-induced MAPK ERK1/2 signaling after 5 min. In summary, this study provides evidence that dehydroepiandrosterone sulfate, independent of its conversion to dehydroepiandrosterone, directs PC-12 cells' differentiation to a neuroendocrine direction. Furthermore, employing membrane-impermeable dehydroepiandrosterone-BSA indicates the involvement of plasma-membrane bound receptors.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Sulfato de Desidroepiandrosterona/farmacologia , Animais , Biomarcadores/metabolismo , Permeabilidade da Membrana Celular/efeitos dos fármacos , Cromogranina A/genética , Cromogranina A/metabolismo , Desidroepiandrosterona/análogos & derivados , Desidroepiandrosterona/farmacologia , Dopamina/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Fator de Crescimento Neural/farmacologia , Neuritos/efeitos dos fármacos , Neuritos/metabolismo , Sistemas Neurossecretores/efeitos dos fármacos , Sistemas Neurossecretores/metabolismo , Células PC12 , Fosforilação/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Soroalbumina Bovina/farmacologia , Esteril-Sulfatase/genética , Esteril-Sulfatase/metabolismo , Proteína 25 Associada a Sinaptossoma/metabolismo , Fatores de Tempo
13.
Mol Cell Endocrinol ; 336(1-2): 41-6, 2011 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-21167252

RESUMO

Rapid activation of adrenal steroid release plays a pivotal role in an organism's first line of defense during sepsis. Adrenal gland function is often suppressed in critically ill patients and negatively impacts the overall survival rate. Increasingly, experimental and clinical evidence suggests that Toll-like receptors (TLRs), components of the innate immune system, play a key role in the mediation of systemic responses to invading pathogens during sepsis. In the present study, we aimed to elucidate the effect of TLR2, TLR4 and CD14 upregulation on adrenocortical cell steroidogenesis. We found that TLR4 and CD14 but not TLR2 overexpression in NCI-H295R cells inhibited basal and acute cortisol and aldosterone production. This effect could be partially explained by reduced expression of enzymes involved in the synthesis of latter steroids--CYP11B1 and CYP11B2. Together, these data suggest that TLR upregulation in the steroid producing cells may be involved in the adrenal gland dysfunction during sepsis.


Assuntos
Córtex Suprarrenal/citologia , Córtex Suprarrenal/metabolismo , Esteroides/biossíntese , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Regulação para Cima , Córtex Suprarrenal/enzimologia , Linhagem Celular , Colesterol/metabolismo , Células Clonais , Humanos , Receptores de Lipopolissacarídeos/metabolismo
14.
J Clin Endocrinol Metab ; 95(12): E421-9, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20826579

RESUMO

CONTEXT: Adrenocortical carcinoma (ACC) is a rare tumor with poor prognosis. The expression of innate immunity receptor Toll-like receptor 4 (TLR4) was recently reported in various human tumors, and TLR4 was shown to regulate tumor immune escape processes, proliferation, and resistance to chemotherapeutical agents. OBJECTIVE: The aim of this study was to investigate TLR4 expression, signaling, and function in the process of tumorigenesis in the human adrenal cortex. MEASUREMENTS AND MAIN RESULTS: Real-time PCR analysis of human ACC (n=8), adenoma (n=8), and ACC cell lines (SW13, NCI-H295R, and HAC15) revealed a significant down-regulation of TLR4, MD2 (myeloid differentiation protein-2), and cluster of differentiation 14 (CD14) mRNA compared with normal human adrenal cortex and adrenocortical cells in primary culture. Furthermore, immunohistochemistry revealed an abrogation of TLR4 and CD14 expression in ACC but not adenoma tissues. Western blot analysis of MAPK, AKT, activator protein-1, and nuclear factor-κB signaling revealed that the ACC cell lines are unresponsive to lipopolysaccharide action. Restoration of TLR4 signaling by stable transfection of TLR4-CD14 plasmid into NCI-H295R cells sensitized them to lipopolysaccharide incubation as shown by nuclear factor-κB activation and decreased cell viability and induced apoptosis in these cells. CONCLUSION: Our results demonstrate a significant reduction in the expression of TLR4 and CD14 and an inactivation of TLR4 signaling in ACCs. Furthermore, our data show that reintroduction of TLR4 expression in ACCs may provide a novel therapeutic strategy for adrenal cancer.


Assuntos
Neoplasias do Córtex Suprarrenal/genética , Receptores de Lipopolissacarídeos/genética , Receptor 4 Toll-Like/genética , Adenoma/genética , Adenoma/patologia , Córtex Suprarrenal/patologia , Córtex Suprarrenal/fisiologia , Neoplasias do Córtex Suprarrenal/imunologia , Neoplasias do Córtex Suprarrenal/cirurgia , Adulto , Animais , Western Blotting , Divisão Celular , Linhagem Celular Tumoral , Regulação para Baixo , Feminino , Regulação da Expressão Gênica , Humanos , Imuno-Histoquímica , Receptores de Lipopolissacarídeos/metabolismo , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase/métodos , RNA Mensageiro/genética , Valores de Referência , Receptor 4 Toll-Like/metabolismo
15.
Proc Natl Acad Sci U S A ; 107(28): 12623-8, 2010 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-20616039

RESUMO

Therapeutic strategies for transplantation of pancreatic islet cells are urgently needed to expand beta-cell mass by stimulating islet cell proliferation and/or prolonging islet cell survival. Control of the islets by different growth factors provides a potential venue for augmenting beta-cell mass. In the present study, we show the expression of the biologically active splice variant-1 (SV-1) of growth hormone-releasing hormone (GHRH) receptor in rat insulinoma (INS-1) cells as well as in rat and human pancreatic islets. In studies in vitro of INS-1 cells, the GHRH agonist JI-36 caused a significant increase in cell proliferation and a reduction of cell apoptosis. JI-36 increased islet size and glucose-stimulated insulin secretion in isolated rat islets after 48-72 h. At the ultrastructural level, INS-1 cells treated with agonist JI-36 revealed a metabolic active stimulation state with increased cytoplasm. Coincubation with the GHRH antagonist MIA-602 reversed the actions of the agonist JI-36, indicating the specificity of this agonist. In vivo, the function of pancreatic islets was assessed by transplantation of rat islets under the kidney capsule of streptozotocin-induced diabetic non-obese diabetic-severe combined immunodeficiency (NOD-SCID) mice. Islets treated with GHRH agonist JI-36 were able to achieve normoglycemia earlier and more consistently than untreated islets. Furthermore, in contrast to diabetic animals transplanted with untreated islets, insulin response to an i.p. glucose tolerance test (IPGTT) in animals receiving islets treated with agonist Jl-36 was comparable to that of normal healthy mice. In conclusion, our study provides evidence that agonists of GHRH represent a promising pharmacological therapy aimed at promoting islet graft growth and proliferation in diabetic patients.


Assuntos
Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/fisiologia , Animais , Apoptose , Proliferação de Células , Glucose/metabolismo , Teste de Tolerância a Glucose , Hormônio do Crescimento/metabolismo , Hormônio Liberador de Hormônio do Crescimento/metabolismo , Hormônios/metabolismo , Hormônio do Crescimento Humano/metabolismo , Humanos , Insulina/biossíntese , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/fisiologia , Insulinoma/cirurgia , Ilhotas Pancreáticas/cirurgia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Ratos , Ratos Wistar
16.
Am J Hypertens ; 23(10): 1061-8, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20559286

RESUMO

BACKGROUND: Serum aldosterone is a causative factor for various metabolic and cardiovascular disorders. Low-density lipoprotein (LDL) is a major cholesterol source for aldosterone steroidogenesis; however, the effect of oxidative modification of LDL on aldosterone release is not known. We studied the effect of hypochlorite-oxidized LDL (oxLDL) on adrenal aldosterone secretion. METHODS: LDL (native LDL (natLDL)) was obtained from healthy volunteers and oxidatively modified in vitro. NCI-H295R cells were stimulated with natLDL and oxLDL, and the aldosterone release was quantified by radioimmunoassay. Molecular changes were studied with western blot analysis and quantitative RT-PCR analysis. RESULTS: NatLDL and oxLDL caused dose-dependent increase in aldosterone release up to threefold. However, the stimulatory effects of modified LDL on aldosterone secretion decreased with increasing degree of LDL oxidation. 24-h incubations with natLDL, mild- and medium-oxidized LDL sensitized the adrenocortical cells to subsequent angiotensin II (Ang II) stimulations by 2.9-, 2.8-, and 2.5-folds, respectively. Heavily oxidized LDL did not sensitize the cells to Ang II stimulations to a similar extent. At the molecular level, the ERK pathway was activated within a minute by both natLDL and oxLDL; however, oxLDL showed a stronger (2.75-fold at 1 and 15 min) and longer (15 min) activation of ERK than natLDL (twofold). CONCLUSIONS: This study demonstrates the following: (i) both natLDL and hypochlorite-oxidized LDL utilize ERK pathway to mediate aldosterone release; (ii) mildly oxidized LDL sensitizes the adrenocortical cells to further stimulations by Ang II similar to natLDL that may have a role in pathological processes; (iii) extensive LDL oxidation counteracts adrenocortical aldosterone release.


Assuntos
Glândulas Suprarrenais/efeitos dos fármacos , Glândulas Suprarrenais/metabolismo , Aldosterona/metabolismo , Lipoproteínas LDL/química , Lipoproteínas LDL/farmacologia , Córtex Suprarrenal/efeitos dos fármacos , Córtex Suprarrenal/metabolismo , Angiotensina II/farmacologia , Western Blotting , Linhagem Celular , Relação Dose-Resposta a Droga , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Ácido Hipocloroso/química , Oxidantes/química , RNA/biossíntese , RNA/isolamento & purificação , Receptores de LDL/efeitos dos fármacos , Receptores de LDL/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Esteroides/biossíntese , Estimulação Química , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo
17.
Mech Ageing Dev ; 131(2): 124-32, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20074583

RESUMO

The potential use of neural stem cells in basic research, drug testing and for development of therapeutic strategies requires large scale in vitro amplification, increasing the probability of genetic instability and transformation. Little is known, however, about potential correlations between long-term culture of neural stem and progenitor cells (NSPCs), changed differentiation and self-renewal capacities, and the occurrence of chromosomal instability. This study investigates the effect of extended culture time on self-renewal, differentiation capacity, cell cycle phase distribution, telomere length, telomerase activity and chromosomal stability on fetal brain-derived cells that form floating sphere colonies (neurospheres). We observed that increased sphere-forming capacity indicative of increased proliferation was accompanied by a decreased ability to differentiate into neural lineages. The high mobility group A (Hmga2) gene positively regulates self-renewal via repression of p16(Ink4a) and p19(ARF) gene expression. This study discerned an upregulation of Hmga2 gene and protein expression and decreased p16(Ink4a) and p19(ARF) gene expression, suggesting that Hmga2 might promote the proliferation of neurosphere cells in long-term culture. Further, our analyses revealed a significant decrease in telomere length after 4 weeks of culturing that is paralleled by a moderate upregulation of telomerase activity. Importantly, regular gain of chromosome 1 with random structural chromosomal aberrations was observed within 16 weeks of neurosphere cell culture. Genetic instability and diminished differentiation capacity seem to be a consequence of long-term culture of neurosphere cells. These data indicate the necessity to analyze self-renewal, differentiation capacity, telomere length, tumor suppressor genes and chromosomal stability in neurosphere cultures prior to their usage in basic research, drug testing or the development of therapeutic strategies.


Assuntos
Ciclo Celular/genética , Diferenciação Celular/genética , Células-Tronco Multipotentes/metabolismo , Sistema Nervoso/metabolismo , Neurônios/metabolismo , Animais , Encéfalo/metabolismo , Células Cultivadas , Embrião de Mamíferos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Sistema Nervoso/embriologia , Esferoides Celulares/metabolismo , Fatores de Tempo
18.
Cytotherapy ; 12(1): 17-30, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19878082

RESUMO

BACKGROUND AIMS: Human adult bone marrow (BM)-derived mesenchymal stromal cells (hMSC) are reported to break germ layer commitment and differentiate into cells expressing neuroectodermal properties. Although it is of pivotal interest for cell replacement therapies for neurologic disorders, no data exist on the influence of the donor's age on this multipotent differentiation behavior. METHODS: We evaluated various epigenetic neuroectodermal conversion protocols in adult hMSC derived from older donors (>45 versus 18-35 years of age) using quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) and immunocytochemistry. The protocols included single- and multi-step conversion-differentiation protocols combined with co-culture techniques. Furthermore, the age dependency of mesodermal differentiation potential and cell senescence were investigated. RESULTS: The neuroectodermal differentiation potential of hMSC derived from old donors was completely lost, with no cells showing mature neuroectodermal phenotypes using single- and multi-step conversion-differentiation protocols and no improvement of neurogenesis by various co-culture conditions. Comparison of young versus old donor-derived hMSC showed fewer cells expressing early neuroectodermal marker proteins in the latter samples. qRT-PCR showed reduced expression of the proliferation marker KI67 and the neuroectodermal gene NES (nestin) in old donor-derived cells compared with young donor hMSC. Telomere length analysis showed no general cell aging. CONCLUSIONS: Our data provide evidence that only young donor-derived hMSC can be epigenetically differentiated in vitro into neuroectodermal cells, pointing towards senescence of multipotentiality of old donor-derived hMSC. There is thus an urgent need to develop better protocols for successful neuroectodermal differentiation of hMSC from old individuals as a prerequisite for autologous cell replacement strategies for neurologic diseases in elderly patients.


Assuntos
Diferenciação Celular/genética , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/fisiologia , Placa Neural/fisiologia , Células Estromais/fisiologia , Células Estromais/transplante , Adolescente , Adulto , Fatores Etários , Idoso , Envelhecimento/fisiologia , Biomarcadores/metabolismo , Proliferação de Células , Senescência Celular/fisiologia , Técnicas de Cocultura , Contraindicações , Feminino , Humanos , Proteínas de Filamentos Intermediários/metabolismo , Antígeno Ki-67/metabolismo , Masculino , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Células-Tronco Mesenquimais/citologia , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/metabolismo , Sistema Nervoso/citologia , Sistema Nervoso/embriologia , Sistema Nervoso/metabolismo , Nestina , Placa Neural/citologia , Células Estromais/citologia , Transplante Autólogo/efeitos adversos , Transplante Autólogo/métodos , Adulto Jovem
19.
Am J Physiol Regul Integr Comp Physiol ; 297(5): R1601-9, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19793950

RESUMO

Hypocretins/orexins act through two receptor subtypes: OX(1) and OX(2). Outside the brain, orexin receptors are expressed in adrenal glands, where orexins stimulate the release of glucocorticoids. To further address the regulation of steroidogenesis, we analyzed the effect of orexins on the expression of steroidogenic enzymes in human adrenocortical National Cancer Institute (NCI) H295R cells by qPCR. In NCI H295R cells, OX(2) receptors were highly expressed, as they were in human adrenal glands. After treatment of NCI H295R cells with orexin A for 12-24 h, the cortisol synthesis rate was significantly increased, whereas 30 min of treatment showed no effect. While CYP11B1 and CYP11B2 mRNA levels were increased already at earlier time points, the expression of HSD3B2 and CYP21 mRNA was significantly up-regulated after treatment with orexin A for 12 h. Likewise, orexin B increased CYP21 and HSD3B2 mRNA levels showing, however, a lower potency compared with orexin A. The mRNA levels of CYP11A and CYP17 were unaffected by orexin A. OX(2) receptor mRNA levels were down-regulated after 12 and 24 h of orexin A treatment. Orexin A increased intracellular Ca(2+) but not cAMP concentrations in NCI H295R cells. Furthermore, inhibition of PKC and MAPK kinase/ERK kinase (MEK1/2) prevented the increase of HSD3B2 expression by orexin A. Accordingly, orexin A treatment of NCI H295R cells markedly enhanced ERK1/2 phosphorylation that was prevented by PKC and, in part, PKA inhibition. In conclusion, orexins may influence adrenal steroidogenesis by differential regulation of the expression of steroidogenic enzymes involving Ca(2+), as well as PKC-ERK1/2 signaling.


Assuntos
Córtex Suprarrenal/efeitos dos fármacos , Córtex Suprarrenal/enzimologia , Peptídeos e Proteínas de Sinalização Intracelular/farmacologia , Neuropeptídeos/farmacologia , Neurotransmissores/farmacologia , Esteroide Hidroxilases/metabolismo , Esteroides/metabolismo , Córtex Suprarrenal/citologia , Cálcio/metabolismo , Linhagem Celular , Células Cultivadas , AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Humanos , Hidrocortisona/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Receptores de Orexina , Orexinas , Progesterona Redutase/metabolismo , Proteína Quinase C/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropeptídeos/metabolismo
20.
J Endocrinol ; 200(1): 45-52, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18835979

RESUMO

Modification of low-density lipoprotein (LDL) and abnormal aldosterone and cortisol metabolism have been implicated in the pathogenesis of type 2 diabetes (DM2) and diabetic vascular disease. Since LDL serves as a major cholesterol source for adrenal steroidogenesis, we investigated whether LDL modification in prediabetic and diabetic subjects influences adrenocortical aldosterone and cortisol release. LDL was isolated from 30 subjects with normal glucose tolerance (NGT-LDL), 30 subjects with impaired glucose tolerance (IGT-LDL), and 26 patients with DM2 (DM2-LDL). Oxidation and glycoxidation characteristics of LDL apolipoprotein B100 of each individual was assessed by gas chromatography-mass spectrometry analysis. Human adrenocortical cells (NCI-H295R) were incubated for 24 h with 100 microg/ml LDL and after removal of supernatants stimulated for a further 24 h with angiotensin II (AngII). In supernatants, aldosterone and cortisol secretion was measured. IGT-LDL and DM2-LDL were substantially more modified than NGT-LDL. Each of the five measured oxidation/glycoxidation markers was significantly positively associated with glycemic control, measured as HbA(1c). LDL from all subjects stimulated both the basal and AngII-induced aldosterone and cortisol release from adrenocortical cells. However, hormone secretion was significantly inversely related to the degree of LDL oxidation/glycoxidation. We conclude that LDL modifications in IGT and DM2 subjects may have significant clinical benefits by counteracting prediabetic and diabetic overactivity of the renin-angiotensin-aldosterone system and enhanced cortisol generation.


Assuntos
Córtex Suprarrenal/metabolismo , Aldosterona/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Hidrocortisona/metabolismo , Lipoproteínas LDL/metabolismo , Estado Pré-Diabético/metabolismo , Adulto , Linhagem Celular , Diabetes Mellitus Tipo 2/sangue , Feminino , Teste de Tolerância a Glucose , Glicosilação , Humanos , Lipoproteínas LDL/sangue , Masculino , Oxirredução , Estado Pré-Diabético/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA