Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
1.
J Dairy Sci ; 102(8): 6820-6829, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31178183

RESUMO

We investigated the effect of different phosphate salts on the structural integrity of micellar casein (MC) at pH 7.0. With the increase of salt concentration, a reduction in turbidity was observed for the MC solutions, and it was modeled using an exponential decay function. The inflection point of the model was defined as the first critical salt concentration (C*), and it is suggested that the salt concentration initiates the disintegration of MC. For linear polyphosphates, C* decreased with the number of phosphate groups. Apparent viscosity (ηapp) of MC solutions increased with the increase of salt concentration, and they recorded a peak while the turbidity decreased to a minimum. The salt concentration that resulted in the highest ηapp was identified as the second critical salt concentration (C**). It is hypothesized that the interactions among protein species present in the mixtures are at an optimum state at C**. Both C* and C** were found to be dependent on the MC concentration. The work presented herein supports an understanding of the concentration effect of phosphate salts on MC for structuring dairy products.


Assuntos
Caseínas/química , Micelas , Fosfatos/análise , Animais , Elasticidade , Concentração de Íons de Hidrogênio , Nefelometria e Turbidimetria , Sais/química , Soluções/química , Viscosidade
2.
Mol Cell Endocrinol ; 382(2): 860-70, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24211300

RESUMO

Somatostatin (SST) and SST receptors (SS1R, SS2R, SS3R, SS4R and SS5R) appear to play a significant role in the progression of human prostate cancer (PCa), which is associated with heterogeneity of SSRs expression and specific cell localization as we already demonstrated in the LNCaP cell line, an in vitro model of human androgen-dependent PCa. In this study, PC-3 and DU-145 human castration-resistant PCa cells were found to express all SSRs, while LNCaP expressed all but SS4R. A 48-h treatment with BIM-23244 (SS2R/SS5R) or BIM-23926 (SS1R) SST analogs was more effective in inhibiting cell proliferation, compared to BIM-23120 (SS2R), BIM-23206 (SS5R) and BIM-23704 (SS1R/SS2R). BIM-23926 (SS1R) treatment increased the amount of p21 and decreased phosphorylated (p) ERK1/2. BIM-23244 (SS2R/SS5R) led to p21 increment only in PC-3 cells, and to pERK1/2 reduction in both cell lines. SS1R/SS2R and SS2R/SS5R receptor dimers were natively present on cell membrane and their amount was increased by BIM-23704 (SS1R/SS2R) or BIM-23244 (SS2R/SS5R) treatment, respectively. SS1R, SS2R and SS5R were differently distributed among nuclear, lysosomal and microsomal compartment, according to their different recycling dynamics. These results show that, in PC-3, DU-145 and LNCaP cells, activation of SS1R and SS2R/SS5R leads to relevant antiproliferative effects.


Assuntos
Proliferação de Células/efeitos dos fármacos , Receptores de Somatostatina/metabolismo , Somatostatina , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Masculino , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Estadiamento de Neoplasias , Especificidade de Órgãos , Próstata/efeitos dos fármacos , Próstata/metabolismo , Próstata/patologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Receptores de Somatostatina/química , Receptores de Somatostatina/genética , Transdução de Sinais , Somatostatina/análogos & derivados , Somatostatina/farmacologia , Quinases Ativadas por p21/genética , Quinases Ativadas por p21/metabolismo
3.
J Neuroendocrinol ; 24(3): 453-63, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22129035

RESUMO

Somatostatin and cortistatin have been shown to act directly on pituitary somatotrophs to inhibit growth hormone (GH) release. However, previous results from nonprimate species indicate that these peptides can also directly stimulate GH secretion, at low concentrations. The relevance of this phenomenon in a nonhuman primate model was investigated in the present study by testing the impact of somatostatin/cortistatin on GH release in primary pituitary cell cultures from baboons. High doses (> 10(-10) m) of somatostatin/cortistatin did not alter basal GH secretion but blocked GH-releasing hormone (GHRH)- and ghrelin-induced GH release. However, at low concentrations (10(-17)-10(-13) m), somatostatin/cortistatin dramatically stimulated GH release to levels comparable to those evoked by GHRH or ghrelin. Use of somatostatin receptor (sst) specific agonists/antagonists, and signal transduction blockers indicated that sst2 and sst1 activation via intact adenylate cylcase and mitogen-activated protein kinase systems mediated the inhibitory actions of high-concentration somatostatin. By contrast, the stimulatory actions of low-dose somatostatin on GH release were mediated by sst5 signalling through adenylate cylcase/cAMP/protein kinase A and intracellular Ca(2+) pathways, and were additive with ghrelin (not GHRH). Notably, low-concentrations of somatostatin, similar to sst5-agonists, inhibited prolactin release. These results clearly demonstrate that the ultimate impact of somatostatin/cortistatin on hormone release is dose-dependent, cell type-selective and receptor-specific, where the stimulatory effects of low-concentration somatostatin/cortistatin on GH release extend to primates, thereby supporting the notion that this action is relevant in regulating GH secretion in humans.


Assuntos
AMP Cíclico/fisiologia , Hormônio do Crescimento/metabolismo , Hipófise/efeitos dos fármacos , Receptores de Somatostatina/fisiologia , Somatostatina/farmacologia , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Feminino , Papio , Hipófise/citologia , Hipófise/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
4.
Horm Metab Res ; 43(12): 854-7, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21989554

RESUMO

A combination of basic research observations concerning the interaction of somatostatin (SST) and dopamine (DA) receptors, and clinical reports of enhanced efficacy of combined SST and DA analogue treatment in suppressing GH hypersecretion, lead to the concept of creating chimeric molecules combining structural features of both compound classes. The resulting SST/DA chimeras retain the ability to interact with receptors of both families and display greatly enhanced potency and efficacy, as compared with that of individual SST or DA receptor agonists. In vitro studies with pituitary adenoma cells from acromegalic patients have demonstrated that the chimeric molecules have exceptional activity with regard to suppression of GH and prolactin secretion. Similarly, potent suppression of ACTH secretion from Cushing's-causing corticotroph tumors, and suppression of nonfunctioning pituitary adenoma proliferation has been observed. The chimeric SST/DA compounds are also quite potent and efficacious in suppressing both GH and IGF1 in vivo when tested in nonhuman primates, with no effect on either insulin secretion or glycemic control. Initial clinical studies examining acute, subcutaneous administration of the chimeric SST/DA compound, BIM-23A760, revealed both prolonged circulating half-life and extended duration of biological effect. With chronic administration, however, BIM-23A760 was found to produce a metabolite with dopaminergic activity that gradually accumulates and interferes with the activity of the parent compound. Consequently, efforts are currently underway to produce a second-generation chimera for treatment of neuroendocrine disease.


Assuntos
Antineoplásicos/uso terapêutico , Dopamina/uso terapêutico , Tumores Neuroendócrinos/tratamento farmacológico , Proteínas Recombinantes/uso terapêutico , Somatostatina/uso terapêutico , Antineoplásicos/química , Dopamina/química , Humanos , Proteínas Recombinantes/química , Somatostatina/química
5.
J Endocrinol ; 207(3): 309-17, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20876239

RESUMO

Somatostatin analogues inhibit in vitro cell proliferation via specific membrane receptors (SSTRs). Recent studies on transfected cell lines have shown a ligand-induced formation of receptor dimers. The aim of this study is 1) to evaluate the role of specific ligands in modulating receptor interactions in the androgen-dependent prostate cancer cell line, LNCaP, and in the non-small cell lung cancer line, Calu-6, by co-immunoprecipitation and immunoblot; and 2) to correlate the antiproliferative effect of these compounds with their ability in modulating receptor interactions. In LNCaP, we have demonstrated the constitutive presence of sstr1/sstr2, sstr2/sstr5, sstr5/dopamine (DA) type 2 receptor (D2R), and sstr2/D2R dimers. BIM-23704 (sstr1- and sstr2-preferential compound) increased the co-immunoprecipitation of sstr1/sstr2 and significantly inhibited proliferation (-30.98%). BIM-23244 (sstr2-sstr5 selective agonist) significantly increased the co-immunoprecipitation of sstr2/sstr5, and induced a -41.36% inhibition of proliferation. BIM-23A760, a new somatostatin/DA chimeric agonist with a high affinity for sstr2 and D2R and a moderate affinity for sstr5, significantly increased the sstr5/D2R and sstr2/D2R complexes and was the most powerful in inhibiting proliferation (-42.30%). The chimeric compound was also the most efficient in modulating receptor interaction in Calu-6, increasing the co-immunoprecipitation of D2R/sstr5 and inhibiting cell proliferation (-30.54%). However, behind BIM-23A760, BIM-53097 (D2R-preferential compound) also significantly inhibited Calu-6 proliferation (-17.71%), suggesting a key role for D2R in receptor cross talk and in controlling cell growth. Indeed, activation of monomeric receptors did not affect receptor co-immunoprecipitation, whereas cell proliferation was significantly inhibited when the receptors were synergistically activated. In conclusion, our data show a dynamic ligand-induced somatostatin and DA receptor interaction, which may be crucial for the antiproliferative effects of the new analogues.


Assuntos
Neoplasias Pulmonares/metabolismo , Neoplasias da Próstata/metabolismo , Receptores de Dopamina D2/metabolismo , Receptores de Somatostatina/metabolismo , Somatostatina/análogos & derivados , Somatostatina/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Dopamina/administração & dosagem , Dopamina/análogos & derivados , Humanos , Imunoprecipitação , Masculino , Receptores de Dopamina D2/análise , Receptores de Somatostatina/administração & dosagem , Receptores de Somatostatina/análise , Somatostatina/administração & dosagem
6.
Front Neuroendocrinol ; 31(1): 44-60, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19896496

RESUMO

Ghrelin, a peptide hormone predominantly produced by the stomach, was isolated as the endogenous ligand for the growth hormone secretagogue receptor. Ghrelin is a potent stimulator of growth hormone (GH) secretion and is the only circulatory hormone known to potently enhance feeding and weight gain and to regulate energy homeostasis following central and systemic administration. Therapeutic intervention with ghrelin in catabolic situations may induce a combination of enhanced food intake, increased gastric emptying and nutrient storage, coupled with an increase in GH thereby linking nutrient partitioning with growth and repair processes. These qualities have fostered the idea that ghrelin-based compounds may have therapeutic utility in treating malnutrition and wasting induced by various sub-acute and chronic disorders. Conversely, compounds that inhibit ghrelin action may be useful for the prevention or treatment of metabolic syndrome components such as obesity, impaired lipid metabolism or insulin resistance. In recent years, the effects of ghrelin on glucose homeostasis, memory function and gastrointestinal motility have attracted considerable amount of attention and revealed novel therapeutic targets in treating a wide range of pathologic conditions. Furthermore, discovery of ghrelin O-acyltransferase has also opened new research opportunities that could lead to major understanding of ghrelin physiology. This review summarizes the current knowledge on ghrelin synthesis, secretion, mechanism of action and biological functions with an additional focus on potential for ghrelin-based pharmacotherapies.


Assuntos
Peso Corporal/fisiologia , Metabolismo Energético/fisiologia , Grelina/fisiologia , Homeostase/fisiologia , Proteína Relacionada com Agouti , Sequência de Aminoácidos , Animais , Glicemia/metabolismo , Barreira Hematoencefálica , Caquexia , Ingestão de Alimentos/fisiologia , Motilidade Gastrointestinal , Grelina/química , Hormônio do Crescimento Humano/metabolismo , Humanos , Resistência à Insulina , Dados de Sequência Molecular , Neuropeptídeo Y , Obesidade , Receptores de Grelina , Aumento de Peso
8.
Mol Cell Endocrinol ; 286(1-2): 214-8, 2008 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-18276067

RESUMO

Clinically "non-functioning" human pituitary adenomas (NFPA) constitute about 35% of pituitary adenomas. Somatostatin receptors (SSTR) expression in these adenomas has previously been described both in vitro and in vivo, without evidence for a correlation with tumor volume or the therapeutic efficacy of somatostatin analogs. This study was performed on 13 surgically removed pituitary macroadenomas, diagnosed before surgery as "non-functioning". In addition, 3 growth hormone (GH)-secreting adenomas served as controls. A specimen from each tumor was dispersed and digested to isolate and culture the tumor cells, and the in vitro effects of SSTR2 and SSTR5 selective analogs and Cortistatin (CST) (100nM) on cell viability were studied. The quantity of viable cells was estimated using the XTT method. RNA purification of tumor samples and subsequent RT-PCR studies for SSTR2 and SSTR5 expression were performed. Somatostatin analog with high affinity for SSTR2 reduced cell viability by 20-80% in 8 of 13 NFPAs studied, all expressing the SSTR2. The inhibitory effect on cell viability of SSTR5-selective analog was 15-80% in 10 of 13 NFPAs studied, all but three expressing the SSTR5. CST, however, effectively reduced cell viability in only 6 NFPAs. Cell viability was inhibited by all peptides studied in 2 out of 3 GH-secreting adenomas, expressing both receptors. The third adenoma responded to SSTR2 analog and expressed only SSTR2. These results suggest the involvement of SSTR2 and SSTR5 in the anti-proliferative effects of somatostatin; however, CST is less potent in reducing cell viability in these tumors.


Assuntos
Adenoma/patologia , Antineoplásicos Hormonais/farmacologia , Neoplasias Hipofisárias/patologia , Somatostatina/análogos & derivados , Somatostatina/farmacologia , Adenoma/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Sobrevivência Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Adenoma Hipofisário Secretor de Hormônio do Crescimento/metabolismo , Adenoma Hipofisário Secretor de Hormônio do Crescimento/patologia , Hormônio do Crescimento Humano/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Neuropeptídeos/farmacologia , Peptídeos Cíclicos/farmacologia , Neoplasias Hipofisárias/metabolismo , Células Tumorais Cultivadas
9.
Eur J Clin Invest ; 37(9): 700-8, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17696959

RESUMO

BACKGROUND: Endothelial cells of human blood vessels (arteries and veins) show high levels of somatostatin subtype-1 receptor (sst(1)). The aim of the present study is to investigate the inhibitory effects of novel somatostatin analogs, highly selective for human sst(1), on in vitro angiogenesis and their modulation of vascular endothelial growth factor (VEGF) and vascular endothelial growth factor receptor-2 (VEGFR-2) expression. MATERIALS AND METHODS: Somatostatin analogs BIM-23745 and BIM-23926 were tested for their ability to prevent proliferation and migration of human endothelial HMEC-1 cells, to modulate VEGF and VEGFR-2 expression and to inhibit sprouting of microvessels from cultured human placental vessel explants in fibrin matrix for 28 days. RESULTS: The somatostatin sst(1 )receptor-selective agonists, BIM-23745 and BIM-23926 showed a suppression of endothelial proliferation (e.g. 10(-6) M BIM-23475, 40.0 +/- 2.1% vs. 100% of controls; 10(-7) M BIM-23926, 55.3 +/- 3.3% vs. 100% of controls), migration (e.g. 10(-7) M BIM-23475, 35.0 +/- 1.56% vs. 100% of controls; 10(-7) M BIM-23926, 53.7 +/- 1.77% vs. 100% of controls) and microvessel sprouting (e.g. 10(-8) M BIM-23475, 42.8 +/- 5.6% vs. 100% of controls; 10(-7) M BIM-23926, 17.2 +/- 11.8% vs. 100% of controls). A small but significant percentage of cells exposed to BIM-23745 and BIM-23926 for 24 h and for 72 h presented typical apoptotic morphology. Moreover, both the analogs significantly inhibit VEGF and VEGFR-2 gene expression in endothelial cells grown for 144 h in a fibrin matrix and the VEGF secretion in conditioned media. CONCLUSIONS: The inhibition of endothelial activities suggests potential therapeutic utility for administration of somatostatin sst(1 )receptor-selective agonists in the proliferative diseases involving angiogenesis.


Assuntos
Receptores de Fatores de Crescimento do Endotélio Vascular/agonistas , Somatostatina/análogos & derivados , Inibidores da Angiogênese/metabolismo , Expressão Gênica , Humanos , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Somatostatina/agonistas
10.
Regul Pept ; 143(1-3): 109-17, 2007 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-17531331

RESUMO

Gastrin regulates ECL cell histamine release and is a critical determinant of acid secretion. ECL cell secretion and proliferation is inhibited by gastrin antagonists and somatostatin but little is known about the role of dopamine agonists in this process. Since the ECL cell exhibits all three classes of receptor we evaluated and compared the effects of the gastrin receptor antagonist, (YF476), lanreotide (SST agonist) and novel dopaminergic agents (BIM53061 and BIM27A760) on ECL cell histamine secretion and proliferation. Highly enriched (>98%) ECL cell preparations prepared from rat gastric mucosa using a FACS approach were studied. Real-time PCR confirmed presence of the CCK2, SS2 and SS5 and D1 receptors on ECL cells. YF476 inhibited histamine secretion and proliferation with IC(50)s of 1.25 nM and 1.3 x 10(-11) M respectively, values 10-1000x more potent than L365,260. Lanreotide inhibited secretion and proliferation (2.2 nM, 1.9 x 10(-10) M) and increased YF476-inhibited proliferation a further 5-fold. The dopamine agonist, BIM53061, inhibited gastrin-mediated ECL cell secretion and proliferation (17 nM, 6 x 10(-10) M) as did the novel dopamine/somatostatin chimera BIM23A760 (22 nM, 4.9 x 10(-10) M). Our studies demonstrate that the gastrin receptor antagonist, YF476, is the most potent inhibitor of ECL cell histamine secretion and proliferation. Lanreotide, a dopamine agonist and a dopamine/somatostatin chimera inhibited ECL cell function but were 10-1000x less potent than YF476. Agents that selectively target the CCK2 receptor may provide alternative therapeutic strategies for gastrin-mediated gastrointestinal cell secretion and proliferation such as evident in the hypergastrinemic gastric carcinoids associated with low acid states.


Assuntos
Celulas Tipo Enterocromafim/metabolismo , Receptor de Colecistocinina B/metabolismo , Receptores Dopaminérgicos/metabolismo , Receptores de Somatostatina/metabolismo , Animais , Benzodiazepinonas/farmacologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Agonistas de Dopamina/farmacologia , Relação Dose-Resposta a Droga , Celulas Tipo Enterocromafim/citologia , Celulas Tipo Enterocromafim/efeitos dos fármacos , Gastrinas/farmacologia , Expressão Gênica/efeitos dos fármacos , Liberação de Histamina/efeitos dos fármacos , Imuno-Histoquímica , Masculino , Peptídeos Cíclicos/farmacologia , Compostos de Fenilureia/farmacologia , Ratos , Ratos Sprague-Dawley , Receptor de Colecistocinina B/antagonistas & inibidores , Receptor de Colecistocinina B/genética , Receptores Dopaminérgicos/genética , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/genética , Somatostatina/análogos & derivados , Somatostatina/farmacologia
11.
J Physiol Pharmacol ; 58(1): 179-88, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17440235

RESUMO

The aim of the study was to examine the effect of somatostatin (SST) and its analogs on the release of chromogranin A (CgA) and alpha-subunit (alpha-SU) from clinically non-functioning pituitary adenomas incubated in vitro. Seven pituitary macroadenomas surgically removed were investigated. All of the tumors were diagnosed before surgery as non-functioning, but they expressed either gonadotropins or their subunits as detected by immunohistochemistry. Two tumors additionally expressed prolactin and growth hormone. All adenomas also expressed chromogranin A (CgA) and at least 3 of 5 subtypes of somatostatin receptors. The cells isolated from the examined tumors were exposed in vitro to either native SST-14 or the following receptor-specific SST analogs: BIM-23926 (agonist of sst1 receptor), BIM-23120 (agonist of sst2 receptor), BIM-23206 (agonist of sst5 receptor) and BIM23A387 (somatostatin/dopamine chimera). The concentration of CgA was measured by means of ELISA method and of alpha-SU was measured by an immunoradiometric method. It was found that the exposure on SST-14 resulted in the decrease of CgA and alpha-SU release from tumor cells in majority of samples, and the effect on CgA was positively correlated with the expression of sst3 and also with the sst2A/sst2B expressions ratio. The inhibitory effect of SST-14 on CgA and alpha-SU seems also to correlate negatively with the expression of sst2B. CgA inhibition also correlates positively with sst5 expression. Among the other compounds studied, only the sst2 agonist decreased the release in all the investigated samples. The remaining substances (agonists of sst1 and sst5 and SST/DA chimera) produced the divergent changes (increased or decreased release, depending on the sample). The data suggest that the inhibition of CgA (and possibly of alpha-SU) release by SST is mediated via subtypes sst2A, sst3 and sst5, whereas sst2B subtype may induce the opposite effect.


Assuntos
Adenoma/metabolismo , Antineoplásicos Hormonais/farmacologia , Biomarcadores Tumorais/metabolismo , Cromogranina A/metabolismo , Subunidade alfa de Hormônios Glicoproteicos/metabolismo , Neoplasias Hipofisárias/metabolismo , Receptores de Somatostatina/agonistas , Somatostatina/farmacologia , Adenoma/patologia , Adenoma/fisiopatologia , Adulto , Idoso , Biomarcadores Tumorais/análise , Ensaio de Imunoadsorção Enzimática , Feminino , Hormônio Foliculoestimulante/análise , Hormônio do Crescimento Humano/análise , Humanos , Imuno-Histoquímica , Ensaio Imunorradiométrico , Hormônio Luteinizante Subunidade beta/análise , Masculino , Pessoa de Meia-Idade , Neoplasias Hipofisárias/patologia , Neoplasias Hipofisárias/fisiopatologia , Prolactina/análise , Receptores de Somatostatina/metabolismo , Somatostatina/análogos & derivados , Células Tumorais Cultivadas
12.
Cell Death Differ ; 14(2): 197-208, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16645635

RESUMO

Somatostatin is a multifunctional hormone that modulates cell proliferation, differentiation and apoptosis. Mechanisms for somatostatin-induced apoptosis are at present mostly unsolved. Therefore, we investigated whether somatostatin receptor subtype 2 (sst2) induces apoptosis in the nontransformed murine fibroblastic NIH3T3 cells. Somatostatin receptor subtype 2 expression induced an executioner caspase-mediated apoptosis through a tyrosine phosphatase SHP-1 (Src homology domain phosphatase-1)-dependent stimulation of nuclear factor kappa B (NF-kappaB) activity and subsequent inhibition of the mitogen-activated protein kinase JNK. Tumor necrosis factor alpha (TNFalpha) stimulated both NF-kappaB and c-Jun NH2-terminal kinase (JNK) activities, which had opposite action on cell survival. Importantly, sst2 sensitized NIH3T3 cells to TNFalpha-induced apoptosis by (1) upregulating TNFalpha receptor protein expression, and sensitizing to TNFalpha-induced caspase-8 activation; (2) enhancing TNFalpha-mediated activation of NF-kappaB, resulting in JNK inhibition and subsequent executioner caspase activation and cell death. We have here unraveled a novel signaling mechanism for a G protein-coupled receptor, which directly triggers apoptosis and crosstalks with a death receptor to enhance death ligand-induced apoptosis.


Assuntos
Apoptose , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Receptores de Somatostatina/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Somatostatina/metabolismo , Animais , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Ativação Enzimática/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Camundongos , Modelos Biológicos , Mimetismo Molecular/efeitos dos fármacos , Células NIH 3T3 , Fosforilação/efeitos dos fármacos , Proteína Fosfatase 1 , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Transdução de Sinais/efeitos dos fármacos , Transfecção , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/farmacologia
13.
Life Sci ; 78(7): 689-93, 2006 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-16115652

RESUMO

The aim of the work was to investigate the effects of somatostatin analogs acting selectively on sst1 (BIM-23926), sst2 (BIM-23120) and sst5 (BIM-23206) receptor subtypes on the viability of "clinically non-functioning" pituitary adenomas in vitro. The effects of native SST (SST-14), a SST/DA chimera (BIM-23A387) and a D(2)-dopamine receptor agonist bromocriptine (BC) were also examined. The study was performed on 10 surgically removed pituitary macroadenomas, diagnosed before surgery as "non-functioning". A part of each tumor was mechanically dispersed and digested with collagenase to isolate the tumoral cells. Another part of each tumor was fixed, embedded in paraffin and immunostained to reveal the pituitary hormones and SST receptor subtypes (sst1, sst2A, sst2B, sst3, sst4, sst5). The tumoral cell suspensions were incubated for 24 h with the substances mentioned above. The quantity of viable cells was estimated using the EZ4U system. The results were compared with the immunohistochemical evaluation of the hormonal profile of adenoma and the sst receptor subtype immunoreactivities present. The findings indicate that selective sst1, sst2 and sst5 receptors agonists, SST/DA chimera and D(2)-dopamine receptor agonist bromocriptine affect the viability of some, but not all, "clinically non-functioning" pituitary adenomas in vitro. The most effective was bromocriptine. The investigated somatostatin analogs including SST/DA chimera exerted roughly similar inhibitory effects. Further studies are needed to fully evaluate the potential usefulness of these compounds in the pharmacological treatment of "non-functioning" pituitary tumors.


Assuntos
Adenoma/tratamento farmacológico , Bromocriptina/farmacologia , Agonistas de Dopamina/farmacologia , Neoplasias Hipofisárias/tratamento farmacológico , Receptores de Somatostatina/agonistas , Proteínas Recombinantes de Fusão/farmacologia , Adenoma/metabolismo , Adenoma/patologia , Adulto , Idoso , Biomarcadores Tumorais/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sinergismo Farmacológico , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Neoplasias Hipofisárias/metabolismo , Neoplasias Hipofisárias/patologia , Receptores Dopaminérgicos/metabolismo , Células Tumorais Cultivadas/efeitos dos fármacos
14.
Obes Rev ; 6(4): 307-22, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16246216

RESUMO

The neuropeptide Y (NPY)/peptide YY (PYY) system has been implicated in the physiology of obesity for several decades. More recently ignited enormous interest in PYY3-36, an endogenous Y2-receptor agonist, as a promising anti-obesity compound. Despite this interest, there have been remarkably few subsequent reports reproducing or extending the initial findings, while at the same time studies finding no anti-obesity effects have surfaced. Out of 41 different rodent studies conducted (in 16 independent labs worldwide), 33 (83%) were unable to reproduce the reported effects and obtained no change or sometimes increased food intake, despite use of the same experimental conditions (i.e. adaptation protocols, routes of drug administration and doses, rodent strains, diets, drug vendors, light cycles, room temperatures). Among studies by authors in the original study, procedural caveats are reported under which positive effects may be obtained. Currently, data speak against a sustained decrease in food intake, body fat, or body weight gain following PYY3-36 administration and make the previously suggested role of the hypothalamic melanocortin system unlikely as is the existence of PYY deficiency in human obesity. We review the studies that are in the public domain which support or challenge PYY3-36 as a potential anti-obesity target.


Assuntos
Fármacos Antiobesidade/farmacologia , Peso Corporal/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Peptídeo YY/farmacologia , Animais , Comportamento Animal , Interpretação Estatística de Dados , Dipeptidil Peptidase 4/metabolismo , Humanos , Fragmentos de Peptídeos , Peptídeo YY/administração & dosagem , Receptores de Neuropeptídeo Y/agonistas , Resposta de Saciedade/efeitos dos fármacos , Especificidade da Espécie , Estresse Fisiológico/fisiopatologia
15.
J Mol Endocrinol ; 35(2): 333-41, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16216913

RESUMO

Dopamine (DA) and somatostatin (SRIF) receptor agonists inhibit growth hormone (GH) secretion by pituitary adenomas. We investigated DA subtype 2 receptor (DR2) and SRIF receptor (sst) subtypes 2 and 5 expression in 25 GH-secreting pituitary adenomas and tested in primary culture the effects on GH and prolactin (PRL) secretion of sst agonists selectively interacting with sst2 (BIM-23120), sst5 (BIM-23206), and sst2 and sst5 (BIM-23244). All adenomas expressed sst2; eight adenomas expressed both sst5 and DR2, eight sst5 but not DR2, and eight DR2 but not sst5. One tissue lacked expression of DR2 and sst5. GH secretion was inhibited by BIM-23120 in all samples, while it was reduced by BIM-23206 only in adenomas not expressing DR2. BIM-23120's inhibitory effects correlated with sst2 and DR2 expression, whereas DR2 expression correlated inversely with BIM-23206 inhibitory effects on GH secretion. In seven mixed GH-/PRL-secreting pituitary adenomas, PRL secretion was inhibited in sst5-expressing tumors by BIM-23206, but not by BIM-23120. BIM-23244 reduced PRL secretion only in adenomas expressing sst2, sst5 and DR2. sst5 and DR2 expression correlated directly with BIM23206 inhibitory effects on PRL secretion. Our results suggest that adenomas expressing DR2 are less likely to respond to clinically available SRIF analogs in terms of GH secretion inhibition. Therefore, drugs interacting also with DR2 might better control secretion of pituitary adenomas.


Assuntos
Adenoma Hipofisário Secretor de Hormônio do Crescimento/metabolismo , Isoformas de Proteínas/metabolismo , Receptores Dopaminérgicos/metabolismo , Receptores de Somatostatina/metabolismo , Somatostatina/análogos & derivados , Acromegalia/metabolismo , Adulto , Idoso de 80 Anos ou mais , Agonistas de Dopamina , Feminino , Adenoma Hipofisário Secretor de Hormônio do Crescimento/patologia , Hormônio do Crescimento Humano/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Prolactina/metabolismo , Isoformas de Proteínas/genética , RNA Mensageiro/metabolismo , Receptores Dopaminérgicos/genética , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/genética , Somatostatina/metabolismo
16.
Eur J Endocrinol ; 153(1): 135-41, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15994755

RESUMO

OBJECTIVE: This study compared the potency of a somatostatin receptor (sstr)2-sstr5 analog, BIM-23244, of an sstr2-dopamine D2 receptor (sstr2-DAD2) molecule, BIM-23A387 and of new somatostatin-dopamine chimeric molecules with differing, enhanced affinities for sstr2, sstr5 and DAD2, BIM-23A758, BIM-23A760 and BIM-23A761, to suppress GH and prolactin (PRL) from 18 human GH adenomas that are partially responsive to octreotide or lanreotide. MATERIALS AND METHODS: The sstr2, sstr5 and DAD2 mRNA levels were determined by RT-PCR. The effect of drugs was tested in cell cultures at various concentrations. RESULTS: In all tumors, the sstr2, sstr5 and DAD2 mRNA levels were coexpressed (mean levels+/-s.e.m. 0.4+/-0.1, 5.3+/-1.9 and 2.0+/-0.4 copy/copy beta-glucuronidase). In 13 tumors, the maximal suppression of GH secretion produced by BIM-23A387 (30+/-3%) and BIM-23244 (28+/-3%) was greater than that produced by octreotide (23+/-3%). In six out of 13 tumors, BIM-23A758, BIM-23A760 and BIM- 23A761 produced greater maximal suppression of GH secretion than octreotide (33+/-5, 38+/-2 and 41+/-2 vs 24+/-2%). Their EC(50) values were 10, 2 and 4 pmol/l. BIM-23A761 was more effective than BIM-23A387 in GH suppression (41+/-2 vs 32+/-4%). The new chimeric molecules produced maximal PRL suppression greater than octreotide (62+/-8 to 74+/-5 vs 46+/-11%). CONCLUSIONS: Novel dopamine-somatostatin chimeric molecules with differing, enhanced activity at sstr2, sstr5 and DAD2, consistently produced significatly greater suppression of GH and PRL than either octreotide or single-receptor-interacting ligands in tumors from patients classified as only partially responsive to octreotide therapy. The higher efficacy of the chimeric compounds was, at least partially, linked to their high affinity for sstr2 (IC50 1-10 pmol/l). The other mechanisms by which such molecules produce an enhanced inhibition of GH remain to be elucidated.


Assuntos
Dopamina/análogos & derivados , Neoplasias Hipofisárias/tratamento farmacológico , Prolactinoma/tratamento farmacológico , Receptores de Dopamina D2/administração & dosagem , Receptores de Somatostatina/administração & dosagem , Somatostatina/análogos & derivados , Acromegalia/sangue , Acromegalia/tratamento farmacológico , Adulto , Antineoplásicos Hormonais/administração & dosagem , Dopamina/administração & dosagem , Resistencia a Medicamentos Antineoplásicos , Feminino , Hormônio do Crescimento Humano/sangue , Hormônio do Crescimento Humano/metabolismo , Humanos , Masculino , Octreotida/administração & dosagem , Neoplasias Hipofisárias/sangue , Neoplasias Hipofisárias/genética , Prolactina/sangue , Prolactina/metabolismo , Prolactinoma/sangue , Prolactinoma/genética , RNA Mensageiro/análise , Receptores de Dopamina D2/genética , Receptores de Somatostatina/genética , Proteínas Recombinantes de Fusão/administração & dosagem , Somatostatina/administração & dosagem , Células Tumorais Cultivadas
17.
Endocrinology ; 146(9): 3836-42, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15919752

RESUMO

Ghrelin was purified from rat stomach as an endogenous ligand for the GH secretagogue (GHS) receptor. As a GHS, ghrelin stimulates GH release, but it also has additional activities, including stimulation of appetite and weight gain. Plasma GH and ghrelin secretory patterns appear unrelated, whereas many studies have correlated ghrelin variations with food intake episodes. To evaluate the role of endogenous ghrelin, GH secretion and food intake were monitored in male rats infused sc (6 mug/h during 10 h) or intracerebroventricularly (5 microg/h during 48 h) with BIM-28163, a full competitive antagonist of the GHS-R1a receptor. Subcutaneous BIM-28163 infusion significantly decreased GH area under the curve during a 6-h sampling period by 54% and peak amplitude by 46%. Twelve hours after the end of treatment these parameters returned to normal. Central treatment was similarly effective (-37 and -42% for area under the curve and -44 and -49% for peak amplitude on the first and second days of infusion, respectively). Neither peripheral nor central BIM-28163 injection modified GH peak number, GH nadir, or IGF-I levels. In this protocol, food intake is not strongly modified and water intake is unchanged. Subcutaneous infusion of BIM-28163 did not change plasma leptin and insulin levels evaluated at 1200 and 1600 h. On the contrary, central BIM-28163 infusion slightly increased leptin and significantly increased insulin concentrations. Thus, endogenous ghrelin, through GHS-R1a, acts as a strong endogenous amplifier of spontaneous GH peak amplitude. The mechanisms by which ghrelin modifies food intake remain to be defined and may involve a novel GHS receptor.


Assuntos
Hormônio do Crescimento/metabolismo , Hormônios Peptídicos/metabolismo , Hormônios Peptídicos/farmacologia , Receptores Acoplados a Proteínas G/metabolismo , Animais , Células CHO , Cricetinae , Ingestão de Alimentos/efeitos dos fármacos , Grelina , Humanos , Injeções Intraventriculares , Injeções Subcutâneas , Insulina/sangue , Leptina/sangue , Masculino , Hormônios Peptídicos/genética , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores de Grelina
18.
J Endocrinol Invest ; 28(11 Suppl International): 21-7, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16625841

RESUMO

We report the comparative efficacy of a somatostatin receptor 1 and 5 subtypes (SSTR2 and SSTR5), and dopamine D2 (DAD2) compound, BIM-23A760, in suppressing GH secretion, in cell culture from human GH-secreting tumors, from patients partially responsive to long-term treatments with octreotide or lanreotide. In 18 tumors tested, the SSTR2, SSTR5, and DAD2 mRNAs were coexpressed. The SSTR2-selective analog, BIM-23197, the SSTR5-selective analog, BIM-23268, and the dopamine (DA) analog, BIM-53097, produced a mean maximal suppression of GH secretion (24 +/- 3, 20 +/- 3, and 20 +/- 3%, respectively) that was similar to that obtained with octreotide (23 +/- 3%). Nevertheless, based on individual responses, 60% of the tumors were mostly sensitive to the SSTR2 analog while 19 and 21% of the tumors were mainly responsive to the SSTR5 analog and to the DA analog, respectively. Among a series of new chimeric compounds that bind the SSTR2, SSTR5, and DAD2 receptors with variable affinities, BIM-23A760 produced greater maximal suppression of GH secretion than octreotide (38 +/- 2 vs 24 +/- 2%; p<0.03). The EC50 for BIM-23A760 was 2 pmol/l. In the presence of sulpride, the dose response inhibition of GH secretion by the trihybrid molecule, BIM-23A760, was partially reversed. The trihybrid produced also a maximal suppression of PRL greater than octreotide (74 +/- 5 vs 46 +/- 11%). When SSTRs pan inhibitors such as BIM-23A779 (binding affinity for SSTR1, SSTR2, SSTR3, SSTR5, respectively: 2.5, 0.3, 0.6, 0.6 nmol/l) or SOM230 were tested for their suppressive effects on GH secretion, they were less potent than the previous dopastatin hybrid molecule. After a brief exposure to a SSTR2-selective analog, BIM-23197, or to a DA analog, BIM-53097, the maximal GH suppression was achieved during 12 h. Under exposure to BIM-23A760, in the same conditions, maximal suppression of GH secretion lasted for 24 h. Such a longer biological effect, yet not explained, probably participates in the higher efficacy of BIM-23A760. The higher efficacy of BIM-23A760 is, at least partially, linked to its high affinity for the SSTR2 receptor subtype (IC50: 3 pmol/l). As compared to the dopastatin compound, the lower efficacy of the universal somatostatin ligands in the inhibition of GH secretion of GH-secreting tumors argues for the use of drugs targeted, according to specific receptors expression and functionality which may vary among the various classes of tumors.


Assuntos
Hormônio do Crescimento Humano/metabolismo , Octreotida/uso terapêutico , Neoplasias Hipofisárias/tratamento farmacológico , Neoplasias Hipofisárias/metabolismo , Receptores Dopaminérgicos/metabolismo , Receptores de Somatostatina/metabolismo , Acromegalia/tratamento farmacológico , Adulto , Feminino , Expressão Gênica , Hormônio do Crescimento Humano/antagonistas & inibidores , Humanos , Masculino , Oligopeptídeos/farmacologia , Piperazinas/farmacologia , Prolactina/metabolismo , RNA Mensageiro/análise , Receptores de Dopamina D2/genética , Receptores de Somatostatina/genética
19.
Eur J Endocrinol ; 151(6): 787-95, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15588247

RESUMO

OBJECTIVE: Ghrelin, a recently identified 28-amino acid peptide is a potent GH secretagogue (GHS) produced predominantly by the stomach. Ghrelin stimulates GH secretion through binding to the GHS receptor in the hypothalamus and pituitary. In addition to the GH-releasing action, ghrelin has been found to be a powerful orexigenic factor. To assess the direct in vitro effects of ghrelin on human pituitary hormone secretion we have produced a panel of novel ghrelin analogs (molecular weight, 3323-3384; human native ghrelin, 3371) with enhanced affinity for the human GHS receptor (IC(50) 0.38-1.09 nM; human ghrelin, 1.2-2.2 nM). METHODS: The peptidic analogs were tested for their effect on GH secretion using dispersed human fetal pituitaries (21 to 23 weeks of gestation) and cultured GH- and prolactin (PRL)-secreting adenomas. The expression of the GHS receptor in normal (fetal and adult) human pituitary tissues, GH- and PRL-cell adenomas was established using RT-PCR. RESULTS: The effects of ghrelin, its analogs and GH-releasing hormone (GHRH) alone or in combination on GH and PRL secretion were compared at various concentrations. The ghrelin analogs stimulated GH release by 35-60% from human fetal pituitary cells (1-10 nM; P<0.05) and by 50-75% from cultured pituitary adenomas (10 nM; P<0.05). This releasing effect was dose-dependent, achieving maximal stimulation with analog concentrations at 100 nM. Human ghrelin was less potent as compared with its analogs in stimulating human GH, in keeping with the improved binding affinity of the analogs for the GHS-1a receptor. The ghrelin analogs and GHRH had comparable effects on GH secretion from both normal and adenomatous cells, and in combination produced an additive stimulatory effect on GH (150%; P<0.0001). In contrast, ghrelin and its analogs induced a comparable increase in PRL release ranging between 25 and 40% (P<0.05) from fetal cells and 30 and 70% (P<0.001) from cultured PRL-cell and mixed GH-PRL adenomas. CONCLUSIONS: Our results have demonstrated for the first time that ghrelin analogs with enhanced affinity for the GHS receptor are potent stimulators of GH secretion from human pituitary cells, and thus may possess potential clinical therapeutic benefits.


Assuntos
Hormônio do Crescimento Humano/metabolismo , Hormônios Peptídicos/farmacologia , Hipófise/metabolismo , Prolactina/metabolismo , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Adenoma/metabolismo , Células Cultivadas , Grelina , Humanos , Ligantes , Hipófise/citologia , Hipófise/efeitos dos fármacos , Neoplasias Hipofisárias/metabolismo , RNA/biossíntese , RNA/isolamento & purificação , Receptores de Grelina , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
Dig Liver Dis ; 36 Suppl 1: S17-25, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15077907

RESUMO

A major challenge in developing somatostatin-based therapies, as well as an important question of basic physiology, is how to achieve functional specificity with an agent that has widespread actions. The natural somatostatin system achieves functional specificity in part through local somatostatin production at the site of action. Further selectivity may be achieved through the recently elucidated somatostatin receptor subtypes. To explore the relationship between the receptor subtypes and somatostatin-mediated functions, we have tested panels of selective somatostatin analogs in specific biological assays. Our studies have demonstrated complex, functional interactions between the somatostatin receptor subtypes. Specific combinations of somatostatin receptor subtypes in specific tissues may be either synergistic or antagonistic. By altering the expression ratio of the interacting receptors, the biological response to somatostatin can be influenced by environmental, hormonal and physiological status. In addition, inappropriate or unbalanced receptor expression may provide a novel mechanism of disease. These concepts take on an even broader context with the demonstration that somatostatin receptor subtypes also interact with other G-protein-coupled receptors. The insight gained from these studies has already resulted in several novel approaches to acromegaly therapy. Our further understanding of these complex cellular codes will provide the conceptual basis for future therapeutics with greatly enhanced efficacy and specificity.


Assuntos
Somatostatina/análogos & derivados , Carcinoma/fisiopatologia , Hormônio do Crescimento/metabolismo , Receptores Dopaminérgicos/fisiologia , Receptores de Somatostatina/fisiologia , Somatostatina/uso terapêutico , Neoplasias da Glândula Tireoide/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA