Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 11(1): 19796, 2021 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-34611283

RESUMO

We previously reported that growth promoter-induced skeletal muscle hypertrophy co-ordinately upregulated expression of genes associated with an integrated stress response (ISR), as well as potential ISR regulators. We therefore used Adeno-Associated Virus (AAV)-mediated overexpression of these genes, individually or in combination, in mouse skeletal muscle to test whether they induced muscle hypertrophy. AAV of each target gene was injected into mouse Tibialis anterior (TA) and effects on skeletal muscle growth determined 28 days later. Individually, AAV constructs for Arginase-2 (Arg2) and Activating transcription factor-5 (Atf5) reduced hindlimb muscle weights and upregulated expression of genes associated with an ISR. AAV-Atf5 also decreased Myosin heavy chain (MyHC)-IIB mRNA, but increased MyHC-IIA and isocitrate dehydrogenase-2 (Idh2) mRNA, suggesting ATF5 is a novel transcriptional regulator of Idh2. AAV-Atf5 reduced the size of both TA oxidative and glycolytic fibres, without affecting fibre-type proportions, whereas Atf5 combined with Cebpg (CCAAT enhancer binding protein-gamma) only reduced the size of glycolytic fibres and tended to increase the proportion of oxidative fibres. It is likely that persistent Atf5 overexpression maintains activation of the ISR, thereby reducing protein synthesis and/or increasing protein degradation and possibly apoptosis, resulting in inhibition of muscle growth, with overexpression of Arg2 having a similar effect.


Assuntos
Fatores Ativadores da Transcrição/genética , Dependovirus/genética , Expressão Gênica , Vetores Genéticos/genética , Músculo Esquelético/metabolismo , Estresse Fisiológico , Transdução Genética , Fatores Ativadores da Transcrição/metabolismo , Animais , Metabolismo Energético , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Camundongos , RNA Mensageiro/genética
2.
PLoS One ; 14(6): e0218970, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31237922

RESUMO

We previously identified PEPCK-M (encoded by the Pck2 gene) to be highly up-regulated in skeletal muscle of pigs treated with Ractopamine, an anabolic beta-adrenergic receptor agonist. To determine whether PEPCK-M had a causative role in modulating the skeletal muscle growth response to Ractopamine, we used adeno-associated virus 1 (AAV1) to over-express Pck2 (AAV-Pck2) in murine skeletal muscle. A contralateral limb design was employed, such that each mouse served as its own control (injected with a GFP-only expressing AAV1, labelled AAV-GFP). Daily injections of Clenbuterol (1 mg/kg for 21 days) or vehicle control were also carried out to assess the effects of AAV-Pck2 overexpression on the anabolic response to a beta-adrenergic agonist. AAV-Pck2 overexpression in leg muscles of male C57BL6/J mice for 4 weeks (6-10 weeks of age) increased Pck2 mRNA (~100-fold), protein (not quantifiable) and enzyme activity (~3-fold). There was a trend (p = 0.0798) for AAV-Pck2 overexpression to reduce TA muscle weights, but there was no significant effect on muscle fibre diameters or myosin heavy chain isoform (MyHC) mRNA expression. When skeletal muscle growth was induced by daily administration of Clenbuterol (for 21 days), overexpression of AAV-Pck2 had no effect on the growth response, nor did it alter the expression of Phosphoserine Aminotransferase-1 (Psat1) or Asparagine Synthetase (Asns) mRNA or the Clenbuterol-induced decreases in MyHC IIa and IIx mRNA expression (p = 0.0065 and p = 0.0267 respectively). However AAV-Pck2 overexpression reduced TA muscle weights (p = 0.0434), particularly in the Control (vehicle treated) mice (p = 0.059 for AAV x Clenbuterol interaction) and increased the expression of Seryl-tRNA Synthetase (Sars) mRNA (p = 0.0477). Hence, contrary to the original hypothesis, AAV-Pck2 overexpression reduced TA muscle weights and did not mimic or alter the muscle hypertrophic effects of the beta-adrenergic agonist, Clenbuterol.


Assuntos
Clembuterol/farmacologia , Dependovirus/metabolismo , Desenvolvimento Muscular/efeitos dos fármacos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Fosfoenolpiruvato Carboxiquinase (ATP)/metabolismo , Agonistas Adrenérgicos beta/farmacologia , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Cadeias Pesadas de Miosina/efeitos dos fármacos , Cadeias Pesadas de Miosina/metabolismo , Fenetilaminas/farmacologia , Isoformas de Proteínas/metabolismo
3.
Exp Physiol ; 103(6): 876-883, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29663541

RESUMO

NEW FINDINGS: What is the central question of this study? The role of FGF21 as an exercise-induced myokine remains controversial. The aim of this study was to determine whether eccentric exercise would augment the release of FGF21 and/or its regulatory enzyme, fibroblast activation protein α (FAP), from skeletal muscle tissue into the systemic circulation of healthy human volunteers. What is the main finding and its importance? Eccentric exercise does not release total or bioactive FGF21 from human skeletal muscle. However, exercise releases its regulatory enzyme, FAP, from tissue(s) other than muscle, which might play a role in the inactivation of FGF21. ABSTRACT: The primary aim of the investigation was to determine whether eccentric exercise would augment the release of the myokine fibroblast growth factor 21 (FGF21) and/or its regulatory enzyme, fibroblast activation protein α (FAP), from skeletal muscle tissue into the systemic circulation of healthy human volunteers. Physically active young healthy male volunteers (age 25.0 ± 10.7 years; body mass index 23.1 ± 7.9 kg m-2 ) completed three sets of 25 repetitions (with 5 min rest in between) of single-leg maximal eccentric contractions using their non-dominant leg, whilst the dominant leg served as a control. Arterialized blood samples from a hand vein and deep venous blood samples from the common femoral vein of the exercised leg, along with blood flow of the superficial femoral artery using Doppler ultrasound, were obtained before and after each exercise bout and every 20 min during the 3 h recovery period. Muscle biopsy samples were taken at baseline, immediately and 3 and 48 h postexercise. The main findings showed that there was no significant increase in total or bioactive FGF21 secreted from skeletal muscle into the systemic circulation in response to exercise. Furthermore, skeletal muscle FGF21 protein content was unchanged in response to exercise. However, there was a significant increase in arterialized and venous FAP concentrations, with no apparent contribution to its release from the exercised leg. These findings raise the possibility that the elevated levels of FAP might play a role in the inactivation of FGF21 during exercise.


Assuntos
Exercício Físico/fisiologia , Fatores de Crescimento de Fibroblastos/sangue , Gelatinases/sangue , Proteínas de Membrana/sangue , Serina Endopeptidases/sangue , Adulto , Endopeptidases , Humanos , Masculino , Proteínas Musculares/sangue , Músculo Esquelético/metabolismo , Fluxo Sanguíneo Regional/fisiologia , Descanso/fisiologia
4.
PLoS One ; 12(2): e0172724, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28235047

RESUMO

VGF (non-acronymic) was first highlighted to have a role in energy homeostasis through experiments involving dietary manipulation in mice. Fasting increased VGF mRNA in the Arc and levels were subsequently reduced upon refeeding. This anabolic role for VGF was supported by observations in a VGF null (VGF-/-) mouse and in the diet-induced and gold-thioglucose obese mice. However, this anabolic role for VGF has not been supported by a number of subsequent studies investigating the physiological effects of VGF-derived peptides. Intracerebroventricular (ICV) infusion of TLQP-21 increased resting energy expenditure and rectal temperature in mice and protected against diet-induced obesity. Similarly, ICV infusion of TLQP-21 into Siberian hamsters significantly reduced body weight, but this was due to a decrease in food intake, with no effect on energy expenditure. Subsequently NERP-2 was shown to increase food intake in rats via the orexin system, suggesting opposing roles for these VGF-derived peptides. Thus to further elucidate the role of hypothalamic VGF in the regulation of energy homeostasis we utilised a recombinant adeno-associated viral vector to over-express VGF in adult male Siberian hamsters, thus avoiding any developmental effects or associated functional compensation. Initially, hypothalamic over-expression of VGF in adult Siberian hamsters produced no effect on metabolic parameters, but by 12 weeks post-infusion hamsters had increased oxygen consumption and a tendency to increased carbon dioxide production; this attenuated body weight gain, reduced interscapular white adipose tissue and resulted in a compensatory increase in food intake. These observed changes in energy expenditure and food intake were associated with an increase in the hypothalamic contents of the VGF-derived peptides AQEE, TLQP and NERP-2. The complex phenotype of the VGF-/- mice is a likely consequence of global ablation of the gene and its derived peptides during development, as well as in the adult.


Assuntos
Peso Corporal/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Neuropeptídeos/biossíntese , Obesidade/tratamento farmacológico , Aumento de Peso/efeitos dos fármacos , Animais , Peso Corporal/fisiologia , Cricetinae , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Camundongos , Camundongos Obesos , Proteínas do Tecido Nervoso/administração & dosagem , Neuropeptídeos/administração & dosagem , Neuropeptídeos/genética , Obesidade/genética , Obesidade/metabolismo , Consumo de Oxigênio/efeitos dos fármacos , Fragmentos de Peptídeos/administração & dosagem , Phodopus , Ratos , Aumento de Peso/fisiologia
5.
J Mol Endocrinol ; 56(2): 123-34, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26643910

RESUMO

The Siberian hamster (Phodopus sungorus) survives winter by decreasing food intake and catabolizing abdominal fat reserves, resulting in a sustained, profound loss of body weight. Hypothalamic tanycytes are pivotal for this process. In these cells, short-winter photoperiods upregulate deiodinase 3, an enzyme that regulates thyroid hormone availability, and downregulate genes encoding components of retinoic acid (RA) uptake and signaling. The aim of the current studies was to identify mechanisms by which seasonal changes in thyroid hormone and RA signaling from tanycytes might ultimately regulate appetite and energy expenditure. proVGF is one of the most abundant peptides in the mammalian brain, and studies have suggested a role for VGF-derived peptides in the photoperiodic regulation of body weight in the Siberian hamster. In silico studies identified possible thyroid and vitamin D response elements in the VGF promoter. Using the human neuroblastoma SH-SY5Y cell line, we demonstrate that RA increases endogenous VGF expression (P<0.05) and VGF promoter activity (P<0.0001). Similarly, treatment with 1,25-dihydroxyvitamin D3 increased endogenous VGF mRNA expression (P<0.05) and VGF promoter activity (P<0.0001), whereas triiodothyronine (T3) decreased both (P<0.01 and P<0.0001). Finally, intra-hypothalamic administration of T3 blocked the short day-induced increase in VGF expression in the dorsomedial posterior arcuate nucleus of Siberian hamsters. Thus, we conclude that VGF expression is a likely target of photoperiod-induced changes in tanycyte-derived signals and is potentially a regulator of seasonal changes in appetite and energy expenditure.


Assuntos
Calcitriol/farmacologia , Fatores de Crescimento Neural/metabolismo , Ativação Transcricional , Tri-Iodotironina/fisiologia , Animais , Diferenciação Celular , Linhagem Celular Tumoral , Proliferação de Células , Cricetinae , Expressão Gênica , Humanos , Masculino , Fatores de Crescimento Neural/genética , Phodopus , Regiões Promotoras Genéticas , Tri-Iodotironina/farmacologia
6.
J Neurosci Methods ; 256: 22-9, 2015 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-26300182

RESUMO

INTRODUCTION: The viral 2A sequence has become an attractive alternative to the traditional internal ribosomal entry site (IRES) for simultaneous over-expression of two genes and in combination with recombinant adeno-associated viruses (rAAV) has been used to manipulate gene expression in vitro. NEW METHOD: To develop a rAAV construct in combination with the viral 2A sequence to allow long-term over-expression of the vgf gene and fluorescent marker gene for tracking of the transfected neurones in vivo. RESULTS: Transient transfection of the AAV plasmid containing the vgf gene, viral 2A sequence and eGFP into SH-SY5Y cells resulted in eGFP fluorescence comparable to a commercially available reporter construct. This increase in fluorescent cells was accompanied by an increase in VGF mRNA expression. Infusion of the rAAV vector containing the vgf gene, viral 2A sequence and eGFP resulted in eGFP fluorescence in the hypothalamus of both mice and Siberian hamsters, 32 weeks post infusion. In situ hybridisation confirmed that the location of VGF mRNA expression in the hypothalamus corresponded to the eGFP pattern of fluorescence. COMPARISON WITH OLD METHOD: The viral 2A sequence is much smaller than the traditional IRES and therefore allowed over-expression of the vgf gene with fluorescent tracking without compromising viral capacity. CONCLUSION: The use of the viral 2A sequence in the AAV plasmid allowed the simultaneous expression of both genes in vitro. When used in combination with rAAV it resulted in long-term over-expression of both genes at equivalent locations in the hypothalamus of both Siberian hamsters and mice, without any adverse effects.


Assuntos
Dependovirus/genética , Técnicas Genéticas , Vetores Genéticos , Proteínas de Fluorescência Verde/metabolismo , Neuropeptídeos/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular Tumoral , Sequência Consenso , DNA Recombinante , DNA Viral , Proteínas de Fluorescência Verde/genética , Humanos , Hipotálamo/metabolismo , Masculino , Mesocricetus , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Fatores de Crescimento Neural , Neuropeptídeos/genética , RNA Mensageiro/metabolismo
7.
J Mol Endocrinol ; 54(3): 241-50, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25878058

RESUMO

Tanycytes play multiple roles in hypothalamic functions, including sensing peripheral nutrients and metabolic hormones, regulating neurosecretion and mediating seasonal cycles of reproduction and metabolic physiology. This last function reflects the expression of TSH receptors in tanycytes, which detect photoperiod-regulated changes in TSH secretion from the neighbouring pars tuberalis. The present overall aim was to determine the signal transduction pathway by which TSH signals in tanycytes. Expression of the TSH receptor in tanycytes of 10-day-old Sprague Dawley rats was observed by in situ hybridisation. Primary ependymal cell cultures prepared from 10-day-old rats were found by immunohistochemistry to express vimentin but not GFAP and by PCR to express mRNA for Dio2, Gpr50, Darpp-32 and Tsh receptors that are characteristic of tanycytes. Treatment of primary tanycyte/ependymal cultures with TSH (100  IU/l) increased cAMP as assessed by ELISA and induced a cAMP-independent increase in the phosphorylation of ERK1/2 as assessed by western blot analysis. Furthermore, TSH (100  IU/l) stimulated a 2.17-fold increase in Dio2 mRNA expression. We conclude that TSH signal transduction in cultured tanycytes signals via Gαs to increase cAMP and via an alternative G protein to increase phosphorylation of ERK1/2.


Assuntos
Células Ependimogliais/metabolismo , Receptores da Tireotropina/metabolismo , Tireotropina/fisiologia , Animais , AMP Cíclico/metabolismo , Feminino , Iodeto Peroxidase/metabolismo , Sistema de Sinalização das MAP Quinases , Masculino , Fosforilação , Cultura Primária de Células , Processamento de Proteína Pós-Traducional , Ratos Sprague-Dawley , Sistemas do Segundo Mensageiro , Iodotironina Desiodinase Tipo II
8.
Artigo em Inglês | MEDLINE | ID: mdl-24616714

RESUMO

Living organisms show seasonality in a wide array of functions such as reproduction, fattening, hibernation, and migration. At temperate latitudes, changes in photoperiod maintain the alignment of annual rhythms with predictable changes in the environment. The appropriate physiological response to changing photoperiod in mammals requires retinal detection of light and pineal secretion of melatonin, but extraretinal detection of light occurs in birds. A common mechanism across all vertebrates is that these photoperiod-regulated systems alter hypothalamic thyroid hormone (TH) conversion. Here, we review the evidence that a circadian clock within the pars tuberalis of the adenohypophysis links photoperiod decoding to local changes of TH signaling within the medio-basal hypothalamus (MBH) through a conserved thyrotropin/deiodinase axis. We also focus on recent findings which indicate that, beyond the photoperiodic control of its conversion, TH might also be involved in longer-term timing processes of seasonal programs. Finally, we examine the potential implication of kisspeptin and RFRP3, two RF-amide peptides expressed within the MBH, in seasonal rhythmicity.

9.
Anat Sci Int ; 85(4): 214-23, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20422334

RESUMO

Electrophysiological recordings and transmission electron microscopy were used to characterize the compound action potential (CAP) and morphology, respectively, of the optic nerve in the Siberian hamster. The CAP was polyphasic in nature, comprising four separate but overlapping peaks, thereby implying that four sub-populations of axons defined by conduction velocity are present in the nerve. The histological analysis of nerves from four animals revealed a cross-sectional area of 128,171 µm(2) containing 78,109 axons. All of the axons were myelinated, and measurements of axon surface area revealed values ranging from 0.09 to 9.92 µm(2), although 68.3% were <1 µm(2). In the regions of the nerve sampled, the area occupied by axons varied from 10.2 to 80.1%, but in 72.5% of these regions the axons occupied between 50 and 70% of the total cross-sectional area. All regions of the nerve expressed small axons, but larger axons (>2.5 µm(2)) were selectively distributed throughout the nerve. We conclude that the CAP recorded from hamster optic nerve displays four distinct peaks; however, morphological analysis failed to reveal a similar distribution of axon sizes.


Assuntos
Potenciais de Ação , Nervo Óptico/fisiologia , Nervo Óptico/ultraestrutura , Animais , Axônios/ultraestrutura , Contagem de Células , Tamanho Celular , Cricetinae , Potenciais Evocados , Microscopia Eletrônica de Transmissão , Nervo Óptico/citologia , Phodopus
10.
Am J Physiol Regul Integr Comp Physiol ; 298(5): R1409-16, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20200136

RESUMO

The objective of this study was to determine whether the previously observed effects of photoperiod on body weight in Siberian hamsters were due to changes in the daily patterns of locomotor activity, energy expenditure, and/or feeding behavior. Adult males were monitored through a seasonal cycle using an automated comprehensive laboratory animal monitoring system (CLAMS). Exposure to a short-day photoperiod (SD; 8:16-h light-dark cycle) induced a significant decline in body weight, and oxygen consumption (Vo(2)), carbon dioxide production (Vco(2)), and heat production all decreased reaching a nadir by 16 wk of SD. Clear daily rhythms in locomotor activity, Vo(2), and Vco(2) were observed at the start of the study, but these all progressively diminished after prolonged exposure to SD. Rhythms in feeding behavior were also detected initially, reflecting an increase in meal frequency but not duration during the dark phase. This rhythm was lost by 8 wk of SD exposure such that food intake was relatively constant across dark and light phases. After 18 wk in SD, hamsters were transferred to a long-day photoperiod (LD; 16:8-h light-dark cycle), which induced significant weight gain. This was associated with an increase in energy intake within 2 wk, while Vo(2), Vco(2), and heat production all increased back to basal levels. Rhythmicity was reestablished within 4 wk of reexposure to long days. These results demonstrate that photoperiod impacts on body weight via complex changes in locomotor activity, energy expenditure, and feeding behavior, with a striking loss of daily rhythms during SD exposure.


Assuntos
Metabolismo Energético/fisiologia , Comportamento Alimentar/fisiologia , Atividade Motora/fisiologia , Phodopus/fisiologia , Fotoperíodo , Estações do Ano , Animais , Regulação da Temperatura Corporal/fisiologia , Peso Corporal/fisiologia , Dióxido de Carbono/metabolismo , Cricetinae , Cabelo/fisiologia , Masculino , Mamíferos , Consumo de Oxigênio/fisiologia
11.
PLoS One ; 4(4): e5322, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19390688

RESUMO

BACKGROUND: Secretion of gonadotropin-releasing hormone (GnRH) produced in neurons in the basal forebrain is the primary regulator of reproductive maturation and function in mammals. Peptidergic signals relating to circadian timing and energy balance are an important influence on the reproductive axis. The aim of this study was to investigate the innervation of GnRH neurons by peptidergic neurons. METHODOLOGY/PRINCIPAL FINDINGS: Immunohistochemistry and confocal microscopy were used to detect appositions of peptidergic fibers (NPY, beta-endorphin, MCH) associated with energy balance and metabolic status in transgenic mice expressing a green fluorescent protein reporter construct in GnRH neurons. The frequency of these appositions was compared to those of vasoactive intestinal peptide (VIP), a hypothalamic neuropeptide likely to convey circadian timing information to the GnRH secretory system. The majority of GnRH neurons (73-87%) were closely apposed by fibers expressing NPY, beta-endorphin, or MCH, and a significant proportion of GnRH neurons (28%) also had close contacts with VIP-ir fibers. CONCLUSIONS/SIGNIFICANCE: It is concluded that GnRH neurons in the mouse receive a high frequency of direct modulatory inputs from multiple hypothalamic peptide systems known to be important in conveying circadian information and signalling energy balance.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/metabolismo , Neuropeptídeos/análise , Animais , Ritmo Circadiano , Proteínas de Fluorescência Verde/metabolismo , Hormônios Hipotalâmicos/metabolismo , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos , Microscopia Confocal , Vias Neurais , beta-Endorfina/análise
12.
Am J Physiol Regul Integr Comp Physiol ; 294(6): R1968-79, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18417646

RESUMO

The genes encoding prokineticin 2 polypeptide (Prok2) and its cognate receptor (Prokr2/Gpcr73l1) are widely expressed in both the suprachiasmatic nucleus and its hypothalamic targets, and this signaling pathway has been implicated in the circadian regulation of behavior and physiology. We have previously observed that the targeted null mutation of Prokr2 disrupts circadian coordination of cycles of locomotor activity and thermoregulation. We have now observed spontaneous but sporadic bouts of torpor in the majority of these transgenic mice lacking Prokr2 signaling. During these torpor bouts, which lasted for up to 8 h, body temperature and locomotor activity decreased markedly. Oxygen consumption and carbon dioxide production also decreased, and there was a decrease in respiratory quotient. These spontaneous torpor bouts generally began toward the end of the dark phase or in the early light phase when the mice were maintained on a 12:12-h light-dark cycle and persisted when mice were exposed to continuous darkness. Periods of food deprivation (16-24 h) induced a substantial decrease in body temperature in all mice, but the duration and depth of hypothermia was significantly greater in mice lacking Prokr2 signaling compared with heterozygous and wild-type littermates. Likewise, when tested in metabolic cages, food deprivation produced greater decreases in oxygen consumption and carbon dioxide production in the transgenic mice than controls. We conclude that Prokr2 signaling plays a role in hypothalamic regulation of energy balance, and loss of this pathway results in physiological and behavioral responses normally only detected when mice are in negative energy balance.


Assuntos
Comportamento Animal/fisiologia , Predisposição Genética para Doença , Hibernação/genética , Mutação/genética , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais/genética , Animais , Temperatura Corporal/fisiologia , Peso Corporal/fisiologia , Dióxido de Carbono/metabolismo , Ritmo Circadiano/fisiologia , Ingestão de Energia/fisiologia , Metabolismo Energético/fisiologia , Feminino , Hibernação/fisiologia , Masculino , Camundongos , Camundongos Transgênicos , Consumo de Oxigênio/fisiologia , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/fisiologia
13.
Neuroendocrinology ; 88(2): 80-7, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18408361

RESUMO

VGF is a 68-kDa polypeptide synthesized in neuronal and neuroendocrine cells. It is cleaved into a number of smaller peptides which are stored in dense core vesicles and are likely to be secreted products. The VGF gene is expressed abundantly in the brain, and in peripheral endocrine tissues including the pituitary gland, the adrenal glands and the pancreas but also in the gastrointestinal tract in both the myenteric plexus and in endocrine cells. Several lines of evidence including observation of changes in hypothalamic VGF expression in catabolic states, a hypermetabolic phenotype in transgenic mice lacking VGF signalling, and demonstration of bioactivity of various peptide fragments have led to the view that VGF and/or its derived peptides are involved in the regulation of both energy balance and reproduction.


Assuntos
Regulação do Apetite/genética , Peso Corporal/genética , Neuropeptídeos/fisiologia , Reprodução/genética , Animais , Peso Corporal/fisiologia , Ritmo Circadiano/genética , Ritmo Circadiano/fisiologia , Ingestão de Alimentos/genética , Ingestão de Alimentos/fisiologia , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/genética , Humanos , Camundongos , Camundongos Transgênicos , Fatores de Crescimento Neural , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Neuropeptídeos/farmacologia , Reprodução/fisiologia , Roedores , Estações do Ano , Distribuição Tecidual
14.
Hum Fertil (Camb) ; 9(3): 127-35, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17008264

RESUMO

The hypogonadal (hpg) mouse is an excellent animal model in which to investigate the mechanism of action of estrogens on spermatogenesis because it has arrested reproductive development without the need for surgical, endocrine, pharmacological or immunological intervention. Hpg mice are hypogonadotrophic and fail to show normal postnatal testicular development due to the congenital inability to synthesize gonadotropin-releasing hormone in the hypothalamus. The hpg testis remains responsive to gonadotropins and androgens in that fertility can be induced by treatment with these hormones. Surprisingly, chronic treatment with low concentrations of estradiol alone induces qualitatively normal spermatogenesis. The induction of testicular development by estradiol in hpg mice is accompanied by a paradoxical increase in FSH production. The actions of estradiol in hpg mice appear to be via genomic estrogen receptors, as concurrent treatment with estrogen-receptor antagonist ICI182,780 completely blocks these pituitary and testis responses. Concurrent treatment with the androgen receptor antagonist bicalutamide does not affect the estradiol-induced increase in pituitary FSH content, but markedly attenuates the estradiol-induced increase in testicular weight. Western blot analyses and immunohistochemistry provide evidence for estrogen-receptor alpha and beta expression in both pituitary gland and testis of the hpg mouse. Estradiol may therefore exert direct actions within the testes and/or indirect neuroendocrine actions via the release of FSH or other hormones from the pituitary gland, but its actions are dependent upon the availability of low levels of androgen within the testis.


Assuntos
Estrogênios/fisiologia , Hipogonadismo , Modelos Animais , Espermatogênese/fisiologia , Animais , Estradiol/farmacologia , Hormônio Foliculoestimulante/biossíntese , Hormônio Foliculoestimulante/metabolismo , Humanos , Hipogonadismo/genética , Masculino , Camundongos , Camundongos Mutantes , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , Receptores de Estrogênio/fisiologia , Células de Sertoli/fisiologia , Espermatogênese/efeitos dos fármacos , Testículo/efeitos dos fármacos , Testículo/crescimento & desenvolvimento
15.
Reproduction ; 130(5): 643-54, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16264094

RESUMO

Testicular development is arrested in the hypogonadal (hpg) mouse due to a congenital deficiency of hypothalamic gonadotropin-releasing hormone synthesis. Previous studies have demonstrated that chronic treatment of these mice with estradiol induces testicular maturation and qualitatively normal spermatogenesis, but it is not known whether these are direct effects via estrogen receptors expressed in the testis, or indirect actions via the pituitary gland. The aim of the current studies was to determine whether the actions of estradiol require the presence of androgens. Sensitive assays revealed that chronic estradiol treatment produced time-dependent increases in pituitary FSH production but no increases in pituitary LH or testicular testosterone content could be detected. As a functional test of androgen dependence, hpg mice were treated for 70 days with estradiol plus Casodex (bicalutamide), an androgen receptor antagonist. Casodex treatment markedly attenuated both the estradiol-induced increase in testicular weight and the proliferation of the seminiferous epithelium, as revealed by morphometric analysis. However, it did not affect the estradiol-induced increase in pituitary FSH content, nor did it affect estradiol-induced increases in the weight of the seminal vesicles and epididymides. We conclude that increased FSH production is not sufficient to explain the increase in testicular development induced by estradiol in hpg mice; there is a requirement for functional androgen receptors for induction of testicular growth.


Assuntos
Androgênios/metabolismo , Estradiol/farmacologia , Hipogonadismo/metabolismo , Espermatogênese/efeitos dos fármacos , Testículo/metabolismo , Antagonistas de Androgênios/farmacologia , Anilidas/farmacologia , Animais , Estradiol/análogos & derivados , Feminino , Hormônio Foliculoestimulante/sangue , Fulvestranto , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Mutantes , Nitrilas , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , Receptores Androgênicos/metabolismo , Epitélio Seminífero/efeitos dos fármacos , Epitélio Seminífero/crescimento & desenvolvimento , Epitélio Seminífero/metabolismo , Testículo/efeitos dos fármacos , Testículo/crescimento & desenvolvimento , Compostos de Tosil , Útero/efeitos dos fármacos
16.
Reprod Biol Endocrinol ; 3: 48, 2005 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-16176578

RESUMO

BACKGROUND: Testicular development is arrested in the hypogonadal (hpg) mouse due to a congenital deficiency in hypothalamic gonadotropin-releasing hormone (GnRH) synthesis. Chronic treatment of male hpg mice with estradiol induces FSH synthesis and secretion, and causes testicular maturation and qualitatively normal spermatogenesis. As estradiol negative feedback normally inhibits FSH production in the male, this study tested whether this paradoxical response to estradiol in the male hpg mouse might be due to inadequate masculinisation or incomplete defeminization in the neonatal period. Previous studies have demonstrated that treatment of hpg mice with testosterone propionate in the immediate neonatal period is necessary to allow full reproductive behaviors to be expressed following suitable endocrine stimulation at adult ages. METHODS: Hpg mice were treated with 100 mug testosterone propionate or vehicle on postnatal day 2. At 35 days of age, subgroups of these mice were treated with silastic implants containing estradiol or cholesterol. Reproductive behavior was scored in tests with steroid-primed female mice, then testicular development was assessed histologically, and measures of pituitary FSH content made at 85 days of age. RESULTS: The neonatal testosterone propionate treatment successfully defeminized female litter mates, as revealed by impaired vaginal opening and deficiencies in lordosis behavior, and it allowed appropriate male reproductive behavior to be expressed in a proportion of the hpg males when tested at an adult age. However, neonatal androgen supplementation did not block or even reduce the subsequent actions of estradiol in increasing pituitary FSH content, nor did it affect the ability of estradiol to induce qualitatively normal spermatogenesis. CONCLUSION: The ability of the hpg male to show a "female" neuroendocrine response to estradiol is not a result of inadequate androgenization during neonatal development, and thus the actions of estradiol revealed in this rodent model are not an artefact of incomplete sexual differentiation, but reflect a physiological role of estradiol occurring during a specific early temporal window of male reproductive development.


Assuntos
Androgênios/farmacologia , Estradiol/farmacologia , Hormônio Foliculoestimulante/biossíntese , Hipogonadismo/fisiopatologia , Espermatogênese/efeitos dos fármacos , Propionato de Testosterona/farmacologia , Animais , Animais Recém-Nascidos , Colesterol/farmacologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Mutantes , Tamanho do Órgão/efeitos dos fármacos , Comportamento Sexual Animal , Testículo/anatomia & histologia , Testículo/efeitos dos fármacos
17.
Reproduction ; 129(6): 675-83, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15923383

RESUMO

Puberty is the attainment of fertility, a process encompassing morphological, physiological and behavioural development. The increased hypothalamic secretion of the gonadotrophin-releasing hormone decapeptide (GnRH) is essential for the activation of the pituitary-gonadal axis at puberty. The GnRH secretory network initially develops and is temporarily active during species-specific periods of fetal/neonatal development, so puberty is the secondary reactivation of an existing system. From a neurobiological perspective, the timing of puberty is therefore a function of changes in the neural systems controlling GnRH release. The large variability between individuals in the onset and progression of puberty indicates that the timing of puberty is not simply a function of chronological age. Rather, the neurotransmitter and neuromodulatory systems that impact upon the GnRH secretory network convey information about metabolic fuels, energy stores and somatic development and, for many species, information about season and social environment. The clear links demonstrated between metabolic fuel availability and reproductive function in many animal models provides evidence that the earlier onset of pubertal development observed in girls in certain US study populations is likely to relate to the increasing prevalence of overweight and obesity in adolescents.


Assuntos
Fertilidade/fisiologia , Hormônio Liberador de Gonadotropina/fisiologia , Puberdade/fisiologia , Adolescente , Animais , Encéfalo/fisiologia , Feminino , Fertilidade/genética , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Hipotálamo/metabolismo , Recém-Nascido , Masculino , Neurônios/fisiologia , Receptores Acoplados a Proteínas G , Receptores de Kisspeptina-1 , Receptores de Neuropeptídeos/fisiologia , Transdução de Sinais/fisiologia
18.
Eur J Neurosci ; 20(2): 338-44, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15233743

RESUMO

Secretion of gonadotropin-releasing hormone (GnRH) at the median eminence is the essential activator of the reproductive axis. The mechanisms by which embryonic GnRH neurons migrate from the olfactory placode to the preoptic area and then elaborate neurites that course through the hypothalamus to terminate at the median eminence are largely unknown. We investigated the hypothesis that GnRH neurite outgrowth is promoted by brain-derived neurotrophic factor (BDNF) because GnRH neurites course through BDNF-rich areas of the forebrain during their development. Confocal microscopy revealed that most (86%) cultured embryonic GnRH cells tagged with a green fluorescent protein reporter were immunoreactive for TrkB. In primary cultures of E12.5 olfactory tissue, treatment with BDNF induced a dose-dependent increase in neurite outgrowth, but had no discernible effect on branching. BDNF induced phosphorylation of Ca(2+)/cAMP response element-binding protein (pCREB) in both GnRH and non-GnRH cells in these cultures. This was not associated with phosphorylation of ERK in GnRH-immunoreactive cells, though BDNF treatment did stimulate pERK in neighbouring non-GnRH cells. Promotion of neurite outgrowth is unlikely therefore to result from activation of the Ras-MAPK/ERK pathway. We conclude that the developing GnRH secretory system is directly sensitive to BDNF and that this polypeptide functions as a neurotrophic factor for GnRH neurons.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/farmacologia , Encéfalo/citologia , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/efeitos dos fármacos , Animais , Encéfalo/embriologia , Contagem de Células/métodos , Células Cultivadas , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Interações Medicamentosas , Embrião de Mamíferos , Hormônio Liberador de Gonadotropina/genética , Proteínas de Fluorescência Verde , Técnicas Imunoenzimáticas/métodos , Imuno-Histoquímica/métodos , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neuritos/efeitos dos fármacos , Neuritos/fisiologia , Neurônios/fisiologia , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Cloreto de Potássio/farmacologia , Receptor trkB/metabolismo
19.
Proc Natl Acad Sci U S A ; 100(5): 2831-5, 2003 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-12598657

RESUMO

Melatonin is produced nocturnally by the pineal gland and is a neurochemical representation of time. It regulates neuroendocrine target tissues through G-protein-coupled receptors, of which MT(1) is the predominant subtype. These receptors are transiently expressed in several fetal and neonatal tissues, suggesting distinct roles for melatonin in development and that specific developmental cues define time windows for melatonin sensitivity. We have investigated MT(1) gene expression in the rat pituitary gland. MT(1) mRNA is confined to the pars tuberalis region of the adult pituitary, but in neonates extends into the ventral pars distalis and colocalizes with luteinizing hormone beta-subunit (LH beta) expression. This accounts for the well documented transient sensitivity of rat gonadotrophs to melatonin in the neonatal period. Analysis of an upstream fragment of the rat MT(1) gene revealed multiple putative response elements for the transcription factor pituitary homeobox-1 (Pitx-1), which is expressed in the anterior pituitary from Rathke's pouch formation. A Pitx-1 expression vector potently stimulated expression of both MT(1)-luciferase and LH beta-luciferase reporter constructs in COS-7 cells. Interestingly, transcription factors that synergize with Pitx-1 to trans-activate gonadotroph-associated genes did not potentiate Pitx-1-induced MT(1)-luciferase activity. Moreover, the transcription factor, early growth response factor-1, which is induced by gonadotrophin-releasing hormone (GnRH) and trans-activates LH beta expression, attenuated Pitx-1-induced MT(1)-luciferase activity. Finally, pituitary MT(1) gene expression was 4-fold higher in hypogonadal (hpg) mice, which do not synthesize GnRH, than in their wild-type littermates. These data suggest that establishment of a mature hypothalamic GnRH input drives the postnatal decline in pituitary MT(1) gene expression.


Assuntos
Regulação para Baixo , Hormônio Liberador de Gonadotropina/fisiologia , Hipófise/embriologia , Receptores de Superfície Celular/biossíntese , Receptores Citoplasmáticos e Nucleares/biossíntese , Animais , Células COS , Clonagem Molecular , DNA Complementar/metabolismo , Genes Reporter , Proteínas de Homeodomínio/metabolismo , Hibridização In Situ , Luciferases/metabolismo , Melatonina/metabolismo , Camundongos , Dados de Sequência Molecular , Fatores de Transcrição Box Pareados , Hipófise/metabolismo , Plasmídeos/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , RNA Mensageiro/metabolismo , Ratos , Receptores de Melatonina , Fatores de Transcrição/metabolismo , Ativação Transcricional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA