Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Mais filtros

Medicinas Complementares
Métodos Terapêuticos e Terapias MTCI
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Comput Biol ; 16(6): e1007848, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32598357

RESUMO

Contraceptive drugs intended for family planning are used by the majority of married or in-union women in almost all regions of the world. The two most prevalent types of hormones associated with contraception are synthetic estrogens and progestins. Hormonal based contraceptives contain a dose of a synthetic progesterone (progestin) or a combination of a progestin and a synthetic estrogen. In this study we use mathematical modeling to understand better how these contraceptive paradigms prevent ovulation, special focus is on understanding how changes in dose impact hormonal cycling. To explain this phenomenon, we added two autocrine mechanisms essential to achieve contraception within our previous menstrual cycle models. This new model predicts mean daily blood concentrations of key hormones during a contraceptive state achieved by administering progestins, synthetic estrogens, or a combined treatment. Model outputs are compared with data from two clinical trials: one for a progestin only treatment and one for a combined hormonal treatment. Results show that contraception can be achieved with synthetic estrogen, with progestin, and by combining the two hormones. An advantage of the combined treatment is that a contraceptive state can be obtained at a lower dose of each hormone. The model studied here is qualitative in nature, but can be coupled with a pharmacokinetic/pharamacodynamic (PKPD) model providing the ability to fit exogenous inputs to specific bioavailability and affinity. A model of this type may allow insight into a specific drug's effects, which has potential to be useful in the pre-clinical trial stage identifying the lowest dose required to achieve contraception.


Assuntos
Anticoncepcionais/uso terapêutico , Contracepção Hormonal , Ciclo Menstrual/efeitos dos fármacos , Progestinas/uso terapêutico , Adulto , Estrogênios/uso terapêutico , Feminino , Hormônio Foliculoestimulante/fisiologia , Humanos , Hipotálamo/efeitos dos fármacos , Hormônio Luteinizante/fisiologia , Modelos Biológicos , Ovário/efeitos dos fármacos , Hipófise/efeitos dos fármacos
2.
Theor Biol Med Model ; 9: 35, 2012 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-22870942

RESUMO

When hormones during the ovulatory cycle are shown in phase plane graphs, reported FSH and estrogen values form a specific pattern that resembles the leaning "&" symbol, while LH and progesterone (Pg) values form a "boomerang" shape. Graphs in this paper were made using data reported by Stricker et al. [Clin Chem Lab Med 2006;44:883-887]. These patterns were used to construct a simplistic model of the ovulatory cycle without the conventional "positive feedback" phenomenon. The model is based on few well-established relations:hypothalamic GnRH secretion is increased under estrogen exposure during two weeks that start before the ovulatory surge and lasts till lutheolysis.the pituitary GnRH receptors are so prone to downregulation through ligand binding that this must be important for their function.in several estrogen target tissue progesterone receptor (PgR) expression depends on previous estrogen binding to functional estrogen receptors (ER), while Pg binding to the expressed PgRs reduces both ER and PgR expression.Some key features of the presented model are here listed:High GnRH secretion induced by the recovered estrogen exposure starts in the late follicular phase and lasts till lutheolysis. The LH and FSH surges start due to combination of accumulated pituitary GnRH receptors and increased GnRH secretion. The surges quickly end due to partial downregulation of the pituitary GnRH receptors (64% reduction of the follicular phase pituitary GnRH receptors is needed to explain the reported LH drop after the surge). A strong increase in the lutheal Pg blood level, despite modest decline in LH levels, is explained as delayed expression of pituitary PgRs. Postponed pituitary PgRs expression enforces a negative feedback loop between Pg levels and LH secretions not before the mid lutheal phase.Lutheolysis is explained as a consequence of Pg binding to hypothalamic and pituitary PgRs that reduces local ER expression. When hypothalamic sensitivity to estrogen is diminished due to lack of local ERs, hypothalamus switches back to the low GnRH secretion rate, leading to low secretion of gonadotropins and to lutheolysis. During low GnRH secretion rates, previously downregulated pituitary GnRH receptors recover to normal levels and thus allow the next cycle.Possible implications of the presented model on several topics related to reproductive physiology are shortly discussed with some evolutionary aspects including the emergence of menopause.


Assuntos
Ciclo Menstrual/fisiologia , Modelos Biológicos , Ovulação/fisiologia , Ativinas/fisiologia , Evolução Biológica , Estrogênios/fisiologia , Retroalimentação Fisiológica , Feminino , Hormônio Foliculoestimulante/fisiologia , Humanos , Hipotálamo/fisiologia , Inibinas/fisiologia , Leptina/fisiologia , Hormônio Luteinizante/fisiologia , Luteólise/fisiologia , Masculino , Hipófise/fisiologia , Progesterona/fisiologia , Puberdade/fisiologia , Receptores de Estrogênio/fisiologia , Receptores LHRH/fisiologia , Receptores de Progesterona/fisiologia , Caracteres Sexuais
3.
Brain Res ; 1364: 103-15, 2010 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-20951683

RESUMO

Gonadotropin-releasing hormone (GnRH) secretion has two modes of release in mammalian species; the surge mode and the pulse mode. The surge mode, which is required for the induction of the preovulatory gonadotropin discharge in most species, is induced by the positive feedback of estrogen secreted by the mature ovarian follicle. The pulse mode of GnRH secretion stimulates tonic luteinizing hormone (LH) and follicle-stimulating hormone (FSH) secretion which drives folliculogenesis, spermatogenesis and steroidogenesis and is negatively fine-tuned by estrogen or androgen. The GnRH pulse-generating mechanism is sensitive to environmental cues, such as photoperiod, nutrition and stress surge-generating mechanism is relatively emancipated from these environmental cues. The present article first provides a brief historical background to the work that led to the concept of the GnRH pulse generator: a hypothalamic network that is central to our understanding of the regulation of reproduction. We then discuss possible neurobiological mechanisms underlying GnRH pulse generation, and conclude by proposing that kisspeptin neurons in the arcuate nucleus are key players in this regard.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Hipotálamo/fisiologia , Neurobiologia , Animais , Dinorfinas/fisiologia , Meio Ambiente , Hormônio Foliculoestimulante/fisiologia , Humanos , Kisspeptinas , Hormônio Luteinizante/metabolismo , Neurocinina B/fisiologia , Neurônios/fisiologia , Esteroides/fisiologia , Proteínas Supressoras de Tumor/fisiologia
4.
Anim Reprod Sci ; 109(1-4): 146-60, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18243600

RESUMO

The comparison of hypothalamic somatostatin (SRIH)-neuronal systems, hypophyseal somatotroph populations and growth hormone (GH) blood plasma patterns among developmental stages, from infancy until puberty, may help to describe the nature of the hypothalamo-hypophyseal mechanisms underlying the changes in GH on the systemic level leading to the somatic, that is growth and sexual maturity in sheep. The aim of this study was to elucidate (i) developmental importance of hypophysiotrophic action of SRIH, (ii) precise time of maturation of this action and (iii) photoperiodic regulation of the postnatal ontogeny in ewes. The central and peripheral activity of the SRIH-GH axis is described through a sequence of histomorphological and functional changes in Merino ewes born after the summer solstice. The actual time of puberty of these animals was delayed until the following breeding season, when the sheep were 14-month old. Histomorphometric examinations have been made in 21 infantile (preweanling, 12-week old), prepubertal (15- and 22-week old), peripubertal (30- and 52-week old) and pubertal (63-week old) ovary-intact sheep. Functional examinations of the GH plasma levels were determined every 1-2 weeks during the period from the 12th to 63rd week of age. The highest GH level was observed at the 13th week of age, on the beginning of the breeding season. The fluctuations in the GH level just after the winter and summer solstice were detected as the one and only deviation from a rule of uniformly low GH concentrations observed until puberty. The age of the fall in serum GH levels corresponded with the postweaning period and the beginning of the phase of the lower daily live-weight gains (growth rate). Thus, the development of GH secretion was finished before the 15th week of age, that is together with the ending of the transitional infantile/prepubertal period, whereas the maturational processing within the hypothalamo-hypophyseal unit prolonged after the 15th week of age until 22 weeks of age and concerned the role of SRIH as the hypophysiotrophic factor regulating somatic maturation, i.e. attenuating growth. Altogether, the pattern of GH secretion during weaning is important for the shift between infancy and prepuberty depended upon an intensive growth and defined as growth maturation. The maturation of the SRIH-GH axis is finished by 22 weeks of age, independently of photoperiodic influences, whereas the neuroendocrine mechanisms to integrate somatic, that is growth and sexual maturation, are seasonal in nature in the ewe. Our observations confirm the hypothesis of the inherent endogenous rhythm controlling somatic maturation in the sheep.


Assuntos
Envelhecimento/fisiologia , Encéfalo/fisiologia , Hormônio do Crescimento/fisiologia , Sistema Hipotálamo-Hipofisário/crescimento & desenvolvimento , Sistemas Neurossecretores/fisiologia , Maturidade Sexual/fisiologia , Ovinos/crescimento & desenvolvimento , Somatostatina/fisiologia , Animais , Feminino , Hormônio Foliculoestimulante/fisiologia , Sistema Hipotálamo-Hipofisário/anatomia & histologia , Sistema Hipotálamo-Hipofisário/fisiologia , Hipotálamo/anatomia & histologia , Hipotálamo/crescimento & desenvolvimento , Hipotálamo/fisiologia , Hormônio Luteinizante/fisiologia , Tamanho do Órgão , Fotoperíodo , Polônia , Estações do Ano
5.
Biol Res Nurs ; 9(1): 49-69, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17601857

RESUMO

Dysfunction in menstrual physiology has pronounced effects on quality of life, involving mood changes, body image, infertility, and pregnancy complications. Light exposure may affect menstrual cycles and symptoms through the influence of melatonin secretion. The purpose of this systematic review is to determine the current state of knowledge about the effects of light and melatonin secretion on menstrual phase and cycle alterations. A brief overview of the influence of melatonin on human physiology is included. There is evidence of a relationship between light exposure and melatonin secretion and irregular menstrual cycles, menstrual cycle symptoms, and disordered ovarian function. In women with a psychopathology such as bipolar disorder or an endocrinopathy such as polycystic ovary syndrome, there seems to be greater vulnerability to the influence of light-dark exposure. Research on the complex role of light-dark exposure in menstrual physiology has implications for treatment of menstrual-associated disorders.


Assuntos
Ritmo Circadiano/fisiologia , Luz , Melatonina/fisiologia , Ciclo Menstrual/fisiologia , Transtornos Cronobiológicos/etiologia , Transtornos Cronobiológicos/metabolismo , Transtornos Cronobiológicos/fisiopatologia , Transtornos Cronobiológicos/terapia , Pesquisa em Enfermagem Clínica , Depressão/etiologia , Feminino , Hormônio Foliculoestimulante/fisiologia , Humanos , Luz/efeitos adversos , Hormônio Luteinizante/fisiologia , Distúrbios Menstruais/etiologia , Distúrbios Menstruais/metabolismo , Distúrbios Menstruais/fisiopatologia , Distúrbios Menstruais/terapia , Ovário/fisiologia , Fototerapia/métodos , Glândula Pineal/fisiologia , Hipófise/fisiologia , Prolactina/fisiologia , Projetos de Pesquisa , Fatores de Risco , Estações do Ano
8.
Mol Cell Endocrinol ; 249(1-2): 64-70, 2006 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-16500744

RESUMO

Meiosis activating sterol (MAS) have been found to be able to promote oocytes meiotic maturation of small animals in vitro, such as mouse, rat and rabbit. But in large animals, whether MAS play the same function, especially the physiological mechanisms of MAS on oocytes maturation are not clear. To our knowledge, this is the first time to investigate the role and signal pathway of MAS on FSH-induced porcine oocytes meiotic resumption. Porcine cumulus-enclosed oocytes (CEOs) isolated from 3 to 5mm follicles were cultured in the FSH-medium for 24h supplemented with 0-50 microM RS21745 or 0-100 microM RS21607 (two specific inhibitors of lanosterol 14alpha-demethylase that converts lanosterol to FF-MAS), or cultured in FSH-medium with 25 microM RS21745 for 0-24h firstly, then transferred into a new FSH-medium (the total culture time is 24h). The results revealed that RS21745 or RS21607 could inhibit FSH-induced porcine CEOs meiotic resumption in a dose and time-dependent manner. Meanwhile, FSH-induced cumulus expansion could also be inhibited dose-dependently by RS21745 or RS21607. Otherwise, AY9944-A-7, an inhibitor of Delta14-reductase which promotes cholesterol accumulation from FF-MAS, had no effect on both denuded oocytes (DOs) cultured for 24 or 44 h and CEOs cultured for 24h meiotic resumption, but it could promote CEOs meiotic resumption after 44 h culture. In addition, we got that 10(-8) to 10(-6)M PMA, an activator of PKC pathway, could reverse the inhibiting effect of RS21745 on FSH-induced CEOs meiotic resumption and enhance the rate of germinal vesicle breakdown (GVBD) of CEOs cultured in medium with hypoxanthine (HX). Moreover, 5-10 microM chelerythrine chloride, an inhibitor of PKC, could enhance the inhibitory effect of RS21745 on FSH-induced porcine oocytes resumption of meiosis. All the data of this study support that endogenous FF-MAS takes part in the FSH-induced porcine oocytes meiotic resumption and might play an active role via PKC signal pathway.


Assuntos
Hormônio Foliculoestimulante/fisiologia , Meiose/fisiologia , Oócitos/citologia , Proteína Quinase C/metabolismo , Esteróis/metabolismo , Compostos de Anilina/farmacologia , Animais , Colestadienóis/metabolismo , Colestenos/metabolismo , Inibidores das Enzimas do Citocromo P-450 , Feminino , Oócitos/efeitos dos fármacos , Oócitos/enzimologia , Folículo Ovariano/citologia , Oxirredutases/antagonistas & inibidores , Proteína Quinase C/antagonistas & inibidores , Esterol 14-Desmetilase , Esteróis/antagonistas & inibidores , Sulfetos/farmacologia , Suínos , Dicloridrato de trans-1,4-Bis(2-clorobenzaminometil)ciclo-hexano/farmacologia
9.
Ann N Y Acad Sci ; 1052: 201-9, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16024763

RESUMO

Epidemiological data showing a predisposition of women to develop Alzheimer disease (AD) led many researchers to investigate the role of sex steroids, namely estrogen, in disease pathogenesis. Although there is circumstantial support for the role of estrogen, the unexpected results of the Women's Health Initiative (WHI) Memory Study, which reported an increase in the risk for probable dementia and impaired cognitive performance in postmenopausal women treated with a combination of estrogen and progestin, have raised serious questions regarding the protective effects of estrogen. Although explanations for these surprising results vary greatly, the WHI Memory Study cannot be correctly interpreted without a complete investigation of the effects of the other hormones of the hypothalamic-pituitary-gonadal (HPG) axis on the aging brain. Certain hormones of the HPG axis, namely, the gonadotropins (luteinizing hormone and follicle-stimulating hormone), are not only involved in regulating reproductive function via a complex feedback loop but are also known to cross the blood-brain barrier. We propose that the increase in gonadotropin concentrations, and not the decrease in steroid hormone (e.g., estrogen) production following menopause/andropause, is a potentially primary causative factor for the development of AD. In this review, we examine how the gonadotropins may play a central and determining role in modulating the susceptibility to, and progression of, AD. On this basis, we suggest that the results of the WHI Memory Study are not only predictable but also avoidable by therapeutically targeting the gonadotropins instead of the sex steroids.


Assuntos
Doença de Alzheimer/etiologia , Estrogênios/fisiologia , Gonadotropinas Hipofisárias/fisiologia , Idoso , Envelhecimento , Barreira Hematoencefálica , Encéfalo/fisiopatologia , Terapia de Reposição de Estrogênios/efeitos adversos , Feminino , Hormônio Foliculoestimulante/fisiologia , Humanos , Hipotálamo/fisiopatologia , Hormônio Luteinizante/fisiologia , Memória , Ovário/fisiopatologia , Hipófise/fisiopatologia , Pós-Menopausa , Pré-Menopausa , Saúde da Mulher
10.
Eur J Obstet Gynecol Reprod Biol ; 118(1): 4-16, 2005 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-15596265

RESUMO

This review focuses on the role of the ovaries in the pathogenesis of the polycystic ovarian syndrome. In particular, the failure of follicular development, hypothalamo-pituitary dysregulation, alterations in adrenal steroid output and derangement of intermediary metabolism are discussed in the context of the ovaries. It is concluded that the central and adrenal alterations associated with PCOS are unlikely to be primary but rather are secondary to the events within the ovary.


Assuntos
Glândulas Suprarrenais/fisiopatologia , Homeostase , Hipotálamo/fisiopatologia , Ovário/fisiopatologia , Hipófise/fisiopatologia , Síndrome do Ovário Policístico/fisiopatologia , Feminino , Hormônio Foliculoestimulante/fisiologia , Hormônios Esteroides Gonadais/fisiologia , Humanos , Hiperandrogenismo , Inibinas/fisiologia , Insulina/fisiologia , Fator de Crescimento Insulin-Like I/fisiologia , Hormônio Luteinizante/fisiologia , Obesidade , Folículo Ovariano/fisiopatologia
11.
Eur J Endocrinol ; 147(3): 425-33, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12213681

RESUMO

The importance of non-genomic signaling as a complementary route for cell regulation has recently become evident. This rapid mechanism is utilized not just by peptide hormones, but also by steroids and other lipid-related substances, resulting in amplification and fine-tuning of the signals. The Sertoli cell is the principal target for hormone action in the seminiferous tubules. The involvement of FSH, testosterone and tri-iodothyronine (T(3)) in the spermatogenetic process is widely known. This paper discusses some rapid responses to FSH, retinol, testosterone and T(3) in the control of Sertoli cell function. Studies employing various methodologies and techniques are described and several hypotheses are considered in an attempt to explain the interactions of hormones with plasma membrane receptors. Recent knowledge about these new signaling mechanisms and cross-talk between them opens new fields of research on the communication and integration of the multiple hormonal signaling systems.


Assuntos
Células de Sertoli/metabolismo , Transdução de Sinais , Animais , Calcitriol/fisiologia , Hormônio Foliculoestimulante/fisiologia , Humanos , Masculino , Testosterona/fisiologia , Tri-Iodotironina/fisiologia , Vitamina A/fisiologia
12.
Recent Prog Horm Res ; 57: 149-79, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12017541

RESUMO

A detailed understanding of the hormonal regulation of spermatogenesis is required for the informed assessment and management of male fertility and, conversely, for the development of safe and reversible male hormonal contraception. An approach to the study of these issues is outlined based on the use of well-defined in vivo models of gonadotropin/androgen deprivation and replacement, the quantitative assessment of germ cell number using stereological techniques, and the directed study of specific steps in spermatogenesis shown to be hormone dependent. Drawing together data from rat, monkey, and human models, we identify differences between species and formulate an overview of the hormonal regulation of spermatogenesis. There is good evidence for both separate and synergistic roles for both testosterone and follicle-stimulating hormone (FSH) in achieving quantitatively normal spermatogenesis. Based on relatively selective withdrawal and replacement studies, FSH has key roles in the progression of type A to B spermatogonia and, in synergy with testosterone, in regulating germ cell viability. Testosterone is an absolute requirement for spermatogenesis. In rats, it has been shown to promote the adhesion of round spermatids to Sertoli cells, without which they are sloughed from the epithelium and spermatid elongation fails. The release of mature elongated spermatids from the testis (spermiation) is also under FSH/testosterone control in rats. Data from monkeys and men treated with steroidal contraceptives indicate that impairment of spermiation is a key to achieving azoospermia. The contribution of 5alpha-reduced androgens in the testis to the regulation of spermatogenesis is also relevant, as 5alpha-reduced androgens are maintained during gonadotropin suppression and may act to maintain low levels of germ cell development. These concepts are also discussed in the context of male hormonal contraceptive development.


Assuntos
Homeostase , Hormônios/fisiologia , Espermatogênese , Androgênios/fisiologia , Animais , Hormônio Foliculoestimulante/farmacologia , Hormônio Foliculoestimulante/fisiologia , Hormônio Liberador de Gonadotropina/fisiologia , Haplorrinos , Hormônios/farmacologia , Humanos , Hipotálamo/fisiologia , Masculino , Hipófise/fisiologia , Ratos , Testículo/fisiologia , Testosterona/farmacologia , Testosterona/fisiologia
13.
Endocrinology ; 141(7): 2377-84, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10875237

RESUMO

Given the evident modulation of FSH-induced steroidogenesis by Ca2+ in granulosa cells, we here test the hypothesis that Ca2+ controls expression of the enzymatically rate-limiting cytochrome P450(scc) (CYP11A) gene. To test this postulate, we quantitated the ability of Ca2+ to regulate: 1) transcriptional activity of a transiently transfected luciferase reporter gene driven by a 2.32-kb 5'-upstream fragment of the porcine P450(scc) gene promoter region; and 2) accumulation of endogenous P450(scc) transcripts in primary monolayer cultures of porcine granulosa cells. To this end, granulosa cells were stimulated for 4 h with FSH (15 ng/ml, NIDDK-oFSH-20) or 8-Bromo-cAMP (8 Br-cAMP, 1 mM) in serum-free medium containing either 1.8 mM Ca2- or no added Ca2+ with 100 microM EGTA or 100 microM CoCl2. In the presence of extracellular Ca2+, FSH and 8 Br-cAMP stimulated expression of the transfected P450(scc) promoter-reporter fusion construct by 5.6 +/- 1.1 and 3.6 +/- 0.67-fold, respectively over Ca2+-containing unstimulated control (P < or = 0.04, n = 5-6 experiments). The foregoing two agonists augmented 4-h progesterone production by cultured granulosa cells by 1.8 +/- 0.11 and 1.6 +/- 0.16-fold, respectively (P < or = 0.001 for FSH and P < or = 0.01 for 8 Br-cAMP). FSH and 8 Br-cAMP also significantly elevated endogenous P450(scc) transcript levels as measured by homologous solution-hybridization RNase protection assay; i.e. by 3.1 +/- 0.49 and 2.9 +/- 0.45-fold, respectively (P < or = 0.001). In Ca2+-free/EGTA-supplemented medium, basal luciferase reporter-gene activity and endogenous P450(scc) messenger RNA accumulation in granulosa cells declined to 34 +/- 12% and 78 +/- 12%, respectively, of corresponding values in control (unstimulated Ca2+-containing) cultures. Extracellular Ca2+ deprivation inhibited the stimulatory effect of FSH (and 8 Br-cAMP) on P450(scc) promoter-luciferase reporter expression to 58 +/- 30% (and 58 +/- 23%), and restrained endogenous P450(scc) message accumulation to 86 +/- 15% (and 96 +/- 18%) of the value in Ca2+-containing control. Extracellular Ca2+ withdrawal suppressed FSH (and 8 Br-cAMP)-driven progesterone production over 4 h to basal levels but did not alter FSH-stimulated cAMP accumulation by granulosa cells. Ca2+-deprived cells exposed to serum-containing media regained P450(scc) responsiveness to both agonists. Antagonism of cellular uptake of Ca2+ and other divalent cations via administration of cobalt chloride (100 microM) inhibited FSH and 8 Br-cAMP's stimulation of endogenous (but not exogenous promoter-driven) P450(scc) gene expression. In contrast, granulosa-cell concentrations of messenger RNA's encoding sterol-carrier protein-2 (SCP-2) and the low density lipoprotein receptor were not altered by Ca2+ withdrawal. In summary, uptake of extracellular Ca2+ by porcine granulosa cells significantly potentiates transactivation of the endogenously expressed and exogenously transfected P450(scc) gene by FSH and 8 Br-cAMP. The agonistic impact of Ca2+ on P450(scc) promoter activity is requisite downstream of FSH-induced cAMP second-messenger signaling.


Assuntos
8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Cálcio/fisiologia , Enzima de Clivagem da Cadeia Lateral do Colesterol/genética , Hormônio Foliculoestimulante/fisiologia , Células da Granulosa/fisiologia , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/fisiologia , Animais , Sinalização do Cálcio/fisiologia , Feminino , Íons , Suínos , Transfecção
14.
Fertil Steril ; 72(6): 1093-9, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10593387

RESUMO

OBJECTIVE: To design a drug that blocks the gonadal actions of lutropins and follitropins. DESIGN: Controlled in vitro study. SETTING: Academic laboratory. PATIENT(S): None. INTERVENTION(S): We removed three glycosylation signals from an hCG-hFSH chimera known to have high affinity for LH and FSH receptors, expecting this would create a bifunctional antagonist (dgCFC). To offset the inhibition of subunit combination caused by deglycosylation of alpha-subunit loop 2, we prepared dgCFC as a single-chain fusion protein containing the alpha-subunit downstream of the chimeric beta-subunit. MAIN OUTCOME MEASURE(S): Receptor binding, cyclic adenosine monophosphate accumulation. RESULT(S): dgCFC bound LH or FSH receptors similar to hCG or hFSH. It was a partial agonist and had one tenth the efficacy of hFSH and two thirds the efficacy of hCG. CONCLUSION(S): The surprising high residual lutropin activity of dgCFC indicated that its FSH residues offset the effects of deglycosylation, suggesting this approach to preparing a bifunctional antagonist is unlikely to lead to a useful drug. The increased lutropin efficacy of dgCFC relative to deglycosylated hCG supports the idea that oligosaccharides modulate glycoprotein hormone efficacy through an influence on hormone conformation.


Assuntos
Hormônio Foliculoestimulante/fisiologia , Hormônio Luteinizante/fisiologia , Receptores do FSH/agonistas , Receptores do LH/agonistas , Sequência de Aminoácidos , Animais , Avaliação Pré-Clínica de Medicamentos , Glicosilação , Humanos , Dados de Sequência Molecular , Ratos
16.
Theriogenology ; 51(3): 559-68, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10729042

RESUMO

The aims of this study were to compare steroïdogenesis (progesterone, androstenedione and estradiol production) and response to LH and FSH challenge by whole perifused follicles 4 to 5.5 mm in diameter, obtained at different periods of the breeding season (onset, middle, end), during anestrus and the luteal phase. We have observed that all follicles do not have the same steroïdogenetic potential and do not respond with the same intensity to LH and FSH. At the middle of the breeding season, LH and FSH supplementation was ineffective in increasing progesterone secretion by follicles (0.19+/-0.05 vs. 0.20+/-0.03 ng/mL). In contrast, gonadotrophin challenge elicited significant (P<0.05) increases in androstenedione (0.94+/-0.34 vs. 0.35+/-0.09 ng/mL) and estradiol (120+/-11 vs. 49+/-10 pg/mL) production immediately after its administration. At the onset of the breeding season, steroidogenesis was identical under both basal and gonadotrophin-stimulated conditions unlike that in middle of the breeding season. However follicles were more sensitive to the gonadotrophin challenge in terms of estradiol production than those collected at the middle of the breeding season (220+/-45 vs. 120+/-11 pg/mL). Follicles obtained at the end of the breeding season featured higher progesterone (2.61+/-0.81 vs. 0.19+/-0.05 ng/mL; P<0.05) and lower estradiol production (10+/-3 vs. 49+/-10 pg/mL; P<0.05) that was not influenced by LH and FSH. Basal androstenedione secretion was comparable to that observed at the middle of the breeding season (0.42+/-0.10 vs. 0.35+/-0.09 ng/mL), but the response to stimulation was significantly higher (1.82+/-0.61 vs. 0.94+/-0.34 ng/mL; P<0.05). In anoestrus and the luteal phase, follicles presented higher progesterone and androstenedione and lower estradiol concentrations (P<0.05) compared with those obtained during the follicular phase at the middle of the breeding season. In the luteal phase, follicles remained capable of responding to LH-FSH challenge by increasing estradiol secretion (9+/-1 before and 21+/-6 pg/mL after LH-FSH; P<0.05). In contrast, in the luteal phase, estradiol production was not increased by LH-FSH challenge (7+/-2 vs. 12+/-4 pg/mL).


Assuntos
Androstenodiona/biossíntese , Estradiol/biossíntese , Estro/fisiologia , Hormônio Foliculoestimulante/fisiologia , Hormônio Luteinizante/fisiologia , Folículo Ovariano/fisiologia , Progesterona/biossíntese , Estações do Ano , Ovinos/fisiologia , Animais , Feminino , Técnicas In Vitro , Fase Luteal , Radioimunoensaio/veterinária
17.
Am J Physiol ; 275(6): E1037-45, 1998 12.
Artigo em Inglês | MEDLINE | ID: mdl-9843747

RESUMO

Hypokalemia produced different effects on steroid sex hormone concentrations in plasma and ovary in the mouse. Estradiol levels were slightly increased, whereas circulating progesterone was markedly decreased in all estrous periods. The preovulatory surge of gonadotropins and the secondary surge of follicle-stimulating hormone (FSH) at estrus were also decreased, but basal levels of both gonadotropins were unaffected. Supplementation with luteinizing hormone (LH), FSH, or gonadotropin-releasing hormone (GnRH) at proestrus rapidly normalized plasma and ovarian progesterone levels at this stage of the estrous cycle. Plasma progesterone levels at diestrus were restored only by combined treatment, at the periovulatory stage, with LH and FSH or GnRH but not by LH or FSH alone. The results demonstrate a lack of steroidogenic activity in the corpus luteum of the potassium-deficient mice and, furthermore, that FSH plays an important role in luteinization in the hypokalemic mice. We conclude that alteration of the transcellular potassium gradient may affect the regulation of the periovulatory surge of gonadotropins and progesterone secretion, probably by altering the release of GnRH from the hypothalamus. In addition, the results suggest that FSH may play a certain role as a luteotropic hormone in mice.


Assuntos
Corpo Lúteo/fisiologia , Estradiol/sangue , Hormônio Foliculoestimulante/fisiologia , Gonadotropinas/sangue , Hipopotassemia/sangue , Progesterona/sangue , Animais , Diestro/fisiologia , Combinação de Medicamentos , Estradiol/metabolismo , Estro/fisiologia , Feminino , Hormônio Foliculoestimulante/farmacologia , Fase Folicular/fisiologia , Hormônio Liberador de Gonadotropina/farmacologia , Hormônio Luteinizante/farmacologia , Camundongos , Camundongos Endogâmicos , Ovário/metabolismo , Proestro/fisiologia , Progesterona/metabolismo
18.
Toxicol Sci ; 45(2): 174-87, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9848124

RESUMO

The testicular-hypothalamic-pituitary axis controls reproductive functions in males. A description of the basic physiological interactions in adult rats among testosterone, luteinizing hormone (LH), and follicle stimulating hormone (FSH) was developed, permitting simulation of hormone levels in testes and blood. This model was used to simulate hormone levels in intact, castrate, ethane dimethanesulfonate-treated, and antiandrogen-treated rats. A large gradient of testosterone concentrations from testicular interstitial fluid to low levels in peripheral blood is created by the testicular blood flow. The dominant feedback loop is positive regulation of testosterone synthesis by LH and negative feedback of testosterone on LH and FSH. The utility of the model for placing in vitro data in the context of in vivo physiology was illustrated for the case of continued synthesis of testosterone by the isolated testes. In the absence of blood flow, very low residual testosterone synthesis can substantially increase testosterone concentration in isolated testes. Effects of an exogenous endocrine active compound were illustrated by modeling altered LH and FSH regulation by testosterone in the presence of an antiandrogen acting as a competitive ligand for the androgen receptor. Increasing concentrations have no effect on steady-state hormone levels until sufficient levels of antiandrogen are achieved to reduce negative feedback of testosterone on LH and FSH. In summary, a model has been developed that provides a basis for initiating evaluations of key issues of concern for the risk assessment of endocrine active compounds including in vitro to in vivo extrapolation and their dose-response behaviors.


Assuntos
Hormônio Foliculoestimulante/fisiologia , Sistema Hipotálamo-Hipofisário/fisiologia , Hormônio Luteinizante/fisiologia , Testículo/fisiologia , Testosterona/fisiologia , Animais , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Masculino , Mesilatos/farmacologia , Modelos Biológicos , Ratos , Testículo/efeitos dos fármacos
19.
Mol Reprod Dev ; 50(3): 273-83, 1998 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9621303

RESUMO

Cell-specific expression of the iron-binding protein transferrin is in part mediated through the regulation of its promoter. Although all cells require iron from serum transferrin produced by hepatocytes, cells that create a blood barrier such as Sertoli cells in the testis and choroid plexus epithelium in the brain also express the transferrin gene to provide iron to cells sequestered within the serum-free environment. The current study provides a complete sequence of the 3-kb mouse transferrin promoter and makes a comparison with the sequence available for the human transferrin promoter. Conserved regulatory elements between these two species are identified and speculated to be potentially important response elements for the regulation of the transferrin gene. The proximal 90 bp of the mouse and human transferrin promoter was found to be 80% homologous. The previously identified protected regions in the proximal human promoter also were conserved in the mouse transferrin promoter. Our sequence analysis data revealed that an E-box response element is also conserved between mouse and human promoters. Deletion mutants of the mouse transferrin promoter were generated in CAT reporter constructs to study the regulation of the transferrin promoter in Sertoli cells. As in the case of the human transferrin promoter, the mouse 581-bp proximal transferrin promoter was sufficient to obtain basal expression. A putative cyclic AMP response element (CRE) in the minimal promoter may be needed for follicle-stimulating hormone (FSH) actions mediated via cyclic AMP. Interestingly, other regulatory agents such as the testicular paracrine factor PModS used elements in the upstream region. A repressor was identified 2.5 kb upstream from the start site of translation. Combined observations suggest for the first time that a minimal promoter is sufficient for basal transcription, but the upstream regions of the promoter are needed for the hormonal regulation of the transferrin gene in Sertoli cells. Conserved response elements between the mouse and human sequences identify potentially important regulatory elements of the promoter and are discussed.


Assuntos
Hormônio Foliculoestimulante/fisiologia , Regiões Promotoras Genéticas , Células de Sertoli/metabolismo , Transferrina/genética , Animais , Sequência de Bases , Células Cultivadas , DNA Complementar , Regulação da Expressão Gênica , Humanos , Masculino , Camundongos , Dados de Sequência Molecular , Ratos , Análise de Sequência de DNA , Células de Sertoli/efeitos dos fármacos
20.
Proc Natl Acad Sci U S A ; 94(17): 9499-503, 1997 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-9256511

RESUMO

Previous studies indicated that there is a separate hypothalamic control of follicle-stimulating hormone (FSH) release distinct from that of luteinizing hormone (LH). An FSH-releasing factor (FSHRF) was purified from rat and sheep hypothalami, but has not been isolated. We hypothesized that FSHRF might be an analogue of mammalian luteinizing hormone-releasing hormone (m-LHRH) and evaluated the activity of many analogues of m-LHRH and of the known LHRHs found in lower forms. Here we demonstrate that lamprey (l) LHRH-III has a potent, dose-related FSH- but not LH-releasing action on incubated hemipituitaries of male rats. l-LHRH-I on the other hand, had little activity to release either FSH or LH. m-LHRH was equipotent to l-LHRH-III to release FSH, but also had a high potency to release LH in contrast to l-LHRH-III that selectively released FSH. Chicken LHRH-II had considerable potency to release both LH and FSH, but no selectivity in its action. Salmon LHRH had much less potency than the others tested, except for l-LHRH-I, and no selectivity in its action. Because ovariectomized, estrogen, progesterone-treated rats are a sensitive in vivo assay for FSH- and LH-releasing activity, we evaluated l-LHRH-III in this assay and found that it had a completely selective stimulatory effect on FSH release at the two doses tested (10 and 100 pmols). Therefore, l-LHRH-III is a highly potent and specific FSH-releasing peptide that may enhance fertility in animals and humans. It may be the long sought after m-FSHRF.


Assuntos
Hormônio Foliculoestimulante/fisiologia , Hipotálamo/fisiologia , Peptídeos/fisiologia , Animais , Feminino , Hormônio Foliculoestimulante/metabolismo , Hormônio Liberador de Gonadotropina/fisiologia , Masculino , Proteínas do Tecido Nervoso/isolamento & purificação , Proteínas do Tecido Nervoso/fisiologia , Peptídeos/isolamento & purificação , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA