Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 159
Filtrar
1.
Science ; 217(4565): 1155-6, 1982 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-6126003

RESUMO

After monolayer cultures of rat islets were exposed to [(125)I]insulin,[(125)I]glucagon, and [(125)I]tyrosinyl somatostatin, specific autoradiographic grains associated with each radioactively labeled ligand were found on B, A, and D cells. The density of labeling of the B, A, and D cells with each labeled ligand correlated well with the known actions of the three hormones on each of the islet cells.


Assuntos
Ilhotas Pancreáticas/metabolismo , Receptor de Insulina/metabolismo , Receptores de Superfície Celular/metabolismo , Animais , Células Cultivadas , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/ultraestrutura , Microscopia Eletrônica , Ratos , Receptores de Glucagon , Receptores de Somatostatina , Somatostatina/metabolismo
2.
Science ; 198(4320): 930-1, 1977 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-337488

RESUMO

Diabetic mice of the C57BL/6J obob and C57BL/Ks dbdb strains show a reduction in pancreatic somatostatin concentration accompanied in the obob strain by a striking decrease in the number of somatostatin-containing cells in the islets. Somatostatin concentration is also decreased in the stomach but increased in the hypothalamus. These findings suggest different control mechanisms for somatostatin in the hypothalamus compared to the gut and pancreas and exclude a primary genetic abnormality of somatostatin cells in the mutants.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Camundongos Obesos/metabolismo , Somatostatina/metabolismo , Animais , Diabetes Mellitus Experimental/genética , Mucosa Gástrica/metabolismo , Hipotálamo/metabolismo , Insulina/sangue , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Pâncreas/metabolismo
3.
Science ; 288(5463): 154-7, 2000 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-10753124

RESUMO

Somatostatin and dopamine are two major neurotransmitter systems that share a number of structural and functional characteristics. Somatostatin receptors and dopamine receptors are colocalized in neuronal subgroups, and somatostatin is involved in modulating dopamine-mediated control of motor activity. However, the molecular basis for such interaction between the two systems is unclear. Here, we show that dopamine receptor D2R and somatostatin receptor SSTR5 interact physically through hetero-oligomerization to create a novel receptor with enhanced functional activity. Our results provide evidence that receptors from different G protein (heterotrimeric guanine nucleotide binding protein)-coupled receptor families interact through oligomerization. Such direct intramembrane association defines a new level of molecular crosstalk between related G protein-coupled receptor subfamilies.


Assuntos
Receptor Cross-Talk , Receptores de Dopamina D2/metabolismo , Receptores de Somatostatina/metabolismo , Animais , Células CHO , Membrana Celular/metabolismo , Córtex Cerebral/metabolismo , Colforsina/farmacologia , Corpo Estriado/metabolismo , Cricetinae , AMP Cíclico/metabolismo , Dimerização , Antagonistas dos Receptores de Dopamina D2 , Guanosina 5'-O-(3-Tiotrifosfato)/farmacologia , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Humanos , Ligantes , Masculino , Neurônios/metabolismo , Células Piramidais/metabolismo , Quimpirol/farmacologia , Ratos , Receptores de Dopamina D2/agonistas , Receptores de Dopamina D2/genética , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/antagonistas & inibidores , Receptores de Somatostatina/genética , Somatostatina/metabolismo , Somatostatina/farmacologia , Espiperona/farmacologia , Sulpirida/farmacologia , Transfecção
4.
J Clin Invest ; 72(6): 2137-43, 1983 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-6139388

RESUMO

Synthesis of somatostatin-14 (S-14) could occur through direct enzymatic processing of precursor somatostatin (prosomatostatin) or via sequential breakdown of prosomatostatin leads to somatostatin-28 (S-28) leads to S-14. If direct processing is important, it should theoretically generate S-14 and a molecule equivalent to prosomatostatin without the S-14 sequence. In an attempt to identify such a molecule, I characterized the molecular forms of S-28(1-12)-like immunoreactivity (S-28(1-12) LI) in the rat pancreas and compared the relative amounts of these forms with those of S-14-like immunoreactivity (S-14 LI). Pancreatic extracts were chromatographed on Sephadex G-50 and Sephadex G-75 columns (Pharmacia Fine Chemicals Inc., Piscataway, NJ) under denaturing conditions and immunoreactivity in the eluting fractions was analyzed by region-specific radioimmunoassays (RIAs). For RIA of S-28(1-12) LI we used a newly developed rabbit antibody R 21 B, 125I-Tyr12 S-28(1-14), and S-28(1-12) standards. This system detects S-28, S-28(1-12), high molecular weight forms of S-28(1-12), but not S-14. S-14 LI was measured using antibody R149, which detects S-14, S-28, and higher molecular weight S-14-like substances, but not S-28(1-12). Three forms of S-28(1-12) LI were identified: Mr 9,000-11,000, Mr 1,200 (corresponding to S-28(1-12), and Mr less than 1,000, comprising, respectively, 35, 53, and 12% of total immunoreactivity. The relative abundance of the 9,000-11,000 mol wt S-28(1-12) LI material was unchanged following removal of S-14 LI from pancreatic extracts by affinity chromatography before gel filtration. Serial dilutions of fractions containing 9-11,000 and 1,200 mol wt materials exhibited parallelism with synthetic S-28(1-12). The total pancreatic concentration of S-28(1-12) LI was 1.56 pmol/mg protein, of which S-28(1-12) accounted for 0.83 pmol/mg protein and 9-11,000 S-28(1-12) LI comprised 0.55 pmol/mg protein. Pancreatic S-14 LI concentration was 2.07 pmol/mg protein, of which 98% corresponded to S-14. S-28-related peaks accounted for <1% of immunoreactivity in both RIAs. I concluded that (a) S-14 is the main form of pancreatic S-14 LI; (b) S-28 is present in very small quantities, in the pancreas; (c) S-28(1-12) LI consist mainly of S-28(1-12) and 9-11,000 mol wt S-28(1-12) LI; (d) 9-11,000 l wt S-28(1-12) LI could represent prosomatostatin without the S-14 sequence; (e) the finding of high concentrations of 9-11,000 mol wt S-28(1-12) LI suggests that S-14 synthesis can occur independently of S-28 and that direct processing of prosomatostatin is an important pathway for S-14 synthesis in the pancreas.


Assuntos
Pâncreas/metabolismo , Somatostatina/biossíntese , Animais , Cromatografia de Afinidade , Cromatografia em Gel , Masculino , Peso Molecular , Radioimunoensaio , Ratos , Ratos Endogâmicos , Somatostatina-28
5.
J Clin Invest ; 80(5): 1455-8, 1987 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-2890658

RESUMO

Quantitative electron microscopic autoradiography was used for comparing the binding of labeled somatostatin-14 (S-14) and somatostatin-28 (S-28 section) to islet cells. Monolayer cultures of rat islet cells were incubated with [125I-Tyr11]S-14 (S-14 section) or [125I-Leu8, D-Trp22, Tyr25]S-28 (S-28 section) in the presence or absence of excess unlabeled peptides. Autoradiographic grains (ARG) associated with individual islet cells were identified and expressed as the mean number per B, A, and D cells. Specific ARG associated with S-14 were found over B and A cells. S-28 section-related specific ARG were concentrated over B, A, as well as D cells. The highest density of S-14 section labeling occurred over A cells, which under conditions of maximum labeling (37 degrees C for 60 min) contained five times as many ARG as did B cells. By contrast, under the same incubation conditions, the labeling density with S-28 section was maximal over B cells, which contained four and five times as many grains as A and D cells, respectively. These observations show preferential association of S-14 section with the A cell and S-28 section with the B cel provide strong evidence for the existence of separate binding sites for S-14 section and S-28 section on A and B cells, respectively, which presumably mediate the previously reported glucagon selective inhibitory effect of S-14 and the insulin-selective action of S-28.


Assuntos
Ilhotas Pancreáticas/metabolismo , Receptores de Neurotransmissores/metabolismo , Somatostatina/metabolismo , Animais , Autorradiografia , Células Cultivadas , Microscopia Eletrônica , Ratos , Receptores de Somatostatina , Somatostatina-28
6.
J Clin Invest ; 84(2): 412-7, 1989 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-2569474

RESUMO

The pathways by which islet B, A, and D cells bind and internalize homologous (self) and heterologous (other) islet hormones were compared. [125I-Tyr]Somatostatin-14 (S-14), 125I-insulin, and 125I-glucagon were incubated with monolayer cultures of neonatal rat islet cells. Tissues were processed for quantitative electron microscopic autoradiography by the probability circle method coupled to morphometry. For all three radioligands and all three cell types surface labeling was rapidly followed by internalization of the radioligands into endocytotic vesicles. The further intracellular movement of the ligand occurred in a time- and temperature-related manner and depended on whether it was homologous or heterologous for the cell in question. Thus [125I-Tyr]S-14 in B and A cells, 125I-insulin in A and D cells, and 125I-glucagon in B and D cells were rapidly transferred from endocytotic vesicles to lysosomal structures. By contrast, [125I-Tyr]S-14 in D cells, 125I-insulin in B cells, and 125I-glucagon in A cells showed poor progression from endocytotic vesicles to downstream vesicular structures. We conclude that (a) each of the three radioligands is internalized by islet cells in a time- and temperature-dependent manner; (b) after initial internalization the further intracellular progression of the endocytosed radioligand occurs freely in cells heterologous for the radioligand but poorly in cells homologous for the radioligand; and (c) binding and endocytosis can be uncoupled from lysosomal degradation of ligand.


Assuntos
Glucagon/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Somatostatina/metabolismo , Animais , Autorradiografia , Células Cultivadas , Endocitose , Ilhotas Pancreáticas/ultraestrutura , Lisossomos/metabolismo , Ratos
7.
J Clin Invest ; 64(3): 737-42, 1979 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-468988

RESUMO

Somatostatin-like immunoreactivity (SLI) is widely distributed in tissues and biological fluids. To determine whether SLI is also present in amniotic fluid, samples obtained by amniocentesis from 30 normal and 27 abnormal pregnancies were studied by radioimmunoassay. Direct incubation of [(125)I-Tyr(1)]tetradecapeptide somatostatin (SRIF) with amniotic fluid resulted in 89% tracer degradation. Damage was reduced to <5% when samples were acidified and boiled before the assay. With this technique, SLI was detectable in all normal amniotic fluid samples; the mean level at 15-20 wk of gestation (320+/-55 pg/ml, n = 15) being 4.5 times higher than the mean at 32-43 wk (70+/-12 pg/ml, n = 15) (P < 0.001). In cases of preeclampsia (n = 6), gestational diabetes (n = 5), anencephaly (n = 1), and meningomyelocele (n = 1), SLI values were in the normal range, but in one juvenile diabetic and one patient with chronic renal failure, SLI was undetectable (<10 pg/ml). In a pair of monochorionic diamniotic twins, SLI levels were very different (33 and 197 pg/ml), which suggests that fetal factors are more important than materno-placental ones in determining amniotic fluid SLI. Serial dilutions of amniotic fluid showed parallelism with standard SRIF. When concentrates of pooled amniotic fluid were chromatographed on Sephadex G-25 columns, all SLI eluted in the void volume ahead of SRIF even after treatment with 8 M urea and dithiothreitol. This "big" SLI incubated in amniotic fluid showed 100% stability over 24 h at 37 degrees C, whereas SRIF was rapidly inactivated (t((1/2)) congruent with 7 min). Extracts of placenta and fetal membranes contained no SLI, but small amounts (6-20% of total amniotic fluid SLI) were found in cells from fresh fluid. Radioimmunoassay of SLI in extracts of seven paired cord arterial and venous plasma samples showed no arteriovenous gradient consistent with fetal origin of cord blood SLI. It is concluded that (a) amniotic fluid contains SLI which is of fetal origin and (b) normal levels vary with gestational age. The SLI has a higher molecular weight (>/=5,000) and is more stable in amniotic fluid than SRIF.


Assuntos
Líquido Amniótico/análise , Somatostatina/análise , Feminino , Sangue Fetal/análise , Feto/análise , Humanos , Placenta/análise , Pré-Eclâmpsia/metabolismo , Gravidez , Gravidez em Diabéticas/metabolismo , Radioimunoensaio , Somatostatina/imunologia , Estômago/análise , Gêmeos
8.
J Clin Invest ; 70(5): 1101-9, 1982 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7130395

RESUMO

The neuronal biosynthesis of somatostatin-like immunoreactivity (SLI) was investigated using mechanically dispersed neonatal rat hypothalamic cells kept in culture for up to 6 wk. Immunohistochemically, SLI was specifically localized to a small subpopulation of parvicellular neurons and their cell processes. By radioimmunoassay the cellular SLI content declined steadily during the first 2 wk in culture (nadir value of 60 fmol/dish at day 15) but then increased progressively to reach a maximum value of 381 fmol/dish at day 46. Gel chromatographic analysis showed this immunoreactivity to consist of forms corresponding to tetradecapeptide somatostatin (S-14), somatostatin-28 (S-28), and a 15,000-mol-wt molecule. After incubation of the cells with [3H]phenylalanine, the cellular extracts, purified by adsorption to C18 silica, contained material that bound specifically to an immobilized antisomatostatin antibody. Analysis by gel chromatography and high performance liquid chromatography of the specifically bound label provided evidence for the presence of labeled S-14, S-28, and the 15,000-mol-wt molecule. Pulse-chase experiments (20-min pulse, 20-min chase) demonstrated a transfer of radioactivity from the 15,000-mol-wt form to material corresponding to S-14 as well as to S-28. These studies demonstrate that cultured hypothalamic neurons are capable of synthesizing three somatostatin-like peptides (15,000-mol-wt SLI, S-28, S-14), one of which (15,000-mol-wt SLI) serve as a biosynthetic precursor for both S-28 and S-14. This in vitro system should provide a powerful tool for further investigation of the biosynthesis and regulation of biosynthesis of somatostatin in the hypothalamus.


Assuntos
Hipotálamo/metabolismo , Neurônios/metabolismo , Biossíntese Peptídica , Peptídeos , Animais , Agregação Celular , Células Cultivadas , Cromatografia em Gel , Cromatografia Líquida de Alta Pressão , Fenilalanina/metabolismo , Ratos , Ratos Endogâmicos
9.
J Clin Invest ; 75(4): 1249-55, 1985 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-2859301

RESUMO

In this study we have characterized the effects of cysteamine (CHS) on the cellular content and release of immunoreactive somatostatin (S-14 LI), insulin (IRI), and glucagon (IRG) from monolayer cultures of neonatal rat islets. Incubation of cultures with 0.1-10 mM CHS for 1 h led to an apparent, dose-dependent reduction of cellular S-14 LI that was 50% of control at 0.3 mM, 87% at 1 mM, and 95% at 10 mM. IRI content was unaffected by CHS up to 1 mM, but at 10 mM 90% loss of IRI occurred. All concentrations were without effect on IRG content. The loss of S-14 LI and IRI was completely reversible with time, but with different recovery rates for the two hormones (48 h for S-14 LI, and 72 h for IRI). Released S-14 LI rose progressively with increasing doses of CHS from 21 +/- 2.5 pg/ml per hour to 41 +/- 1.4 pg/ml per hour at CHS concentrations of 5 mM and 10 mM. IRI and IRG secretion were both also significantly enhanced (by 55% and 88%, respectively), despite the elevated medium S-14 LI. Since CHS reduced cellular S-14 LI but augmented medium S-14 LI, the relative effects of CHS (1 mM) and immunoneutralization with antibody to S-14 LI on IRI and IRG secretion were tested. Anti S-14 LI alone stimulated basal IRG (67%) but not IRI. Cultures rendered S-14 LI deficient with both CHS and anti-S-14 LI exhibited threefold and 2.3-fold potentiation of IRG and IRI secretions, respectively, greater than that expected from the separate effects of the two agents. Increasing medium glucose from 2.8 mM to 16.7 mM stimulated IRI release by 86% and suppressed IRG by 53%. CHS (1 mM) and anti-S-14 LI further augmented stimulated IRI release, by 30%; although 16.7 mM glucose suppression of IRG was still maintained under these conditions, the quantitative IRG response was significantly greater. These results suggest that CHS induces an apparent loss of islet S-14 LI, and at high doses, of IRI as well, but has no effect on A cells. Complete islet S-14 LI deficiency augments IRI and IRG secretion over a wide range of glucose concentrations, suggesting a physiological role of D cells on B cell and A cell regulation. D cell modulation of B cells requires cellular but not extracellular S-14 LI, being mediated probably though direct intracellular communication, whereas the A cells seem to be regulated by both direct contact as well as through locally secreted S-14 LI.


Assuntos
Anticorpos/imunologia , Cisteamina/farmacologia , Glucagon/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Somatostatina/metabolismo , Animais , Células Cultivadas , Glucose/farmacologia , Secreção de Insulina , Ilhotas Pancreáticas/metabolismo , Ratos , Ratos Endogâmicos , Somatostatina/deficiência , Somatostatina/imunologia
10.
J Clin Invest ; 107(12): 1571-80, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11413165

RESUMO

Pulsatile growth hormone (GH) secretion differs between males and females and regulates the sex-specific expression of cytochrome P450s in liver. Sex steroids influence the secretory dynamics of GH, but the neuroendocrine mechanisms have not been conclusively established. Because periventricular hypothalamic somatostatin (SST) expression is greater in males than in females, we generated knockout (Smst(-/-)) mice to investigate whether SST peptides are necessary for sexually differentiated GH secretion and action. Despite marked increases in nadir and median plasma GH levels in both sexes of Smst(-/-) compared with Smst(+/+) mice, the mutant mice had growth curves identical to their sibling controls and retained a normal sexual dimorphism in weight and length. In contrast, the liver of male Smst(-/-) mice was feminized, resulting in an identical profile of GH-regulated hepatic mRNAs between male and female mutants. Male Smst(-/-) mice show higher expression of two SST receptors in the hypothalamus and pituitary than do females. These data indicate that SST is required to masculinize the ultradian GH rhythm by suppressing interpulse GH levels. In the absence of SST, male and female mice exhibit similarly altered plasma GH profiles that eliminate sexually dimorphic liver function but do not affect dimorphic growth.


Assuntos
Hormônio do Crescimento/fisiologia , Fígado/metabolismo , Caracteres Sexuais , Somatostatina/fisiologia , Animais , Peso Corporal , Feminino , Genótipo , Hormônio do Crescimento/metabolismo , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Knockout , Hipófise/metabolismo , RNA Mensageiro/biossíntese , Receptores de Somatostatina/biossíntese , Receptores de Somatostatina/genética , Recombinação Genética , Somatostatina/genética , Transcrição Gênica
11.
Trends Pharmacol Sci ; 16(3): 86-8, 1995 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-7792934

RESUMO

There is considerable controversy about the classification and nomenclature of somatostatin receptors. To date, five distinct receptor genes have been cloned and named chronologically according to their respective publication dates, but two were unfortunately given the same appellation (SSTR4). Consensually, a nomenclature for the recombinant receptors has been agreed according to IUPHAR guidelines (sst1, sst2, sst3, sst4, and sst5). However, a more informative classification is to be preferred for the future, employing all classification criteria in an integrated scheme. It is already apparent that the five recombinant receptors fall into two classes or groups, on the basis of not only structure but also pharmacological characteristics. One class (already referred to by some as SRIF1) appears to comprise sst2, sst3 and sst5 receptor subtypes. The other class (SRIF2) appears to comprise the other two recombinant receptor subtypes (sst1 and sst4). This promising approach is discussed but it is acknowledged that much more data from endogenous receptors in whole tissues are needed before further recommendations on somatostatin receptor nomenclature can be made.


Assuntos
Receptores de Somatostatina/classificação , Somatostatina/química , Sequência de Aminoácidos , Dados de Sequência Molecular , Octreotida/análogos & derivados , Octreotida/química , Peptídeos Cíclicos/química , Receptores de Somatostatina/antagonistas & inibidores , Receptores de Somatostatina/química , Proteínas Recombinantes/química , Proteínas Recombinantes/classificação , Somatostatina/análogos & derivados , Terminologia como Assunto
12.
Diabetes ; 36(7): 849-52, 1987 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-2884158

RESUMO

We previously reported that the BB diabetic rat is characterized by a reduction in pancreatic immunoreactive somatostatin (SLI) content, delta-cell mass, and delta-cell secretory reserve. Despite this, portal plasma SLI levels are elevated in diabetic animals and normalized by insulin therapy. These findings comprise indirect evidence for SLI hypersecretion by the gut in untreated BB rats. This study was undertaken with isolated stomach perfusions to investigate directly the secretory status of gastric delta-cells in this diabetic model. Isolated stomachs of three groups of insulin-treated diabetic, untreated diabetic, and nondiabetic control rats were perfused in situ under basal and glucagon-stimulated (5 nM) conditions. Untreated diabetic BB rats exhibited significant enhancement of basal and glucagon-stimulated gastric SLI release. Insulin treatment reduced gastric SLI release to significantly subnormal levels. More than 95% of basal and stimulated SLI released in diabetic BB and normal control rats coeluted with synthetic somatostatin-14 on Sephadex G-50 columns. We conclude that basal and stimulated gastric SLI release is increased in untreated BB rats and is suppressed with insulin therapy, gastric delta-cell hyperfunction accounts for portal vein hypersomatostatinemia characteristic of untreated diabetic BB rats, and somatostatin-14 is the main molecular form of SLI released from normal and diabetic stomachs.


Assuntos
Diabetes Mellitus/tratamento farmacológico , Mucosa Gástrica/metabolismo , Insulina/uso terapêutico , Somatostatina/metabolismo , Animais , Cromatografia em Gel , Diabetes Mellitus/metabolismo , Modelos Animais de Doenças , Mucosa Gástrica/efeitos dos fármacos , Glucagon , Masculino , Perfusão , Veia Porta , Ratos , Ratos Endogâmicos , Somatostatina/sangue
13.
Diabetes ; 38(6): 752-7, 1989 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-2470628

RESUMO

In streptozocin-induced diabetes in rats, there is a marked increase in the content and release of immunoreactive somatostatin (SLI) from the pancreas and upper gut. To elucidate whether these SLI changes are associated with alterations in somatostatin gene transcription, we measured somatostatin mRNA (SmRNA) accumulation in these and other SLI-producing tissues. Pancreas, stomach, jejunum, hypothalamus, and cerebral cortex were removed from control rats, 6-wk-diabetic rats, and diabetic rats treated with insulin for 6 wk. Total RNA was isolated by centrifugation through CsCl and fractionated on agarose gels. A sensitive radiodensitometric hybridization assay was used to determine SmRNA levels in absolute amounts by in vitro synthesized sense-strand RNA as a quantitative standard and antisense cRNA as a specific probe. SLI was determined by radioimmunoassay. SmRNA exhibited size heterogeneity between the different control and diabetic tissues. A 2- to 3-fold increase in total SmRNA was found in pancreas and stomach of the diabetic rats that suppressed toward normal with insulin treatment. These two tissues also exhibited significant 1.6- to 2.6-fold increases in SLI, respectively. The remaining tissues showed no diabetes-related changes in SLI or SmRNA. We conclude that in insulinopenic diabetes, tissue SLI and SmRNA accumulation undergo parallel changes; are increased in pancreas and upper gut, reflecting augmented somatostatin synthesis; are reciprocally related to insulin acting directly or indirectly on somatostatin-producing cells; and are unchanged in the lower gut and brain, suggesting tissue-specific regulation of somatostatin gene transcription in diabetes.


Assuntos
Diabetes Mellitus Experimental/genética , RNA Mensageiro/genética , Somatostatina/genética , Transcrição Gênica , Animais , Diabetes Mellitus Experimental/metabolismo , Masculino , Especificidade de Órgãos , RNA/genética , RNA Antissenso , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/isolamento & purificação , Ratos , Ratos Endogâmicos , Valores de Referência
14.
Diabetes ; 29(9): 757-61, 1980 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-6108276

RESUMO

Immunoreactive somatostatin (IRS) was measured in extracted plasma obtained from the hepatic portal vein (PV) and inferior vena cava (IVC) of acute, untreated, spontaneously diabetic Wistar rats (BBL), insulin-treated diabetic rats, and nondiabetic controls. Acetic acid extracts of the pancreas and entire gastrointestinal tract were assayed for IRS, and the volume density of pancreatic D-, A-, and B-cells was determined by quantitative morphometry. The concentration of IRS in the PV and IVC of the untreated diabetic rats was significantly elevated compared with controls, with a much greater percent increase in the IVC compared with the PV. Insulin treatment for 4-6 wk restored the elevated PV and IVC levels to control values. The pancreatic content of IRS and the volume density of D-cells was severely reduced in the diabetic groups whereas gut IRS was unchanged. These data suggest that the elevated blood levels are secondary to insulin deficiency and result from altered peripheral metabolism and/or increased secretion of IRS most probably from the gut. The increased peripheral blood concentration of IRS raises the possibility of an endocrine role of circulating somatostatin in diabetes. The reduction in pancreatic IRS found in this model is probably secondary to insulitis and contrasts with the D-cell augmentation reported in streptozotocin-diabetic rats.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Insulina/farmacologia , Somatostatina/sangue , Animais , Peso Corporal , Imunofluorescência , Mucosa Gástrica/metabolismo , Glucagon/metabolismo , Glucose/metabolismo , Insulina/metabolismo , Mucosa Intestinal/metabolismo , Ilhotas Pancreáticas/metabolismo , Ratos , Somatostatina/metabolismo
15.
Diabetes ; 28(1): 35-40, 1979 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-759249

RESUMO

The isolated perfused canine pancreas with duodenal exclusion was used to examine islet hormone output in response to arginine and exogenous glucagon and insulin. Exogenous glucagon (100 ng/ml) stimulated insulin and somatostatin secretion, which occurred in a biphasic pattern. The insulin response to glucagon was markedly enhanced by increased perfusate glucose, unlike the somatostatin response, which was little affected. The insulin and somatostatin responses were seen between 15 and 45 s after the glucagon stimulus. Pancreatic polypeptide secretion was uninfluenced by exogenous glucagon. Biphasic release of glucagon, somatostatin, and pancreatic polypeptide was evoked by 10 mM arginine, the responses first being apparent within less than 30 s. Exogenous insulin (50 mU/ml) infused for 10 min had no statistically significant effect on glucagon, somatostatin, or pancreatic polypeptide secretion. This study suggests that these four islet hormones may all be involved in the dynamic mechanisms of nutrient metabolism. In addition, potential intra-islet paracrine effects are identified.


Assuntos
Arginina/farmacologia , Glucagon/farmacologia , Insulina/farmacologia , Pâncreas/efeitos dos fármacos , Polipeptídeo Pancreático/metabolismo , Somatostatina/metabolismo , Animais , Cães , Glucose/administração & dosagem , Técnicas In Vitro , Pâncreas/metabolismo
16.
Diabetes ; 48(1): 77-85, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9892225

RESUMO

We have developed a panel of rabbit polyclonal antipeptide antibodies against the five human somatostatin receptor subtypes (hSSTR1-5) and used them to analyze the pattern of expression of hSSTR1-5 in normal human islet cells by quantitative double-label confocal fluorescence immunocytochemistry. All five hSSTR subtypes were variably expressed in islets. The number of SSTR immunopositive cells showed a rank order of SSTR1 > SSTR5 > SSTR2 > SSTR3 > SSTR4. SSTR1 was strongly colocalized with insulin in all beta-cells. SSTR5 was also an abundant isotype, being colocalized in 87% of beta-cells. SSTR2 was found in 46% of beta-cells, whereas SSTR3 and SSTR4 were relatively poorly expressed. SSTR2 was strongly colocalized with glucagon in 89% of alpha-cells, whereas SSTR5 and SSTR1 colocalized with glucagon in 35 and 26% of alpha-cells, respectively. SSTR3 was detected in occasional alpha-cells, and SSTR4 was absent. SSTR5 was preferentially expressed in 75% of SST-positive cells and was the principal delta-cell SSTR subtype, whereas SSTR1-3 were colocalized in only a few delta-cells, and SSTR4 was absent. These studies reveal predominant expression of SSTR1, SSTR2, and SSTR5 in human islets. Beta-cells, alpha-cells, and delta-cells each express multiple SSTR isoforms, beta-cells being rich in SSTR1 and SSTR5, alpha-cells in SSTR2, and delta-cells in SSTR5. Although there is no absolute specificity of any SSTR for an islet cell type, SSTR1 is beta-cell selective, and SSTR2 is alpha-cell selective. SSTR5 is well expressed in beta-cells and delta-cells and moderately well expressed in alpha-cells, and thereby it lacks the islet cell selectivity displayed by SSTR1 and SSTR2. Subtype-selective SSTR expression in islet cells could be the basis for preferential insulin suppression by SSTR1-specific ligands and of glucagon inhibition by SSTR2-selective compounds.


Assuntos
Ilhotas Pancreáticas/metabolismo , Receptores de Somatostatina/metabolismo , Animais , Linhagem Celular , Glucagon/metabolismo , Humanos , Imuno-Histoquímica , Insulina/metabolismo , Ilhotas Pancreáticas/citologia , Isomerismo , Coelhos , Somatostatina/metabolismo , Especificidade por Substrato , Distribuição Tecidual
17.
Trends Endocrinol Metab ; 8(10): 398-405, 1997 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18406829

RESUMO

The diverse biological effects of somatostatin (SRIF) are mediated by a family of G protein-coupled receptors (termed sst) that are encoded by five nonallelic genes located on separate chromosomes. The receptors can be further divided into two subfamilies: sst(2,3,5) react with octapeptide and hexapeptide SRIF analogues and belong to one subclass; sst(1,4) react poorly with these compounds and fall into another subclass. This review focuses on the molecular pharmacology and function of these receptors, with particular emphasis on the ligand-binding domain, subtype-selective analogues, agonist-dependent receptor regulation and desensitization responses, subtype-specific effector coupling, and signal transduction pathways responsible for inhibiting cell secretion and cell growth or induction of apoptosis.

18.
Mol Endocrinol ; 10(12): 1688-96, 1996 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8961277

RESUMO

Somatostatin (SST) exerts direct antiproliferative effects in tumor cells, triggering either growth arrest or apoptosis. The cellular actions of SST are transduced through a family of five distinct somatostatin receptor subtypes (SSTR1-5). Whereas growth inhibition has been reported to follow stimulation of protein tyrosine phosphatase via SSTR2 or inhibition of Ca2+ channels via SSTR5 in heterologous expression systems, the subtype selectivity for signaling apoptosis has not been investigated. The tumor suppressor protein p53 and the protooncogene product c-Myc regulate cell cycle progression (growth factors present) or apoptosis (growth factors absent). The p53-induced G1 arrest requires induction of p21, an inhibitor of cyclin-dependent kinases, whereas apoptosis requires induction of Bax. c-Myc is capable of abrogating p53-induced G1 arrest by interfering with the inhibitory action of p21 on cyclin-dependent kinases. We have, therefore, investigated the regulation of p53, p21, c-Myc, and Bax and cellular apoptosis in relation to cell cycle progression in CHO-K1 cells stably expressing individual human SSTR1-5. We demonstrate that apoptosis is signaled uniquely through human SSTR3 and is associated with dephosphorylation-dependent conformational change in wild-type (wt) p53 as well as induction of Bax. The induction of wt p53 occurs rapidly and precedes the onset of apoptosis. We show that the increase in wt p53 is not associated with the induction of p21 or c-Myc when octreotide-induced apoptosis becomes evident, suggesting that such apoptosis does not require G1 arrest and is not c-Myc dependent. These findings provide the first evidence for hormonal induction of wt p53-associated apoptosis via G protein-coupled receptor in a subtype-selective manner.


Assuntos
Apoptose/fisiologia , Ciclo Celular/fisiologia , Proteínas Proto-Oncogênicas c-bcl-2 , Receptores de Somatostatina/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Antineoplásicos Hormonais/farmacologia , Apoptose/efeitos dos fármacos , Células CHO/efeitos dos fármacos , Células CHO/metabolismo , Ciclo Celular/efeitos dos fármacos , Cricetinae , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/efeitos dos fármacos , Ciclinas/genética , Ciclinas/metabolismo , Proteínas de Ligação ao GTP/efeitos dos fármacos , Proteínas de Ligação ao GTP/metabolismo , Regulação Neoplásica da Expressão Gênica , Hormônios , Humanos , Octreotida/farmacologia , Proteínas Proto-Oncogênicas/efeitos dos fármacos , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-myc/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Receptores de Somatostatina/genética , Transdução de Sinais , Proteína Supressora de Tumor p53/efeitos dos fármacos , Proteína Supressora de Tumor p53/genética , Proteína X Associada a bcl-2
19.
Mol Endocrinol ; 13(1): 82-90, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9892014

RESUMO

Ligand-activated somatostatin receptors (SSTRs) initiate cytotoxic or cytostatic antiproliferative signals. We have previously shown that cytotoxicity leading to apoptosis was signaled solely via human (h) SSTR subtype 3, whereas the other four hSSTR subtypes initiated a cytostatic response that led to growth inhibition. In the present study we characterized the antiproliferative signaling mediated by hSSTR subtypes 1, 2, 4, and 5 in CHO-K1 cells. We report here that cytostatic signaling via these subtypes results in induction of the retinoblastoma protein Rb and G1 cell cycle arrest. Immunoblot analysis revealed an increase in hypophosphorylated form of Rb in agonist-treated cells. The relative efficacy of these receptors to initiate cytostatic signaling was hSSTR5 > hSSTR2 > hSSTR4 approximately = hSSTR1. Cytostatic signaling via hSSTR5 also induced a marginal increase in cyclin-dependent kinase inhibitor p21. hSSTR5-initiated cytostatic signaling was G protein dependent and protein tyrosine phosphatase (PTP) mediated. Octreotide treatment induced a translocation of cytosolic PTP to the membrane, whereas it did not stimulate PTP activity when added directly to the cell membranes. C-tail truncation mutants of hSSTR5 displayed progressive loss of antiproliferative signaling proportional to the length of deletion, as reflected by the marked decrease in the effects of octreotide on membrane translocation of cytosolic PTP, and induction of Rb and G1 arrest. These data demonstrate that the C-terminal domain of hSSTR5 is required for cytostatic signaling that is PTP dependent and leads to induction of hypophosphorylated Rb and G1 arrest.


Assuntos
Fase G1/fisiologia , Receptores de Somatostatina/metabolismo , Proteína do Retinoblastoma/metabolismo , Animais , Células CHO/efeitos dos fármacos , Células CHO/metabolismo , Cricetinae , Citosol/metabolismo , Fase G1/efeitos dos fármacos , Proteínas de Ligação ao GTP/efeitos dos fármacos , Proteínas de Ligação ao GTP/metabolismo , Hormônios/farmacologia , Humanos , Proteínas de Membrana , Mutação , Octreotida/farmacologia , Fosforilação , Proteína Tirosina Fosfatase não Receptora Tipo 1 , Proteínas Tirosina Fosfatases/efeitos dos fármacos , Proteínas Tirosina Fosfatases/metabolismo , Receptores de Somatostatina/efeitos dos fármacos , Receptores de Somatostatina/genética , Proteína do Retinoblastoma/genética , Somatostatina/metabolismo , Somatostatina/farmacologia , Fatores de Virulência de Bordetella/farmacologia
20.
Endocrinology ; 136(6): 2389-96, 1995 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-7750460

RESUMO

We previously reported that dexamethasone (DEX) induces dose-dependent biphasic effects on steady state somatostatin (SS) messenger RNA (mRNA) levels in normal rat islet and islet SS-producing tumor cells (1027B2), characterized by stimulation at low doses and marked inhibition at high doses. The stimulatory effect is transcriptionally mediated, whereas the molecular mechanism underlying DEX-induced suppression of SS mRNA levels is unknown. In the present study, we investigated these mechanisms in human thyroid medullary carcinoma (TT) cells, which exhibit only inhibition of SS mRNA with DEX. Cultured TT cells synthesized and secreted large quantities of SS-like immunoreactivity (content, 90 ng/10(6) cells; release, 18 ng/10(6) cells/24h). DEX produced a dose-dependent reduction of both SS-like immunoreactivity secretion and SS mRNA levels, with a maximum inhibition of 60% at 10(-6) M at 48 h. In time-course studies, DEX inhibition of SS function occurred after a lag period of about 12 h, suggesting a posttranscriptional mechanism. To exclude a transcriptional effect of DEX on the SS gene, chloramphenicol acetyltransferase (CAT) activity was determined in TT cells acutely transfected with SS promoter (-750 base pairs) ligated to the receptor CAT gene. No inhibition of CAT activity occurred with DEX (10(-6) M) for 48 h. Furthermore, DEX did not influence the rate of SS gene transcription determined by nuclear run-on assay compared to approximately 2-fold stimulation by cAMP. Actinomycin D (inhibitor of mRNA synthesis) reduced the size of the SS mRNA transcript and rendered it resistant to DEX-induced degradation when coincubated with DEX, but not when it was added after a delay of 12 h, indicating that DEX destabilizes SS mRNA by an active process requiring ongoing gene transcription. Cycloheximide (inhibitor of protein synthesis) reduced SS mRNA levels to the same level as DEX, suggesting that the two agents promote SS mRNA degradation through a common pathway. We conclude that glucocorticoids inhibit steady state SS mRNA levels in TT cells. This effect is not mediated through direct transcriptional inhibition of the SS gene. It requires transcription of another gene(s) whose product(s) accelerates SS mRNA degradation.


Assuntos
Dexametasona/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Somatostatina/genética , Carcinoma Medular/genética , Carcinoma Medular/metabolismo , Cicloeximida/farmacologia , Dactinomicina/farmacologia , Dexametasona/administração & dosagem , Relação Dose-Resposta a Droga , Expressão Gênica/efeitos dos fármacos , Humanos , Cinética , Somatostatina/metabolismo , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/metabolismo , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA