Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Med Virol ; 82(11): 1950-7, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20872723

RESUMO

Despite evidence supporting an association between enterovirus (EV) infection and type 1 diabetes, the etiological mechanism(s) for EV-induced beta cell destruction is(are) not well understood. In this study, the effects of Coxsackievirus B (CVB) 1-6 on cell lysis and cytokine/chemokine expression in the insulinoma-1 (INS-1) beta cell line were investigated. Cytolysis was assessed using tissue culture infectious dose 50 (TCID(50)). Quantitative RT-PCR was used to measure viral RNA and mRNA of cytokines interferon (IFN)-α, IFN-ß, IFN-γ, tumor necrosis factor (TNF)-α, and chemokine (C-X-C motif) ligand 10 (CXCL10), chemokine (C-C motif) ligand 2 (CCL2), and chemokine (C-C motif) ligand 5 (CCL5) in infected INS-1 cells. CVB2, 4, 5, and 6 lysed and replicated in INS-1 cells; TCID(50) was lowest for CVB5 and highest for CVB6. IFN-γ, CXCL10, and CCL5 mRNA levels all increased significantly following infection with CVB2, 4, 5, and 6 (P<0.05). CCL2 mRNA increased with CVB2, 5, and 6 (P<0.05), IFN-α mRNA increased with CVB5 infection (P<0.05), while TNF-α mRNA and IFN-ß mRNA (P<0.001) increased with CVB2 infection. Dose-dependent effects of infection on cytokine mRNA levels were observed for all (P<0.01) except IFN-γ. Following inoculation of INS-1 cells with CVB1 and 3, viral RNA was not detected and cytokine/chemokine mRNA levels were unchanged. In conclusion, CVB2, 4, 5, and 6 induce dose-dependent cytokine and chemokine mRNA production from INS-1 cells suggesting that pro-inflammatory cytokine and chemokine secretion by beta cells is a potential mechanism for EV-induced beta cell damage in type 1 diabetes.


Assuntos
Quimiocinas/metabolismo , Citocinas/metabolismo , Enterovirus Humano B/imunologia , Enterovirus Humano B/patogenicidade , Inflamação/imunologia , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/metabolismo , Animais , Linhagem Celular Tumoral , Quimiocina CXCL10/metabolismo , Quimiocinas CC/metabolismo , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/virologia , Humanos , Inflamação/metabolismo , Células Secretoras de Insulina/virologia , Insulinoma/imunologia , Insulinoma/metabolismo , Insulinoma/virologia , Interferons/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Fator de Necrose Tumoral alfa/metabolismo
2.
Metabolism ; 58(2): 239-45, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19154958

RESUMO

Little is known about the associations between adiponectin and its oligomeric isoforms with female sex steroids, and the relevance of these relationships to insulin sensitivity in women. In a cross-sectional study of 32 healthy women (12 premenopausal, 10 postmenopausal, and 10 early pregnant), we investigated the correlations of total adiponectin and the high-, medium-, and low-molecular weight oligomers (HMW, MMW, and LMW, respectively) with estrogen, progesterone, adiposity, and insulin resistance. Fat mass and serum concentrations of estradiol, progesterone, insulin, glucose, and total and isoform adiponectin were measured. The homeostasis model assessment of insulin resistance (HOMA-IR) was calculated. Serum concentrations of total and HMW adiponectin were highest in postmenopausal women and lowest in pregnant women. Concentrations of the MMW and LMW isoforms were not significantly different between the 3 groups. Total adiponectin, HMW adiponectin, and MMW adiponectin were negatively associated with estradiol and progesterone; but no associations between the LMW isoform and female sex steroids were observed. Fat mass and HOMA-IR were highest in pregnant women and lowest in premenopausal women. The HOMA-IR was positively associated with fat mass, estradiol, and progesterone, and negatively associated with total, HMW, and MMW adiponectin. Multivariate stepwise regression analysis revealed that fat mass explained 34% of the variance in HOMA-IR and that total and isoform adiponectin contributed an additional 10% to 15%. In the multivariate linear regression analysis, there were significant interactions of estradiol and progesterone with adiponectin or fat mass in the associations with HOMA-IR. In conclusion, there are strong negative associations of serum adiponectin and some of its isoforms with estradiol and progesterone. Female sex steroids are likely to affect insulin sensitivity through modulation of adiponectin and body fat.


Assuntos
Tecido Adiposo/metabolismo , Estradiol/sangue , Resistência à Insulina/fisiologia , Progesterona/sangue , Adiponectina/sangue , Adiponectina/química , Adulto , Idoso , Glicemia/metabolismo , Estudos Transversais , Feminino , Homeostase/fisiologia , Humanos , Insulina/sangue , Isomerismo , Modelos Lineares , Peso Molecular , Análise Multivariada , Gravidez
3.
Am J Physiol Endocrinol Metab ; 293(1): E364-71, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17456639

RESUMO

Muscle is a target of growth hormone (GH) action and a major contributor to whole body metabolism. Little is known about how GH regulates metabolic processes in muscle or the extent to which muscle contributes to changes in whole body substrate metabolism during GH treatment. To identify GH-responsive genes that regulate substrate metabolism in muscle, we studied six hypopituitary men who underwent whole body metabolic measurement and skeletal muscle biopsies before and after 2 wk of GH treatment (0.5 mg/day). Transcript profiles of four subjects were analyzed using Affymetrix GeneChips. Serum insulin-like growth factor I (IGF-I) and procollagens I and III were measured by RIA. GH increased serum IGF-I and procollagens I and III, enhanced whole body lipid oxidation, reduced carbohydrate oxidation, and stimulated protein synthesis. It induced gene expression of IGF-I and collagens in muscle. GH reduced expression of several enzymes regulating lipid oxidation and energy production. It reduced calpain 3, increased ribosomal protein L38 expression, and displayed mixed effects on genes encoding myofibrillar proteins. It increased expression of circadian gene CLOCK, and reduced that of PERIOD. In summary, GH exerted concordant effects on muscle expression and blood levels of IGF-I and collagens. It induced changes in genes regulating protein metabolism in parallel with a whole body anabolic effect. The discordance between muscle gene expression profiles and metabolic responses suggests that muscle is unlikely to contribute to GH-induced stimulation of whole body energy and lipid metabolism. GH may regulate circadian function in skeletal muscle by modulating circadian gene expression with possible metabolic consequences.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Hormônio do Crescimento Humano/farmacologia , Hipopituitarismo/genética , Hipopituitarismo/metabolismo , Redes e Vias Metabólicas/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Análise de Sequência com Séries de Oligonucleotídeos , Adulto , Idoso , Colágeno/genética , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/genética , Perfilação da Expressão Gênica , Terapia de Reposição Hormonal , Hormônio do Crescimento Humano/uso terapêutico , Humanos , Hipopituitarismo/tratamento farmacológico , Fator de Crescimento Insulin-Like I/genética , Masculino , Redes e Vias Metabólicas/genética , Pessoa de Meia-Idade , Músculo Esquelético/metabolismo
4.
Endocrinology ; 148(5): 2417-23, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17272397

RESUMO

Activation of the Janus kinase 2 (JAK2)/signal transducer and activator of transcription 5 (STAT5) pathway by GH is terminated by the suppressors of cytokine signaling (SOCSs) and protein tyrosine phosphatases, Src homology 2 domain-containing protein tyrosine phosphatase (SHP)-1 and SHP-2. Based on our recent report that estrogen inhibits GH signaling by stimulating SOCS-2 expression, we investigated the effects of selective estrogen receptor modulators (SERMs) on GH signaling in human embryonic kidney (HEK293) and breast cancer (MDA-MB-231) cells expressing human GH receptor and estrogen receptor-alpha. 17beta-estradiol (E(2)) suppressed GH activation of a STAT5-responsive luciferase reporter and JAK2 phosphorylation in both cell models. 4-hydroxytamoxifen and raloxifene augmented these actions of GH in HEK293 cells but not breast cancer cells. SOCS-2 expression in both cell types was stimulated by E(2) but unaffected by SERMs. In HEK293 cells, SHP-1 was inhibited by raloxifene and 4-hydroxytamoxifen, whereas the latter additionally inhibited SHP-2. The phosphatases were unaffected by E(2). In breast cancer cells, phosphatase activity was not altered by SERMs or E(2). In summary, estrogen inhibited the JAK2/STAT5 signaling of GH and stimulated SOCS-2 expression in both HEK293 and breast cancer cells. By contrast, SERMs augmented GH signaling by reducing SHP activities in HEK293 cells and had no effect on both in breast cancer cells. We provide the first evidence for a novel mechanism regulating GH signaling, in which SERMs enhance GH activation of the JAK2/STAT5 pathway in a cell-type-dependent manner by attenuating protein tyrosine phosphatase activities.


Assuntos
Hormônio do Crescimento Humano/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Receptores de Estrogênio/antagonistas & inibidores , Transdução de Sinais/fisiologia , Neoplasias da Mama , Linhagem Celular Tumoral , Dactinomicina/farmacologia , Antagonistas de Estrogênios/farmacologia , Estrogênios/farmacologia , Humanos , Janus Quinase 2/metabolismo , Rim/citologia , Inibidores da Síntese de Proteínas/farmacologia , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Cloridrato de Raloxifeno/farmacologia , Receptores de Estrogênio/metabolismo , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Tamoxifeno/farmacologia
5.
J Clin Endocrinol Metab ; 90(7): 3897-903, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15855258

RESUMO

CONTEXT: The endocrine and metabolic functions of the liver are affected by estrogen. Oral estrogen reduces IGF-I and suppresses fat oxidation despite augmenting GH secretion. The aim of this study was to determine whether selective estrogen receptor modulators display similar effects and whether these effects are magnified in GH-deficient (GHD) women because of the loss of GH feedback. DESIGN: This was an open-label, randomized, two-period, crossover study comparing treatment (raloxifene vs. estradiol) and group (normal vs. GHD). SETTING: The setting of this study was a clinical research unit. PARTICIPANTS: Twelve postmenopausal women and 12 women with hypopituitarism participated in this study. INTERVENTION: Two 4-wk treatments with 17beta-estradiol (E2; 2 mg, followed by 4 mg) or raloxifene (60 mg, followed by 120 mg) were given, crossing over to the alternate treatment after a 4-wk washout period. OUTCOME MEASURES: Endocrine [GH, IGF-I, IGF-binding protein-3 (IGFBP-3), GH-binding protein, and SHBG] and metabolic (fat oxidation) end points were used as outcome measures. RESULTS: E2 reduced serum IGF-I levels in a dose-dependent manner in both groups, with effects greater (P < 0.05) than raloxifene. Raloxifene reduced IGF-I levels in the GHD group (P < 0.001), but not in the postmenopausal group. E2 reduced (P < 0.05), and raloxifene increased (P < 0.05), IGFBP-3 levels in both groups. E2, but not raloxifene, increased GH (P < 0.05) in postmenopausal women. The effects of E2 and raloxifene on IGF-I, IGFBP-3, IGF-I/IGFBP-3 molar ratio, GH-binding protein, and SHBG were significantly different (P < 0.05). E2 and raloxifene reduced (P < 0.05) fat oxidation equally in GHD, whereas the decrease in postmenopausal women was not significant. CONCLUSION: E2 and raloxifene exert different hepatic endocrine, but not lipid oxidative, effects. The greater effects seen in GHD women may be explained by the loss of endogenous GH feedback.


Assuntos
Estradiol/farmacologia , Hormônio do Crescimento Humano/deficiência , Pós-Menopausa/metabolismo , Cloridrato de Raloxifeno/farmacologia , Adolescente , Adulto , Estudos Cross-Over , Metabolismo Energético/efeitos dos fármacos , Feminino , Hormônio do Crescimento Humano/sangue , Humanos , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/sangue , Fator de Crescimento Insulin-Like I/análise , Metabolismo dos Lipídeos , Pessoa de Meia-Idade , Oxirredução , Globulina de Ligação a Hormônio Sexual/análise
6.
J Clin Endocrinol Metab ; 90(7): 3989-94, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15827107

RESUMO

CONTEXT: Symptoms of fluid retention in GH-deficient patients during GH replacement are greater in men than in women, suggesting that testosterone may augment or estradiol may attenuate the antinatriuretic actions of GH. The mechanisms underlying the sodium-retaining effects of GH are poorly understood. AIM: The aim of this study was to investigate the effects of GH and testosterone, alone and in combination, on extracellular water (ECW) and the hormonal mechanisms involved. DESIGN: Two separate, open-label, randomized, two-period, crossover studies were performed; the first compared the effects of GH alone with those of GH and testosterone, and the second compared the effects of testosterone alone with those of GH and testosterone. PARTICIPANTS: Twelve hypopituitary men with GH deficiency and hypogonadism were studied. INTERVENTION: During the weeks of intervention, GH (0.5 mg/d) and testosterone enanthate (250 mg) were administered by im injection. OUTCOME MEASURES: The outcome measures were ECW, IGF-I, plasma renin activity (PRA), aldosterone (Aldo), and atrial natriuretic peptide (ANP). RESULTS: GH treatment significantly increased (P < 0.05) both IGF-I and ECW, and these changes were enhanced by cotreatment with testosterone (P = 0.07 for both). PRA, Aldo, and ANP levels did not change. Testosterone treatment alone did not change the IGF-I concentration, whereas cotreatment with GH induced a marked increase. Testosterone alone increased (P < 0.05) ECW, and the effect was augmented (P < 0.01) by cotreatment with GH. Although PRA and ANP did not change, plasma Aldo decreased after single and combined treatments. CONCLUSION: GH and testosterone exerted independent and additive effects on ECW. The mechanisms of fluid retention for both hormones are likely to be exerted on the renal tubules. This is the first direct evidence that testosterone increases ECW.


Assuntos
Água Corporal/metabolismo , Líquido Extracelular/metabolismo , Terapia de Reposição Hormonal , Hormônio do Crescimento Humano/deficiência , Hormônio do Crescimento Humano/uso terapêutico , Hipopituitarismo/tratamento farmacológico , Testosterona/uso terapêutico , Adulto , Idoso , Aldosterona/sangue , Fator Natriurético Atrial/sangue , Estudos Cross-Over , Sinergismo Farmacológico , Humanos , Hipopituitarismo/metabolismo , Fator de Crescimento Insulin-Like I/análise , Masculino , Pessoa de Meia-Idade
7.
Endocr Rev ; 25(5): 693-721, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15466938

RESUMO

GH plays a pivotal role in regulating body growth and development, which is modulated by sex steroids. A close interplay between estrogen and GH leads to attainment of gender-specific body composition during puberty. The physiological basis of the interaction is not well understood. Most previous studies have focused on the effects of estrogen on GH secretion. There is also strong evidence that estrogen modulates GH action independent of secretion. Oral but not transdermal administration of estrogen impairs the metabolic action of GH in the liver, causing a fall in IGF-I production and fat oxidation. This results in a loss of lean tissue and a gain of body fat in postmenopausal women and an impairment of GH effect in hypopituitary women on GH replacement. The negative metabolic sequelae are potentially important because of the widespread use of oral estrogen and estrogen-related compounds. Estrogen affects GH action at the level of receptor expression and signaling. More recently, estrogen has been shown to inhibit Janus kinase/signal transducer and activator of transcription signaling by GH via the induction of suppressor of cytokine signaling-2, a protein inhibitor for cytokine signaling. This represents a novel paradigm of steroid regulation of cytokine receptors and is likely to have significance for a diverse range of cytokine function.


Assuntos
Estrogênios/fisiologia , Hormônio do Crescimento/fisiologia , Homeostase , Animais , Composição Corporal , Proteínas de Transporte/fisiologia , Citocinas/fisiologia , Estrogênios/farmacologia , Estrogênios/uso terapêutico , Feminino , Crescimento , Humanos , Fator de Crescimento Insulin-Like I/fisiologia , Masculino , Receptores Citoplasmáticos e Nucleares/fisiologia , Receptores de Estrogênio/fisiologia , Receptores da Somatotropina/fisiologia , Transdução de Sinais
8.
Endocrinology ; 145(12): 5525-31, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15319356

RESUMO

Suppressors of cytokine signaling (SOCS) are important negative regulators of cytokine action. We recently reported that estrogen stimulates SOCS-2 expression and inhibits GH signaling in kidney cells. The effects of estrogen on SOCS expression in other tissues are unclear. The aim of this study was to investigate in vivo and in vitro whether estrogen affected SOCS expression in the liver, a major target organ of GH. The in vivo hepatic effects of estrogen on ovariectomized mice lacking estrogen receptor (ER)-alpha, ERbeta, or both and their wild-type littermates were examined by DNA microarray analysis. In vitro, the effects of estrogen on SOCS expression in human hepatoma cells were examined by reverse transcription quantitative PCR. Long-term (3 wk) estrogen treatment induced a 2- to 3-fold increase in hepatic expression of SOCS-2 and -3 in wild-type and ERbeta knockout mice but not in those lacking ERalpha or both ER subtypes. Short-term treatment (at 24 h) increased the mRNA level of SOCS-3 but not SOCS-2. In cultured hepatoma cells, estrogen increased SOCS-2 and -3 mRNA levels by 2-fold in a time- and dose-dependent manner (P < 0.05). Estrogen induced murine SOCS-3 promoter activity by 2-fold (P < 0.05) in constructs containing a region between nucleotides -1862 and -855. Moreover, estrogen and GH had additive effects on the SOCS-3 promoter activity. In summary, estrogen, via ERalpha, up-regulated hepatic expression of SOCS-2 and -3, probably through transcriptional activation. This indicates a novel mechanism of estrogen regulation of cytokine action.


Assuntos
Proteínas de Ligação a DNA/genética , Estradiol/farmacologia , Fígado/fisiologia , Proteínas Repressoras/genética , Transdução de Sinais/efeitos dos fármacos , Transativadores/genética , Animais , Carcinoma Hepatocelular , Linhagem Celular Tumoral , Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/genética , Feminino , Hormônio do Crescimento/farmacologia , Humanos , Técnicas In Vitro , Neoplasias Hepáticas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Regiões Promotoras Genéticas/fisiologia , Transdução de Sinais/fisiologia , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina , Fatores de Transcrição/genética , Regulação para Cima/efeitos dos fármacos
9.
Clin Endocrinol (Oxf) ; 59(6): 690-8, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14974909

RESUMO

OBJECTIVE: Oral but not transdermal oestrogen administration reduces IGF-I, and increases GH binding protein (GHBP) reflecting effects on hepatic endocrine function in postmenopausal women. As progestogens attenuate the effects of oestrogen on circulating lipid levels according to their androgenic properties, we have investigated the impact of progestogen types on the hepatic endocrine effects of oestrogen. DESIGN: Four progestogens differing in androgenicity were co-administered in a monthly cyclical regimen in random order to postmenopausal women receiving either oral (n = 9, premarin 1.25 mg) or transdermal (n = 10, Estraderm 100 microg patches twice weekly). The four progestogens were cyproterone acetate (CA 5 mg, antiandrogenic), dydrogesterone (20 mg, neutral), medroxyprogesterone acetate (MPA 10 mg, mildly androgenic), norethisterone (2.5 mg, androgenic). PATIENTS: Nineteen postmenopausal women (age 57 +/- 3 years, mean +/- SE) were studied. MEASUREMENTS: The effects of oestrogen alone and the combined effects with each progestogen type on IGF-I, GHBP, SHBG, cholesterol, triglycerides and lipoprotein(a) were investigated. RESULTS: Mean IGF-I fell while GHBP and SHBG levels increased with oral (P < 0.01) but not transdermal oestrogen administration. When the combined effects were examined, progestogens did not affect IGF-I, GHBP and SHBG during oral oestrogen treatment, while they significantly increased (P < 0.01) mean IGF-I levels during transdermal therapy. Among the progestogen types, only norethisterone prevented the fall in IGF-I induced by oral oestrogen. During transdermal therapy, MPA and norethisterone but not CA or dydrogesterone significantly increased (P < 0.005) IGF-I. The rise in GHBP induced by oral oestrogens tended to be lower during co-administration of MPA and norethisterone. The increase in SHBG induced by oral oestrogen was attenuated (P < 0.05) by norethisterone which was the only progestogen that lowered SHBG (P < 0.05) during transdermal oestrogen treatment. Mean IGF-I was higher (P < 0.001), GHBP and SHBG lower during co-administration of androgenic progestogens (MPA and norethisterone). CONCLUSIONS: Oestrogen effects on IGF-I, GHBP and SHBG are dependent on the route of administration with progestogens having variable effects. Among the progestogen types, norethisterone, the most androgenic, had the greatest effect, particularly on IGF-I. Progestogens modulate the effects of oestrogen on hepatic endocrine function in relation to their intrinsic androgenic properties. The modulatory effects of progestogens on IGF-I during oestrogen therapy may have long-term implications for lean body mass.


Assuntos
Estradiol/uso terapêutico , Terapia de Reposição de Estrogênios , Estrogênios Conjugados (USP)/uso terapêutico , Fígado/efeitos dos fármacos , Pós-Menopausa/metabolismo , Progestinas/uso terapêutico , Análise de Variância , Proteínas de Transporte/sangue , Acetato de Ciproterona/uso terapêutico , Didrogesterona/uso terapêutico , Feminino , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Lipídeos/sangue , Fígado/metabolismo , Acetato de Medroxiprogesterona/uso terapêutico , Pessoa de Meia-Idade , Noretindrona/uso terapêutico , Globulina de Ligação a Hormônio Sexual/análise
10.
Am J Physiol Endocrinol Metab ; 283(4): E836-43, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12217902

RESUMO

The 20-kDa growth hormone (GH) is generated from alternative splicing of the primary transcript of full-length 22-kDa GH. We have studied the regulation of 20-kDa GH over a range of pathophysiological conditions and in response to pharmacological stimulation using isoform-specific enzyme-linked immunosorbent assays (ELISAs). Mean 24-h levels of 20- and 22-kDa GH were higher in acromegaly and lower in GH deficiency than in normal subjects, with the 20-to-22-kDa ratio not different between the three groups. In normal subjects, 20-kDa GH was secreted in a pulsatile manner throughout the day, with peaks coinciding with those of 22-kDa GH. However, the half-life of 20-kDa GH (18.7 +/- 0.8 min) was significantly longer than that of 22-kDa GH (14.7 +/- 0.8 min; P < 0.02). Insulin-induced hypoglycemia, androgen, and oral estrogen caused a parallel and proportionate increase in both isoforms. Octreotide suppressed 20-kDa less rapidly than 22-kDa GH in blood. Administration of recombinant 22-kDa GH in normal subjects rapidly reduced the 20-kDa GH levels. In conclusion, 20-kDa GH is cosecreted with and circulates at a constant proportion of 22-kDa GH. The 20-kDa GH level is reduced by administration of exogenous 22-kDa GH, suggesting rapid negative feedback regulation on pituitary release.


Assuntos
Acromegalia/tratamento farmacológico , Acromegalia/metabolismo , Hormônio do Crescimento Humano/administração & dosagem , Hormônio do Crescimento Humano/sangue , Hipopituitarismo/tratamento farmacológico , Hipopituitarismo/metabolismo , Adolescente , Adulto , Idoso , Estrogênios/administração & dosagem , Retroalimentação Fisiológica/efeitos dos fármacos , Retroalimentação Fisiológica/fisiologia , Feminino , Hormônios Esteroides Gonadais/administração & dosagem , Hormônios/administração & dosagem , Hormônio do Crescimento Humano/deficiência , Humanos , Hipoglicemiantes/administração & dosagem , Insulina/administração & dosagem , Masculino , Pessoa de Meia-Idade , Octreotida/administração & dosagem , Testosterona/administração & dosagem
11.
Clin Endocrinol (Oxf) ; 56(3): 397-403, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11940053

RESUMO

OBJECTIVES: Galanin (GAL) is a neuropeptide widely expressed in the central and peripheral nervous system and in neuroendocrine tissue, including the adenohypophysis where, in humans, it is expressed in corticotrophs and in ACTH-producing adenomas. Previous analyses of human tissue have used antiserum against porcine GAL for detection of GAL immunoreactivity (GAL-IR) and no pathophysiological correlates have been reported. Given significant differences between the sequence of porcine and human GAL peptides, the aim of this study was to use antiserum raised against synthetic human GAL to investigate GAL-IR in non tumorous pituitaries and in pituitary adenomas, and to correlate GAL-IR with the clinical and hormonal characteristics of patients with Cushing's disease. PATIENTS: Six nontumorous pituitaries were obtained from autopsy and 151 pituitary adenomas, comprising 62 functioning (16 corticotroph, 26 somatotroph, 19 lactotroph and one thyrotroph) and 89 nonfunctioning adenomas, were obtained by surgery. RESULTS: All non tumorous pituitary glands showed GAL-IR in corticotrophs, in basophil cells within the neurohypophysis and in nerve fibres of the neurohypophysis. GAL-IR was found in a subset (10 of 16) of patients with ACTH-secreting tumours causing Cushing's syndrome. GAL-IR was rarely expressed in somatotroph adenomas and prolactinomas, but was expressed in approximately one-third of nonfunctioning tumours. GAL-IR was found in almost 90% of nonfunctioning tumours that were positive for ACTH. There were no significant differences in sex ratio, age at presentation or 24-h urinary free cortisol secretion in the subset of patients with Cushing's disease positive (n = 10) or negative (n = 6) for GAL-IR. However, Cushing's patients positive for GAL-IR tended to have smaller tumours and achieved a higher cure rate than those without (100 vs. 50%, P = 0.017). CONCLUSIONS: Galanin is present in normal and tumorous human pituitaries. In addition, GAL colocalizes exclusively in corticotrophs of normal pituitaries and is coexpressed almost exclusively in corticotrophs from functioning and nonfunctioning tumours. The finding that corticotroph adenomas can function irrespective of the presence of GAL suggests that GAL may not play a pathophysiological role in Cushing's disease. However, the better surgical outcome observed in patients with Cushing's disease who had tumours positive for GAL-IR suggests that the expression of GAL confers a less aggressive tumour phenotype.


Assuntos
Adenoma/química , Biomarcadores Tumorais/análise , Galanina/análise , Hipófise/química , Neoplasias Hipofisárias/química , Adenoma/complicações , Adenoma/cirurgia , Hormônio Adrenocorticotrópico/análise , Adulto , Síndrome de Cushing/etiologia , Síndrome de Cushing/metabolismo , Síndrome de Cushing/cirurgia , Feminino , Humanos , Técnicas Imunoenzimáticas , Masculino , Pessoa de Meia-Idade , Neoplasias Hipofisárias/complicações , Neoplasias Hipofisárias/cirurgia , Prognóstico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA