Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Eur J Epidemiol ; 37(10): 1087-1105, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36245062

RESUMO

The Food Chain Plus (FoCus) cohort was launched in 2011 for population-based research related to metabolic inflammation. To characterize this novel pathology in a comprehensive manner, data collection included multiple omics layers such as phenomics, microbiomics, metabolomics, genomics, and metagenomics as well as nutrition profiling, taste perception phenotyping and social network analysis. The cohort was set-up to represent a Northern German population of the Kiel region. Two-step recruitment included the randomised enrolment of participants via residents' registration offices and via the Obesity Outpatient Centre of the University Medical Center Schleswig-Holstein (UKSH). Hence, both a population- and metabolic inflammation- based cohort was created. In total, 1795 individuals were analysed at baseline. Baseline data collection took place between 2011 and 2014, including 63% females and 37% males with an age range of 18-83 years. The median age of all participants was 52.0 years [IQR: 42.5; 63.0 years] and the median baseline BMI in the study population was 27.7 kg/m2 [IQR: 23.7; 35.9 kg/m2]. In the baseline cohort, 14.1% of participants had type 2 diabetes mellitus, which was more prevalent in the subjects of the metabolic inflammation group (MIG; 31.8%). Follow-up for the assessment of disease progression, as well as the onset of new diseases with changes in subject's phenotype, diet or lifestyle factors is planned every 5 years. The first follow-up period was finished in 2020 and included 820 subjects.


Assuntos
Diabetes Mellitus Tipo 2 , Feminino , Humanos , Masculino , Estudos de Coortes , Diabetes Mellitus Tipo 2/epidemiologia , Cadeia Alimentar , Inflamação , Obesidade/epidemiologia , Adolescente , Adulto Jovem , Adulto , Pessoa de Meia-Idade , Idoso , Idoso de 80 Anos ou mais
2.
Nat Commun ; 11(1): 4291, 2020 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-32855407

RESUMO

The annual photoperiod cycle provides the critical environmental cue synchronizing rhythms of life in seasonal habitats. In 1936, Bünning proposed a circadian-based coincidence timer for photoperiodic synchronization in plants. Formal studies support the universality of this so-called coincidence timer, but we lack understanding of the mechanisms involved. Here we show in mammals that long photoperiods induce the circadian transcription factor BMAL2, in the pars tuberalis of the pituitary, and triggers summer biology through the eyes absent/thyrotrophin (EYA3/TSH) pathway. Conversely, long-duration melatonin signals on short photoperiods induce circadian repressors including DEC1, suppressing BMAL2 and the EYA3/TSH pathway, triggering winter biology. These actions are associated with progressive genome-wide changes in chromatin state, elaborating the effect of the circadian coincidence timer. Hence, circadian clock-pituitary epigenetic pathway interactions form the basis of the mammalian coincidence timer mechanism. Our results constitute a blueprint for circadian-based seasonal timekeeping in vertebrates.


Assuntos
Fatores de Transcrição ARNTL/genética , Relógios Circadianos/fisiologia , Fotoperíodo , Hipófise/fisiologia , Ovinos/fisiologia , Fatores de Transcrição ARNTL/metabolismo , Animais , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Epigênese Genética , Regulação da Expressão Gênica , Masculino , Melatonina/genética , Melatonina/metabolismo , Estações do Ano
3.
J Neuroendocrinol ; 27(4): 277-84, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25650820

RESUMO

In seasonal mammals living in temperate zones, photoperiod regulates prolactin secretion, such that prolactin plasma concentrations peak during the summer months and are lowest during the winter. In sheep, a short-day breeder, circulating prolactin has important modulatory effects on the reproductive system via inhibitory actions on pituitary gonadotrophs and hypothalamic gonadotrophin-releasing hormone release. The exact cellular mechanisms that account for the chronic hypersecretion of prolactin during the summer is not known, although evidence supports an intrapituitary mechanism regulated by melatonin. Folliculo-stellate (FS) cells are non-endocrine cells that play a crucial role in paracrine communication within the pituitary and produce factors controlling prolactin and gonadotrophin release. The present study examined the morphology of the FS and lactotroph cell populations and their distribution in the sheep pituitary during the annual reproductive cycle. Ovine pituitary glands were collected in the winter (breeding season; BS) and summer (nonbreeding season; NBS) and were prepared for quantitative electron microscopy to assess the effects of season on FS and lactotroph cell density, morphology and distribution, as well as on junctional contacts between cells. It was found that lactotrophs in the NBS are larger in size and contain more numerous PRL granules than lactotrophs in the BS. FS cells were also larger in the NBS compared to BS and showed altered morphology such that, in the BS, long cell processes surrounded clusters of adjacent secretory cells. Although no significant change in the number of junctions was observed between lactotrophs and FS cells, or lactotrophs and gonadotrophs, there was a significant increase in the number of adherens junctions between lactotrophs and between FS cells. These findings demonstrate seasonal plasticity in the morphology of lactotrophs and FS cells that reflect changes in PRL secretion.


Assuntos
Lactotrofos/ultraestrutura , Hipófise/ultraestrutura , Reprodução/fisiologia , Ovinos , Animais , Comunicação Celular , Feminino , Lactotrofos/citologia , Microscopia Eletrônica , Hipófise/citologia , Estações do Ano , Ovinos/anatomia & histologia
4.
J Neuroendocrinol ; 26(11): 785-95, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25039914

RESUMO

To investigate the relationship between gonadotroph function and ultrastructure, we have compared, in parallel in female mice, the effects of several different mutations that perturb the hypothalamic-pituitary-gonadal axis. Specifically, serum and pituitary gonadotrophin concentrations, gonadotrophin gene expression, gonadotroph structure and number were measured. Follicle-stimulating hormone ß knockout (FSHßKO), follicle-stimulating hormone receptor knockout (FSHRKO), luteinising hormone receptor knockout (LuRKO), hypogonadal (hpg) and ovariectomised mice were compared with control wild-type or heterozygote female mice. Serum levels of LH were elevated in FSHßKO and FSHRKO compared to heterozygote females, reflecting the likely decreased oestrogen production in KO females, as demonstrated by the threadlike uteri and acyclicity. As expected, there was no detectable FSH in the serum or pituitary and an absence of expression of the FSHß subunit gene in FSHßKO mice. However, there was a significant increase in expression of the FSHß and LHß subunit genes in FSHRKO female mice. The morphology of FSHßKO and FSHRKO gonadotrophs was not significantly different from the control, except that secretory granules in FSHRKO gonadotrophs were larger in diameter. In LuRKO and ovariectomised mice, stimulation of LHß and FSHß mRNA, as well as serum protein concentrations, were reflected in subcellular changes in gonadotroph morphology, including more dilated rough endoplasmic reticula and fewer, larger secretory granules. In the gonadotophin-releasing hormone deficient hpg mouse, gonadotrophin mRNA and protein levels were significantly lower than in control mice and gonadotrophs were correspondingly smaller with less abundant endoplasmic reticula and reduced numbers of secretory granules. In summary, major differences in pituitary content and serum concentrations of the gonadotrophins LH and FSH were found between control and mutant female mice. These changes were associated with changes in expression of the gonadotrophin subunit genes and were reflected in the cellular structure and secretory granule appearance within the gonadotroph cells.


Assuntos
Hormônio Foliculoestimulante/metabolismo , Expressão Gênica , Hipogonadismo/metabolismo , Hormônio Luteinizante/metabolismo , Hipófise/metabolismo , Animais , Feminino , Hormônio Foliculoestimulante/genética , Hipogonadismo/genética , Hormônio Luteinizante/genética , Camundongos , Camundongos Knockout , Hipófise/citologia , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
5.
PLoS One ; 9(4): e93860, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24710599

RESUMO

OBJECTIVE: The aim of the present study was to examine to what extent different social network mechanisms are involved in the pathogenesis of obesity and insulin-resistance. DESIGN: We used nonparametric and parametric regression models to analyse whether individual BMI and HOMA-IR are determined by social network characteristics. SUBJECTS AND METHODS: A total of 677 probands (EGO) and 3033 social network partners (ALTER) were included in the study. Data gathered from the probands include anthropometric measures, HOMA-IR index, health attitudes, behavioural and socio-economic variables and social network data. RESULTS: We found significant treatment effects for ALTERs frequent dieting (p<0.001) and ALTERs health oriented nutritional attitudes (p<0.001) on EGO's BMI, establishing a significant indirect network effect also on EGO's insulin resistance. Most importantly, we also found significant direct social network effects on EGO's insulin resistance, evidenced by an effect of ALTERs frequent dieting (p = 0.033) and ALTERs sport activities (p = 0.041) to decrease EGO's HOMA-IR index independently of EGO's BMI. CONCLUSIONS: Social network phenomena appear not only to be relevant for the spread of obesity, but also for the spread of insulin resistance as the basis for type 2 diabetes. Attitudes and behaviour of peer groups influence EGO's health status not only via social mechanisms, but also via socio-biological mechanisms, i.e. higher brain areas might be influenced not only by biological signals from the own organism, but also by behaviour and knowledge from different human individuals. Our approach allows the identification of peer group influence controlling for potential homophily even when using cross-sectional observational data.


Assuntos
Resistência à Insulina/fisiologia , Obesidade/fisiopatologia , Grupo Associado , Apoio Social , Adulto , Idoso , Índice de Massa Corporal , Estudos Transversais , Feminino , Nível de Saúde , Humanos , Masculino , Pessoa de Meia-Idade , Obesidade/psicologia
6.
J Neuroendocrinol ; 26(2): 111-20, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24176035

RESUMO

Vascular endothelial growth factor (VEGF) is an endothelial cell mitogen responsible for physiological and pathological angiogenesis. Abnormal regulation of VEGF expression in anterior pituitary folliculostellate (FS) cells has been implicated in pituitary tumour progression. FS and endocrine cells express VEGF, which is considered to be secreted by the constitutive pathway. The present study investigated the mechanism of VEGF secretion in TtT/GF cells, a mouse FS cell line. TtT/GF cells were shown to express VEGF(164), the most potent and bioavailable isoform of VEGF. Immunofluorescence and immunogold electron microscopy localised VEGF to the cytoplasm and small electron-lucent vesicles. Pituitary adenylate cyclase-activating polypeptide (PACAP), a well-documented stimulant of VEGF secretion, caused a robust increase in VEGF secretion over 24 h. Glyburide, an ABCA1 and K(ATP) channel blocker, also caused an increase in VEGF secretion when applied alone, and amplified the response to PACAP. Other ABCA1 transport blockers did not affect VEGF secretion. Exposure of TtT/GF cells to cycloheximide with PACAP or glyburide inhibited the increased secretion of VEGF, consistent with control of secretion at the transcription level. The SUR2B/Kir6.1 form of K(ATP) channels was shown to be expressed by TtT/GF cells. Diazoxide, a K(ATP) activator, inhibited PACAP- and PACAP + glyburide-stimulated VEGF secretion but not that of glyburide alone. These data suggest that K(ATP) channels are expressed by FS cells and play a significant role in the control of VEGF secretion, and also that activation of K(ATP) channels inhibits the secretion of VEGF at the level of transcription.


Assuntos
Canais KATP/fisiologia , Hipófise/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Sequência de Bases , Células Cultivadas , Cicloeximida/farmacologia , Primers do DNA , Ensaio de Imunoadsorção Enzimática , Imunofluorescência , Camundongos , Microscopia Imunoeletrônica/métodos , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Hipófise/citologia , Biossíntese de Proteínas/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
J Neuroendocrinol ; 25(10): 863-75, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23895394

RESUMO

To investigate brain-pituitary-gonadal inter-relationships, we have compared the effects of mutations that perturb the hypothalamic-pituitary-gonadal axis in male mice. Specifically, serum and pituitary gonadotrophin concentrations, gonadotrophin gene expression, and gonadotroph structure and number were measured. Follicle-stimulating hormone (FSH)ß knockout (FSHßKO), FSH receptor knockout (FSHRKO), luteinising hormone (LH) receptor knockout (LuRKO), hypogonadal (hpg), testicular feminised (tfm) and gonadectomised mice were compared with control wild-type mice or heterozygotes. Serum levels of LH were similar in FSHßKO, FSHRKO and heterozygote males despite decreased androgen production in KO males. As expected, there was no detectable FSH in the serum or pituitary and an absence of expression of the FSHß subunit gene in FSHßKO mice. However, there was a significant increase in expression of the common α and LHß subunit genes in FSHRKO males. The morphology of FSHßKO and FSHRKO gonadotrophs was not significantly different from controls, except that the subpopulation of granules consisting of an electron-dense core and electron-lucent 'halo' was not observed in FSHßKO gonadotrophs and the granules were smaller in diameter. In the gonadotrophin-releasing hormone deficient hpg mouse, gonadotrophin mRNA and hormone levels were significantly lower compared to control mice and gonadotrophs were correspondingly smaller, with less abundant endoplasmic reticulum and reduced secretory granules. In LuRKO, tfm and gonadectomised mice, hyperstimulation of LHß and FSHß mRNA and serum protein concentrations was reflected by subcellular changes in gonadotroph morphology, including more dilated rough endoplasmic reticulum and more secretory granules distributed adjacent to the plasma membrane. In summary, major differences in pituitary content and serum concentrations of the gonadotrophins LH and FSH have been found between normal and mutant male mice. These changes are associated with changes in transcriptional activity of the gonadotrophin subunit genes and are reflected by changes in the cellular structure and secretory granule architecture within the gonadotroph cells.


Assuntos
Expressão Gênica , Hipófise/metabolismo , Hormônios Hipofisários/metabolismo , Animais , Sequência de Bases , Primers do DNA , Masculino , Camundongos , Camundongos Knockout , Camundongos Mutantes , Microscopia Eletrônica , Hormônios Hipofisários/genética , Reação em Cadeia da Polimerase em Tempo Real
8.
Xenobiotica ; 38(7-8): 1072-106, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18668440

RESUMO

1. The monocarboxylate transporter (MCT, SLC16) family comprises 14 members, of which to date only MCT1-4 have been shown to carry monocarboxylates, transporting important metabolic compounds such as lactate, pyruvate and ketone bodies in a proton-coupled manner. The transport of such compounds is fundamental for metabolism, and the tissue locations, properties and regulation of these isoforms is discussed. 2. Of the other members of the MCT family, MCT8 (a thyroid hormone transporter) and TAT1 (an aromatic amino acid transporter) have been characterized more recently, and their physiological roles are reviewed herein. The endogenous substrates and functions of the remaining members of the MCT family await elucidation. 3. The MCT proteins have the typical twelve transmembrane-spanning domain (TMD) topology of membrane transporter proteins, and their structure-function relationship is discussed, especially in relation to the future impact of the single nucleotide polymorphism (SNP) databases and, given their ability to transport pharmacologically relevant compounds, the potential impact for pharmacogenomics.


Assuntos
Transportadores de Ácidos Monocarboxílicos/metabolismo , Família Multigênica/fisiologia , Sistemas de Transporte de Aminoácidos Neutros/genética , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Animais , Transporte Biológico/fisiologia , Humanos , Transportadores de Ácidos Monocarboxílicos/genética , Especificidade de Órgãos/fisiologia , Farmacogenética , Estrutura Terciária de Proteína/fisiologia
9.
J Neuroendocrinol ; 20(3): 309-22, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18208550

RESUMO

The elevation in baseline circulating growth hormone (GH) that occurs in pregnant rats is thought to arise from increased pituitary GH secretion, but the underlying mechanism remains unclear. Distribution, Fourier and algorithmic analyses confirmed that the pregnancy-induced increase in circulating GH in 3-week pregnant rats was due to a 13-fold increase in baseline circulating GH (P < 0.01), without any significant alteration in the parameters of episodic secretion. Electron microscopy revealed that pregnancy resulted in a reduction in the proportion of mammosomatotrophs (P < 0.01) and an increase in type II lactotrophs (P < 0.05), without any significant change in the somatotroph population. However, the density of the secretory granules in somatotrophs from 3-week pregnant rats was reduced (P < 0.05), and their distribution markedly polarised; the granules being grouped nearest the vasculature. Pituitary GH content was not increased, but steady-state GH mRNA levels declined progressively during pregnancy (P < 0.05). In situ hybridisation revealed that pregnancy was accompanied by a suppression of GH-releasing hormone mRNA expression in the arcuate nuclei (P < 0.05) and enhanced somatostatin mRNA expression in the periventricular nuclei (P < 0.05), an expression pattern normally associated with increased GH feedback. Although gastric ghrelin mRNA expression was elevated by 50% in 3-week pregnant rats (P < 0.01), circulating ghrelin, GH-secretagogue receptor mRNA expression and the GH response to a bolus i.v. injection of exogenous ghrelin were all largely unaffected during pregnancy. Although trace amounts of 'pituitary' GH could be detected in the placenta with radioimmunoassay, significant GH-immunoreactivity could not be observed by immunohistochemistry, indicating that rat placenta itself does not produce 'pituitary' GH. Although not excluding the possibility that the pregnancy-associated elevation in baseline circulating GH could arise from alternative extra-pituitary sources (e.g. the ovary), our data indicate that this phenomenon is most likely to result from a direct alteration of somatotroph function.


Assuntos
Grelina/fisiologia , Hormônio do Crescimento/sangue , Prenhez , Adiposidade/fisiologia , Animais , Anticorpos Monoclonais/metabolismo , Peso Corporal/fisiologia , Feminino , Grelina/genética , Grelina/metabolismo , Grelina/farmacologia , Hormônio do Crescimento/genética , Hormônio do Crescimento/metabolismo , Crescimento e Desenvolvimento/fisiologia , Hormônios Hipotalâmicos/metabolismo , Masculino , Placenta/metabolismo , Gravidez , Ratos , Ratos Sprague-Dawley , Receptores de Grelina/genética , Receptores de Grelina/metabolismo , Somatotrofos/fisiologia , Fatores de Tempo , Regulação para Cima
10.
J Neuroendocrinol ; 19(8): 605-13, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17620102

RESUMO

In the male rat anterior pituitary, three morphological subtypes of cells secreting primarily prolactin (PRL) (lactotrophs) have been described. Type I contain predominantly large irregularly shaped granules, whereas type II and type III lactotrophs contain smaller spherical granules. We have previously shown that oestradiol and testosterone exert a rapid stimulatory effect selectively on type II lactotrophs but it is not known how the lactotroph subtypes respond to peptide secretagogues. We have therefore examined which cell subtype(s) release PRL in response to vasoactive intestinal peptide (VIP), thyrotrophin-releasing hormone (TRH) and prolactin-releasing peptide (PrRP-31). Pituitary segments were incubated in medium containing tannic acid (to capture exocytosis of secretory granules), either alone or with secretagogue peptide. VIP (1-10 nM), TRH (10 nM) and PrRP-31 (10 nM) all caused a significant increase (P < 0.05) in the amount of PRL granule exocytosis from type II and III lactotrophs, but had no effect on PRL exocytosis from type I. Dopamine (100 nM) inhibited basal exocytosis of immunoreactive (ir)-PRL from type I, II and III lactotrophs and PrRP-31-stimulated ir-PRL granule exocytosis from II and III lactotrophs. Treatment of lactating female rats with the dopamine D(2) receptor antagonist sulpiride (40 microg/kg) produced a significant increase (P < 0.05) in PRL granule exocytosis from type I and type III lactotrophs and a significant increase (P < 0.05) in the proportion of type I and II cells undergoing exocytosis of PRL. In conclusion, VIP, TRH and PrRP-31 selectively stimulate exocytosis from type II and III lactotrophs in the male rat, whereas all three lactotroph types are sensitive to dopamine inhibition of exocytosis in male and female rats.


Assuntos
Dopamina/farmacologia , Hormônios Hipotalâmicos/farmacologia , Lactotrofos/efeitos dos fármacos , Neuropeptídeos/farmacologia , Prolactina/metabolismo , Caracteres Sexuais , Hormônio Liberador de Tireotropina/farmacologia , Peptídeo Intestinal Vasoativo/farmacologia , Animais , Antagonistas de Dopamina/farmacologia , Feminino , Lactação/efeitos dos fármacos , Lactação/metabolismo , Lactotrofos/classificação , Lactotrofos/metabolismo , Lactotrofos/ultraestrutura , Masculino , Microscopia Eletrônica , Adeno-Hipófise/citologia , Adeno-Hipófise/efeitos dos fármacos , Adeno-Hipófise/metabolismo , Hormônio Liberador de Prolactina , Radioimunoensaio , Ratos , Ratos Sprague-Dawley , Sulpirida/farmacologia
11.
FASEB J ; 21(4): 1037-46, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17218541

RESUMO

The N-formyl peptide receptors (FPRs) are a family of G-protein coupled receptors that respond to proinflammatory N-formylated bacterial peptides (e.g., formyl-Met-Leu-Phe, fMLF) and, thus, contribute to the host response to bacterial infection. Paradoxically, a growing body of evidence suggests that some members of this receptor family may also be targets for certain anti-inflammatory molecules, including annexin A1 (ANXA1), which is an important mediator of glucocorticoid (GC) action. To explore further the potential role of FPRs in mediating ANXA1 actions, we have focused on the pituitary gland, where ANXA1 has a well-defined role as a cell-cell mediator of the inhibitory effects of GCs on the secretion of corticotrophin (ACTH), and used molecular, genetic, and pharmacological approaches to address the question in well-established rodent models. Reverse transcriptase-polymerase chain reaction (RT-PCR) analysis identified mRNAs for four FPR family members in the mouse anterior pituitary gland, Fpr-rs1, Fpr-rs2, Fpr-rs6, and Fpr-rs7. Functional studies confirmed that, like dexamethasone, ANXA1 and two ANXA1-derived peptides (ANXA1(1-188) and ANXA1(Ac2-26)) inhibit the evoked release of ACTH from rodent anterior pituitary tissue in vitro. Fpr1 gene deletion failed to modify the pituitary responses to dexamethasone or ANXA1(Ac2-26). However, lipoxin A4 (LXA4, 0.02-2 microM, a lipid mediator with high affinity for Fpr-rs1) mimicked the inhibitory effects of ANXA1 on ACTH release as also did fMLF in high (1-100 microM) but not lower (10-100 nM) concentrations. Additionally, a nonselective FPR antagonist (Boc1, 100 microM) overcame the effects of dexamethasone, ANXA1(1-188), ANXA1(Ac2-26), fMLF, and LXA4 on ACTH release, although at a lower concentration (50 microM), it was without effect. Together, the results suggest that the actions of ANXA1 in the pituitary gland are independent of Fpr1 but may involve other FPR family members, in particular, Fpr-rs1 or a closely related receptor. They thus provide the first evidence for a role of the FPR family in the regulation of neuroendocrine function.


Assuntos
Hormônio Adrenocorticotrópico/metabolismo , Anexina A1/metabolismo , Bactérias/metabolismo , Regulação da Expressão Gênica , Lipoxinas/metabolismo , Peptídeos/química , Receptores de Formil Peptídeo/química , Animais , Anti-Inflamatórios/farmacologia , Glucocorticoides/metabolismo , Masculino , Camundongos , Camundongos Knockout , Hipófise/metabolismo , Ratos , Receptores de Formil Peptídeo/metabolismo
12.
J Neuroendocrinol ; 18(12): 949-59, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17076770

RESUMO

Perinatal glucocorticoid (GC) treatment is increasingly associated with long-term disturbances in hypothalamo-pituitary-adrenocortical function. In the male rat, such treatment induces profound molecular, morphological and functional changes in the anterior pituitary gland at adulthood. To determine whether these effects are sex-specific, we have examined the effects of perinatal dexamethasone treatment on the female pituitary gland, focusing on (i) the integrity of the annexin 1 (ANXA1) dependent regulatory effects of GCs on adrenocorticotrophic hormone (ACTH) release and (ii) corticotroph and folliculo-stellate (FS) cell morphology. Dexamethasone was given to pregnant (gestational days 16-19) or lactating (days 1-7 post partum) rats via the drinking water (1 microg/ml); controls received normal drinking water. Pituitary tissue from the female offspring was examined ex vivo at adulthood (60-90 days). Both treatment regimes reduced the intracellular and cell surface ANXA1 expression, as determined by western blot analysis and quantitative immunogold electron microscopic histochemistry. In addition, they compromised the ability of dexamethasone to suppress the evoked release of ACTH from the excised tissue in vitro, a process which requires the translocation of ANXA1 from the cytoplasm to the cell surface of FS cells. Although neither treatment regime affected the number of FS cells or corticotrophs, both altered the subcellular morphology of these cells. Thus, prenatal dexamethasone treatment increased while neonatal treatment decreased FS cell size and cytoplasmic area. By contrast, corticotroph size was unaffected by either treatment, as also was the size of the secretory granules. Corticotroph granule density and margination were, however, increased markedly by the prenatal treatment, while the neonatal treatment had no effect on granule density but decreased granule margination. Thus, perinatal dexamethasone treatment exerts long-term effects on the female pituitary gland, altering gene expression, cell morphology and the ANXA1-dependent GC regulation of ACTH secretion. The changes are similar but not identical to those reported in the male.


Assuntos
Hormônio Adrenocorticotrópico/metabolismo , Anexina A1/metabolismo , Glucocorticoides/fisiologia , Adeno-Hipófise/fisiologia , Efeitos Tardios da Exposição Pré-Natal , Hormônio Adrenocorticotrópico/efeitos dos fármacos , Fatores Etários , Animais , Anexina A1/efeitos dos fármacos , Corticotrofos/efeitos dos fármacos , Corticotrofos/ultraestrutura , Dexametasona/farmacologia , Retroalimentação Fisiológica/fisiologia , Feminino , Glucocorticoides/farmacologia , Imuno-Histoquímica , Técnicas In Vitro , Masculino , Neurônios/efeitos dos fármacos , Neurônios/ultraestrutura , Adeno-Hipófise/efeitos dos fármacos , Adeno-Hipófise/ultraestrutura , Gravidez , Ratos , Ratos Sprague-Dawley , Fatores Sexuais , Fatores de Tempo
13.
J Neuroendocrinol ; 18(11): 835-46, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17026533

RESUMO

Annexin 1 (ANXA1) is a member of the annexin family of phospholipid- and calcium-binding proteins with a well demonstrated role in early delayed (30 min to 3 h) inhibitory feedback of glucocorticoids in the pituitary. We have examined corticotrophs in wild-type and ANXA1 knockout mice to determine the effects of lack of ANXA1 in male and female animals. Anterior pituitary tissue from ANXA1 wild-type, heterozygote and null mice was fixed and examined (i) by confocal immunocytochemistry to determine the number of corticotrophs and (ii) by electron microscopy to examine the size, secretory granule population and secretory machinery of corticotrophs. No differences in these parameters were detected in female mice. In male ANXA1 null mice, there were approximately four-fold more corticotrophs than in wild-type animals. However, the corticotrophs in ANXA1 null mice were smaller and had reduced numbers of secretory granules (the reduction in granules paralleled the reduction in cell size). No differences in the numerical density of folliculo-stellate, gonadotroph, lactotroph or somatotroph cells were detected in male ANXA1 null mice. Plasma corticosterone, adrenocorticotrophic hormone (ACTH) and pituitary pro-opiomelanocortin mRNA were unchanged but pituitary ACTH content was increased in male ANXA1 null mice. Interleukin (IL)-6 pituitary content was significantly elevated in male and reduced in female ANXA1 null mice compared to wild-type. In conclusion, these data indicate that ANXA1 deficiency is associated with gender-specific changes in corticotroph number and structure, via direct actions of ANXA1 and/or indirect changes in factors such as IL-6.


Assuntos
Hormônio Adrenocorticotrópico/sangue , Anexina A1/metabolismo , Corticotrofos/citologia , Interleucina-6/metabolismo , Animais , Anexina A1/genética , Tamanho Corporal , Contagem de Células , Tamanho Celular , Corticosterona/sangue , Corticotrofos/metabolismo , Corticotrofos/ultraestrutura , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hipófise/metabolismo , Hipófise/ultraestrutura , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo , RNA Mensageiro/análise , Fatores Sexuais
14.
J Neuroendocrinol ; 18(10): 719-31, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16965290

RESUMO

Growth hormone (GH) is known to regulate peripheral components of the hypothalamo-pituitary gonadal (HPG) axis, but it remains unclear whether GH exerts a significant influence on the activity of the hypothalamo-pituitary components of the HPG axis. In this study, we investigated the development of HPG axis function in the male transgenic growth retarded (Tgr) rat, a model of moderate systemic GH deficiency caused by hypothalamic expression of human (h)GH. Impaired postnatal somatotroph expansion and moderate GH deficiency in male Tgr rats were accompanied by a two- to three-fold increase in pituitary gonadotrophin content, but without a significant change in the pituitary gonadotroph population. A three- to nine-fold elevation in basal circulating luteinising hormone concentration was seen in postpubertal Tgr rats, with a smaller increase in follicle-stimulating hormone. Despite this hypergonadotrophism, there was no corresponding increase in steroidogenic (circulating testosterone and seminal vesicle weights) or gametogenic (spermatozoa counts in seminiferous tubules) activity in the postpubertal Tgr testis. Following puberty, the plasma leptin concentration also became progressively elevated in Tgr males. Circulating gonadotrophin and leptin levels were normalised in Tgr rats by peripheral physiological replacement of rat GH, but plasma testosterone concentration was unaffected. These results confirm that hGH exerts a positive influence on the central control of gonadotrophin secretion in the Tgr rat, but the absence of a corresponding elevation in the steroidogenic or gametogenic function of the Tgr testis implies that the peripheral GH/insulin-like growth factor I axis may also exert a permissive influence on testicular function. The relative contribution of somatogenic and lactogenic mechanisms and the potential influence of elevated leptin and decreased sensitivity to androgen feedback to the development of postpubertal hypergonadotrophism in Tgr males remain to be determined.


Assuntos
Gonadotropinas/metabolismo , Hormônio do Crescimento Humano/metabolismo , Hormônio do Crescimento Humano/fisiologia , Hipotálamo/metabolismo , Animais , Animais Geneticamente Modificados , Hormônios/sangue , Humanos , Masculino , Microscopia Eletrônica , Hipófise/citologia , Hipófise/metabolismo , Hipófise/ultraestrutura , Ratos , Contagem de Espermatozoides , Testículo/efeitos dos fármacos , Testículo/crescimento & desenvolvimento
15.
FASEB J ; 20(9): 1498-500, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16720734

RESUMO

Annexin A1 (ANXA1) has an important role in cell-cell communication in the host defense and neuroendocrine systems. In both systems, its actions are exerted extracellularly via membrane-bound receptors on adjacent sites after translocation of the protein from the cytoplasm to the cell surface of adjacent cells. This study used molecular, microscopic, and pharmacological approaches to explore the mechanisms underlying the cellular exportation of ANXA1 in TtT/GF (pituitary folliculo-stellate) cells. LPS caused serine-phosphorylation of ANXA1 (ANXA1-S27-PO4) and translocation of the phosphorylated protein to the cell membrane. The fundamental requirement of phosphorylation for membrane translocation was confirmed by immunofluorescence microscopy on cells transfected with wild-type or mutated (S27/A) ANXA1 constructs tagged with enhanced green fluorescence protein. The trafficking of ANXA1-S27-PO4 to the cell surface was dependent on PI3-kinase and MAP-kinase. It also required HMG-coenzyme A and myristoylation. The effects of HMG-coenzyme A blockade were overcome by mevalonic acid (the product of HMG-coenzyme A) and farnesyl-pyrophosphate but not by geranyl-geranylpyrophosphate or cholesterol. Together, these results suggest that serine-27 phosphorylation is essential for the translocation of ANXA1 across the cell membrane and also identify a role for isoprenyl lipids. Such lipids could target consensus sequences in ANXA1. Alternatively, they may target other proteins in the signal transduction cascade (e.g., transporters).


Assuntos
Anexina A1/metabolismo , Membrana Celular/metabolismo , Processamento de Proteína Pós-Traducional , Animais , Anexina A1/genética , Comunicação Celular , Linhagem Celular Tumoral , Citoplasma/metabolismo , Inibidores Enzimáticos/farmacologia , Genes Reporter , Lipopolissacarídeos/farmacologia , Ácido Mevalônico/farmacologia , Camundongos , Mutagênese Sítio-Dirigida , Fosforilação , Neoplasias Hipofisárias , Transporte Proteico , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais
16.
Endocrinology ; 147(4): 1904-15, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16439449

RESUMO

This study aimed to test the hypothesis that the tuberoinfundibular dopaminergic neurons of the arcuate nucleus and/or the lactotroph cells of the anterior pituitary gland are key targets for the programming effects of perinatal glucocorticoids (GCs). Dexamethasone was administered noninvasively to fetal or neonatal rats via the mothers' drinking water (1 mug/ml) on embryonic d 16-19 or neonatal d 1-7, and control animals received normal drinking water. At 68 d of age, the numbers of tyrosine hydroxylase-positive (TH+) cells in the arcuate nucleus and morphometric parameters of pituitary lactotrophs were analyzed. In control animals, striking sex differences in TH+ cell numbers, lactotroph cell size, and pituitary prolactin content were observed. Both pre- and neonatal GC treatment regimens were without effect in adult male rats, but in females, the overriding effect was to abolish the sex differences by reducing arcuate TH+ cell numbers (pre- and neonatal treatments) and reducing lactotroph cell size and pituitary prolactin content (prenatal treatment only) without changing lactotroph cell numbers. Changes in circulating prolactin levels represented a net effect of hypothalamic and pituitary alterations that exhibited independent critical windows of susceptibility to perinatal GC treatments. The dopaminergic neurons of the hypothalamic periventricular nucleus and the pituitary somatotroph populations were not significantly affected by either treatment regimen in either sex. These data show that the adult female hypothalamo-lactotroph axis is profoundly affected by perinatal exposure to GCs, which disrupts the tonic inhibitory tuberoinfundibular dopaminergic pathway and changes lactotroph morphology and prolactin levels in the pituitary and circulation. These findings provide new evidence for a long-term disruption in prolactin-dependent homeostasis in females, but not males, after inappropriate GC exposure in perinatal life.


Assuntos
Dexametasona/toxicidade , Feto/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Prolactina/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/patologia , Dopamina/análise , Feminino , Hormônio do Crescimento/análise , Hormônio do Crescimento/metabolismo , Sistema Hipotálamo-Hipofisário/fisiologia , Masculino , Hipófise/patologia , Gravidez , Prolactina/análise , Prolactina/sangue , Ratos , Ratos Sprague-Dawley , Caracteres Sexuais , Tirosina 3-Mono-Oxigenase/análise
17.
Endocrinology ; 146(11): 4804-13, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16099861

RESUMO

Stress or glucocorticoid (GC) treatment in perinatal life can induce long-term changes in the sensitivity of the hypothalamo-pituitary-adrenocortical axis to the feedback actions of GCs and, hence, in GC secretion. These changes have been ascribed largely to changes in the sensitivity of the limbic system, and possibly the hypothalamus, to GCs. Surprisingly, the possibility that early life stress/GC treatment may also exert irreversible effects at the pituitary level has scarcely been addressed. Accordingly, we have examined the effects of pre- and neonatal dexamethasone treatment on the adult male pituitary gland, focusing on the following: 1) the integrity of the acute annexin 1 (ANXA1)-dependent inhibitory actions of GCs on ACTH secretion, a process requiring ANXA1 release from folliculostellate (FS) cells; and 2) the morphology of FS cells and corticotrophs. Dexamethasone was given to pregnant (d 16-19) or lactating (d 1-7 postpartum) rats via the drinking water (1 microg/ml); controls received normal drinking water. Pituitary tissue from the offspring was examined ex vivo at d 90. Both treatment regimens reduced ANXA1 expression, as assessed by Western blotting and quantitative immunogold labeling. In particular, the amount of ANXA1 located on the outer surface of the FS cells was reduced. By contrast, IL-6 expression was increased, particularly by the prenatal treatment. Pituitary tissue from untreated control rats responded to dexamethasone with an increase in cell surface ANXA1 and a reduction in forskolin-induced ACTH release. In contrast, pituitary tissue from rats treated prenatally or neonatally with dexamethasone was unresponsive to the steroid, although, like control tissue, it responded readily to ANXA1, which readily inhibited forskolin-driven ACTH release. Prenatal dexamethasone treatment reduced the size but not the number of FS cells. It also caused a marked reduction in corticotroph number and impaired granule margination without affecting other aspects of corticotroph morphology. Similar but less marked effects on pituitary cell morphology and number were evident in tissue from neonatally treated rats. Our study shows that, when administered by a noninvasive process, perinatal GC treatment exerts profound effects on the adult pituitary gland, impairing the ANXA1-dependent GC regulation of ACTH release and altering the cell profile and morphology.


Assuntos
Animais Recém-Nascidos , Anexina A1/metabolismo , Dexametasona/farmacologia , Glucocorticoides/farmacologia , Adeno-Hipófise/citologia , Adeno-Hipófise/fisiologia , Efeitos Tardios da Exposição Pré-Natal , Caracteres Sexuais , Hormônio Adrenocorticotrópico/antagonistas & inibidores , Hormônio Adrenocorticotrópico/metabolismo , Animais , Anexina A1/antagonistas & inibidores , Western Blotting , Colforsina/farmacologia , Feminino , Imuno-Histoquímica , Interleucina-6/metabolismo , Masculino , Microscopia Eletrônica , Adeno-Hipófise/efeitos dos fármacos , Adeno-Hipófise/metabolismo , Gravidez , Ratos , Distribuição Tecidual
18.
J Neuroendocrinol ; 15(12): 1134-43, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14636175

RESUMO

Annexin 1 (ANXA1) is a key mediator of the inhibitory effects of glucocorticoids on adrenocorticotropic hormone (ACTH) release, which develop within 1-2 h of a steroid challenge. Our previous studies, which showed that (i) ANXA1 is expressed principally by the nonsecretory folliculo-stellate cells in the pituitary gland; (ii) glucocorticoids cause the exportation of ANXA1 from these cells; and (iii) corticotrophs express specific ANXA1 binding sites, led us to propose that ANXA1 serves as a paracrine or juxtacrine mediator of glucocorticoids. To address this hypothesis, we examined ANXA1-dependent glucocorticoid actions in co-cultures of murine corticotroph (AtT20 clone D1) and folliculo-stellate (TtT/GF) cell lines. ANXA1 mRNA and protein were found in abundance in TtT/GF cells but neither was detectable in the AtT20 cells. AtT20 cells (alone and in co-culture with TtT/GF cells) responded to corticotropin-releasing hormone (CRH) (0.1-1 micro m) with increased ACTH release. The CRH-stimulated release of ACTH from AtT20 cells cultured alone was unaffected by preincubation with dexamethasone (Dex, 100 nm); by contrast, in co-cultures of AtT20 and TtT/GF cells, the steroid readily inhibited the secretory response to CRH. The effects of Dex on ACTH release were mimicked by N-terminal ANXA1 fragments (ANXA1Ac2-26, 2 micro g/ml and ANXA11-188, 0.1 ng/ml) and reversed by mifepristone (1 micro m) and by an antisense oligodeoxynucleotide (ODN) to ANXA1 (50 nm) but not by control ODNs. The antisense ODN also specifically blocked the Dex-induced externalization of ANXA1 from TtT/GF cells. Immunofluorescence imaging of the co-cultures localized the exported protein to the vicinity of the AtT20 cells and identified ANXA1 binding sites on these cells. These results provide functional and histological evidence to support our premise that the early inhibitory effects of glucocorticoids on ACTH release are dependent upon paracrine/juxtacrine actions of ANXA1 derived from folliculo-stellate cells.


Assuntos
Hormônio Adrenocorticotrópico/metabolismo , Anexina A1/metabolismo , Dexametasona/farmacologia , Glucocorticoides/farmacologia , Hipófise/citologia , Hipófise/metabolismo , Neoplasias Hipofisárias , Animais , Anexina A1/genética , Linhagem Celular Tumoral , Técnicas de Cocultura , Antagonistas de Hormônios/farmacologia , Mifepristona/farmacologia , Comunicação Parácrina/efeitos dos fármacos , Comunicação Parácrina/fisiologia , RNA Mensageiro/análise
19.
J Neuroendocrinol ; 15(10): 946-57, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12969239

RESUMO

Our previous studies have identified a role for annexin 1 (ANXA1), a protein produced by the pituitary folliculostellate cells, as a paracrine/juxtacrine mediator of the acute regulatory effects of glucocorticoids on the release of adrenocorticotropic hormone and other pituitary hormones. In the present study, we focused on the secretion of thyroid stimulating hormone (TSH) and luteinizing hormone (LH) and used a battery of ANXA1-derived peptides to identify the key domains in the ANXA1 molecule that are critical to the inhibition of peptide release. In addition, as ANXA1 is a substrate for protein kinase C (PKC) and tyrosine kinase, we examined the roles of these kinases in the manifestation of the ANXA1-dependent inhibitory actions of dexamethasone on TSH and LH release. Dexamethasone suppressed the forskolin-induced release of TSH and LH from rat anterior pituitary tissue in vitro. Its effects were mimicked by human recombinant ANXA1 (hrANXA1) and a truncated protein, ANXA1(1-188). ANXA1(Ac2-26), also suppressed stimulated peptide release but it lacked both the potency and the efficacy of the parent protein. Shorter N-terminal ANXA1 sequences were without effect. The PKC inhibitor PKC(19-36) abolished the inhibitory actions of dexamethasone on the forskolin-evoked release of TSH and LH; it also attenuated the inhibitory actions of ANXA1(Ac2-26). Similar effects were produced by annexin 5 (ANXA5) which sequesters PKC in other systems. By contrast, the tyrosine kinase inhibitors, p60v-src (137-157) and genistein, had no effect on the secretion of TSH or LH alone or in the presence of forskolin and/or dexamethasone. Dexamethasone caused the translocation of a tyrosine-phosphorylated species of ANXA1 to the surface of pituitary cells. The total amount of ANXA1 exported from the cells in response to the steroid was unaffected by tyrosine kinase blockade. However, the degree of tyrosine-phosphorylation of the exported protein was markedly reduced by genistein. These results suggest that (i) the ANXA1-dependent inhibitory actions of dexamethasone on the release of TSH and LH require PKC and sequences in the N-terminal domain of ANXA1, but are independent of tyrosine kinase, and (ii) while dexamethasone induces the cellular exportation of a tyrosine-phosphorylated species of ANXA1, tyrosine phosphorylation per se is not critical to the steroid-induced passage of ANXA1 across the membrane.


Assuntos
Anexina A1/farmacologia , Glucocorticoides/farmacologia , Hormônio Luteinizante/metabolismo , Fosfotransferases/fisiologia , Tireotropina/metabolismo , Sequência de Aminoácidos , Animais , Anexina A5/farmacologia , Western Blotting , Colforsina/farmacologia , Eletroforese em Gel de Poliacrilamida , Técnicas In Vitro , Masculino , Proteínas de Membrana/química , Dados de Sequência Molecular , Peptídeos/farmacologia , Fosforilação , Fosfotransferases/antagonistas & inibidores , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , Adeno-Hipófise/efeitos dos fármacos , Adeno-Hipófise/metabolismo , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/metabolismo , Radioimunoensaio , Ratos , Ratos Sprague-Dawley
20.
Arch Physiol Biochem ; 110(1-2): 54-61, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11935401

RESUMO

Prolactin secretion is controlled by the hypothalamus, and by circulating steroids; oestrogens stimulate, but glucocorticoids inhibit prolactin release. Lactotrophs express intracellular receptors for oestrogens, but apparently not glucocorticoids. Therefore, a genomic effect of oestrogens could be direct, but that of glucocorticoids appears to be indirect. Lactotrophs are not a homogeneous cell population: some have large irregular dense-cored vesicles, others have small round vesicles, but the functional significance of this inhomogeneity is far from clear. Oestradiol and testosterone can stimulate rapid release of prolactin selectively from type II lactotrophs characterised by small round vesicles. Progesterone and other steroids do not exert this effect, which results from a non-genomic action of oestradiol and testosterone. Glucocorticoid inhibition of secretagogue-induced prolactin secretion is mimicked by annexin 1 (lipocortin 1), a protein induced by glucocorticoids in the pituitary and many other tissues, and can be blocked by annexin 1 immunoneutralisation and antisense. Glucocorticoid inhibition of ACTH and growth hormone secretion also involves annexin 1. Pituitary annexin 1 is located in folliculo-stellate cells; these express glucocorticoid receptors, and glucocorticoids induce annexin-1 synthesis. Annexin 1 is externalised from folliculo-stellate cells in response to glucocorticoids, despite the fact that it lacks a secretory signal sequence and is not packaged in vesicles. Inhibition of annexin 1 externalisation by glyburide suggests involvement of an ABC (ATP-binding cassette) transporter in externalisation. Both oestradiol and glucocorticoids therefore influence the secretion of prolactin by novel direct and indirect mechanisms, in addition to their much better understood effects on transcription via classical intracellular steroid receptors.


Assuntos
Anexina A1/fisiologia , Esteroides/fisiologia , Animais , Estradiol/metabolismo , Glucocorticoides/fisiologia , Glibureto/metabolismo , Imuno-Histoquímica , Adeno-Hipófise/citologia , Adeno-Hipófise/metabolismo , Prolactina/metabolismo , Prolactina/fisiologia , Ratos , Receptores de Estrogênio/metabolismo , Receptores de Glucocorticoides/fisiologia , Receptores de Esteroides/biossíntese , Receptores de Esteroides/fisiologia , Testosterona/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...