Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 181
Filtrar
1.
Endocrine ; 84(2): 345-349, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38400880

RESUMO

PURPOSE: Disorders/differences of sex development (DSD) result from variants in many different human genes but, frequently, have no detectable molecular cause. METHODS: Detailed clinical and genetic phenotyping was conducted on a family with three children. A Sec31a animal model and functional studies were used to investigate the significance of the findings. RESULTS: By trio whole-exome DNA sequencing we detected a heterozygous de novo nonsense SEC31A variant, in three children of healthy non-consanguineous parents. The children had different combinations of disorders that included complete gonadal dysgenesis and multiple pituitary hormone deficiency. SEC31A encodes a component of the COPII coat protein complex, necessary for intracellular anterograde vesicle-mediated transport between the endoplasmic reticulum (ER) and Golgi. CRISPR-Cas9 targeted knockout of the orthologous Sec31a gene region resulted in early embryonic lethality in homozygous mice. mRNA expression of ER-stress genes ATF4 and CHOP was increased in the children, suggesting defective protein transport. The pLI score of the gene, from gnomAD data, is 0.02. CONCLUSIONS: SEC31A might underlie a previously unrecognised clinical syndrome comprising gonadal dysgenesis, multiple pituitary hormone deficiencies, dysmorphic features and developmental delay. However, a variant that remains undetected, in a different gene, may alternatively be causal in this family.


Assuntos
Hipopituitarismo , Animais , Humanos , Feminino , Masculino , Camundongos , Hipopituitarismo/genética , Hipopituitarismo/metabolismo , Disgenesia Gonadal/genética , Proteínas de Transporte Vesicular/genética , Linhagem , Criança , Camundongos Knockout , Pré-Escolar , Hormônios Hipofisários/deficiência , Hormônios Hipofisários/genética
2.
Hum Mol Genet ; 32(3): 367-385, 2023 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-35951005

RESUMO

Congenital hypopituitarism is a genetically heterogeneous condition that is part of a spectrum disorder that can include holoprosencephaly. Heterozygous mutations in SIX3 cause variable holoprosencephaly in humans and mice. We identified two children with neonatal hypopituitarism and thin pituitary stalk who were doubly heterozygous for rare, likely deleterious variants in the transcription factors SIX3 and POU1F1. We used genetically engineered mice to understand the disease pathophysiology. Pou1f1 loss-of-function heterozygotes are unaffected; Six3 heterozygotes have pituitary gland dysmorphology and incompletely ossified palate; and the Six3+/-; Pou1f1+/dw double heterozygote mice have a pronounced phenotype, including pituitary growth through the palate. The interaction of Pou1f1 and Six3 in mice supports the possibility of digenic pituitary disease in children. Disruption of Six3 expression in the oral ectoderm completely ablated anterior pituitary development, and deletion of Six3 in the neural ectoderm blocked the development of the pituitary stalk and both anterior and posterior pituitary lobes. Six3 is required in both oral and neural ectodermal tissues for the activation of signaling pathways and transcription factors necessary for pituitary cell fate. These studies clarify the mechanism of SIX3 action in pituitary development and provide support for a digenic basis for hypopituitarism.


Assuntos
Holoprosencefalia , Hipopituitarismo , Criança , Humanos , Heterozigoto , Hipopituitarismo/genética , Fatores de Transcrição/genética , Mutação , Hormônios Hipofisários/genética , Fator de Transcrição Pit-1/genética
3.
Front Endocrinol (Lausanne) ; 13: 1008306, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36237189

RESUMO

Combined pituitary hormone deficiency (CPHD) is not a rare disorder, with a frequency of approximately 1 case per 4,000 live births. However, in most cases, a genetic diagnosis is not available. Furthermore, the diagnosis is challenging because no clear correlation exists between the pituitary hormones affected and the gene(s) responsible for the disorder. Next-generation sequencing (NGS) has recently been widely used to identify novel genes that cause (or putatively cause) CPHD. This review outlines causative genes for CPHD that have been newly reported in recent years. Moreover, novel variants of known CPHD-related genes (POU1F1 and GH1 genes) that contribute to CPHD through unique mechanisms are also discussed in this review. From a clinical perspective, variants in some of the recently identified causative genes result in extra-pituitary phenotypes. Clinical research on the related symptoms and basic research on pituitary formation may help in inferring the causative gene(s) of CPHD. Future NGS analysis of a large number of CPHD cases may reveal new genes related to pituitary development. Clarifying the causative genes of CPHD may help to understand the process of pituitary development. We hope that future innovations will lead to the identification of genes responsible for CPHD and pituitary development.


Assuntos
Hipopituitarismo , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Hipopituitarismo/diagnóstico , Hipopituitarismo/genética , Hormônios Hipofisários/genética , Fatores de Transcrição/genética
4.
PLoS Genet ; 16(12): e1009244, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33301440

RESUMO

The genetic origin of human skin pigmentation remains an open question in biology. Several skin disorders and diseases originate from mutations in conserved pigmentation genes, including albinism, vitiligo, and melanoma. Teleosts possess the capacity to modify their pigmentation to adapt to their environmental background to avoid predators. This background adaptation occurs through melanosome aggregation (white background) or dispersion (black background) in melanocytes. These mechanisms are largely regulated by melanin-concentrating hormone (MCH) and α-melanocyte-stimulating hormone (α-MSH), two hypothalamic neuropeptides also involved in mammalian skin pigmentation. Despite evidence that the exogenous application of MCH peptides induces melanosome aggregation, it is not known if the MCH system is physiologically responsible for background adaptation. In zebrafish, we identify that MCH neurons target the pituitary gland-blood vessel portal and that endogenous MCH peptide expression regulates melanin concentration for background adaptation. We demonstrate that this effect is mediated by MCH receptor 2 (Mchr2) but not Mchr1a/b. mchr2 knock-out fish cannot adapt to a white background, providing the first genetic demonstration that MCH signaling is physiologically required to control skin pigmentation. mchr2 phenotype can be rescued in adult fish by knocking-out pomc, the gene coding for the precursor of α-MSH, demonstrating the relevance of the antagonistic activity between MCH and α-MSH in the control of melanosome organization. Interestingly, MCH receptor is also expressed in human melanocytes, thus a similar antagonistic activity regulating skin pigmentation may be conserved during evolution, and the dysregulation of these pathways is significant to our understanding of human skin disorders and cancers.


Assuntos
Hormônios Hipotalâmicos/metabolismo , Melaninas/metabolismo , Hormônios Hipofisários/metabolismo , Pigmentação da Pele/genética , Animais , Hormônios Hipotalâmicos/genética , Hipotálamo/citologia , Hipotálamo/metabolismo , Melaninas/genética , Hormônios Estimuladores de Melanócitos/genética , Hormônios Estimuladores de Melanócitos/metabolismo , Melanócitos/metabolismo , Neurônios/metabolismo , Hormônios Hipofisários/genética , Peixe-Zebra
5.
Zoolog Sci ; 37(6): 563-574, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33269872

RESUMO

Amphibians exhibit phenotypic plasticity, which allows flexible adaptation to fluctuating environments. Although genes involved in expression of plastic phenotypes have been identified, the endocrine bases of plastic responses are largely unknown. Larvae of the Hokkaido salamander (Hynobius retardatus) plastically display distinct phenotypes, an "offensive phenotype" characterized as larger body with broadened gape and a "defensive phenotype" characterized as enlarged gills and tail and less active behavior, in the presence of prey larval amphibians and predatory larval dragonfly, respectively. In the presence of both prey and predators, the degree of induction of both phenotypes is reduced, suggesting cross-talk between the molecular signaling pathways of these phenotypes. We conducted a transcriptomic analysis to examine how endocrine regulation affects the phenotypic expression by focusing on the pituitary gland. We found that five endocrine genes, i.e., calcitonin related polypeptide alpha (CALCA), growth hormone (GH), neuropeptide B (NPB), parathyroid hormone 2 (PTH2), and prolactin 1 (PRL1), were involved in the expression of both phenotypes. However, we conducted only RNA-seq analysis, and no confirmation of significant up-regulation or down-regulation has been conducted. These results suggest that these genes were up-regulated for induction of the offensive phenotype and down-regulated for induction of the defensive phenotype. Phylogenetic analysis indicated that possible gene duplications of PRL and CALCA have occurred during amphibian evolution. Based on these findings, it is suggested that a trade-off of molecular signaling pathways exists between the two distinct phenotypic expressions. The results also suggest that hormonal-gene duplications might have contributed to the acquisition of phenotypic plasticity in amphibians.


Assuntos
Adaptação Fisiológica/genética , Comportamento Predatório , Urodelos/genética , Animais , Perfilação da Expressão Gênica , Larva/genética , Larva/metabolismo , Odonatos , Filogenia , Hipófise/fisiologia , Hormônios Hipofisários/genética , Hormônios Hipofisários/metabolismo , Ranidae , Urodelos/crescimento & desenvolvimento , Urodelos/metabolismo
6.
Zhongguo Ying Yong Sheng Li Xue Za Zhi ; 36(3): 245-249, 2020 May.
Artigo em Chinês | MEDLINE | ID: mdl-32981280

RESUMO

Objective: To explore the effects of repeated immobilization stress on hypothalamic-pituitary-ovarian axis in female rats. Methods: Forty female SD rats were randomly divided into two groups: control group (n=20) and experimental group (n=20). One group was fed normally, the other group was subjected to incremental load restraint stress. Brake stress once a day in the retainer (starting at 9: 00 a.m.), braking for 2 hours on the first day, increasing load by 0.5 hours a day for two weeks. Body weight, estrous cycle, sex hormone, organ coefficient, pathology and expression of related genes were detected to explore the harm of hypothalamic-pituitary-ovarian axis. Results: Repeated immobilization stress caused weight loss, prolonged estrous cycle, and changed the organ coefficient and morphology of ovaries and uterus. QPCR technique was used to detect the related genes. It was found that the expressions of gonadotropin releasing hormone, pituitary gonadotropin releasing hormone receptor, follicle stimulating hormone and luteinizing hormone mRNA were decreased significantly, while the expressions of ovarian follicle stimulating hormone and luteinizing hormone receptor mRNA were increased significantly. The expression of estrogen receptor mRNA in ovary and uterus was decreased significantly. Conclusion: Repeated immobilization stress may disrupt the estrous cycle by interfering with the endocrine regulation of the hypothalamic-pituitary-ovarian axis, thus damaging the gonadal and reproductive endocrine function of female animals.


Assuntos
Regulação da Expressão Gênica , Hipotálamo , Imobilização , Ovário , Hipófise , Hormônios Hipofisários , Estresse Fisiológico , Animais , Feminino , Hormônio Foliculoestimulante/genética , Regulação da Expressão Gênica/fisiologia , Hormônio Liberador de Gonadotropina/genética , Hipotálamo/fisiopatologia , Imobilização/fisiologia , Imobilização/psicologia , Hormônio Luteinizante/genética , Ovário/fisiopatologia , Hipófise/fisiopatologia , Hormônios Hipofisários/genética , Ratos , Ratos Sprague-Dawley
7.
Mol Cell Endocrinol ; 513: 110858, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32413385

RESUMO

Kisspeptin2 is a neuropeptide widely found in the brain and multiple peripheral tissues in the zebrafish. The pituitary is the center of synthesis and secretes various endocrine hormones. However, Kiss2 innervation in the zebrafish pituitary is unknown. In this study, the organization of Kiss2 cells and structures in the zebrafish pituitary by promoter-driving mCherry-labeling Kiss2 neurons were investigated. Kiss2 neurons in the hypothalamus do not project into the pituitary. Kiss2 cells are found in the female pituitary. Unidentified Kiss2 cells and extensions are located in the proximal pars distalis (PPD), similar to the distribution of Gnrh3 fibers. Kiss2 structures reside alongside Gnrh3 fibers. No Kiss2 structures are found in the male pituitary. The transcriptional expression of the kisspeptin receptor kiss1rb is detected in both female and male pituitaries. In situ hybridization shows that kiss1rb-positive cells are located in the PPD and pars intermedia (PI). In vitro Kiss2-10 treatment stimulates Akt and Erk phosphorylation and significantly induces lhß, fshß, and prl1 mRNA expression in the female pituitary. The results in this study suggest that Kiss2 and Kiss1rb may form an independent paracrine or autocrine system in the female zebrafish pituitary. Kiss2 and Kiss1rb signaling regulates the expression of pituitary hormones.


Assuntos
Kisspeptinas/fisiologia , Hipófise/metabolismo , Hormônios Hipofisários/genética , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra , Animais , Animais Geneticamente Modificados , Células Cultivadas , Feminino , Subunidade beta do Hormônio Folículoestimulante/genética , Subunidade beta do Hormônio Folículoestimulante/metabolismo , Regulação da Expressão Gênica , Hormônio Luteinizante Subunidade beta/genética , Hormônio Luteinizante Subunidade beta/metabolismo , Hormônios Hipofisários/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
8.
Front Endocrinol (Lausanne) ; 11: 629077, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33613457

RESUMO

Glucagon (GCG) plays a stimulatory role in pituitary hormone regulation, although previous studies have not defined the molecular mechanism whereby GCG affects pituitary hormone secretion. To this end, we identified two distinct proglucagons, Gcga and Gcgb, as well as GCG receptors, Gcgra and Gcgrb, in Nile tilapia (Oreochromis niloticus). Using the cAMP response element (CRE)-luciferase reporter system, tilapia GCGa and GCGb could reciprocally activate the two GCG receptors expressed in human embryonic kidney 293 (HEK293) cells. Quantitative real-time PCR analysis revealed that differential expression of the Gcga and Gcgb and their cognate receptors Gcgra and Gcgrb was found in the various tissues of tilapia. In particular, the Gcgrb is abundantly expressed in the neurointermediate lobe (NIL) of the pituitary gland. In primary cultures of tilapia NIL cells, GCGb effectively stimulated SL release, with parallel rises in the mRNA levels, and co-incubation with the GCG antagonist prevented GCGb-stimulated SL release. In parallel experiments, GCGb treatment dose-dependently enhanced intracellular cyclic adenosine monophosphate (cAMP) accumulation with increasing inositol 1,4,5-trisphosphate (IP3) concentration and the resulting in transient increases of Ca2+ signals in the primary NIL cell culture. Using selective pharmacological approaches, the adenylyl cyclase (AC)/cAMP/protein kinase A (PKA) and phospholipase C (PLC)/IP3/Ca2+/calmodulin (CaM)/CaMK-II pathways were shown to be involved in GCGb-induced SL release and mRNA expression. Together, these results provide evidence for the first time that GCGb can act at the pituitary level to stimulate SL release and gene expression via GCGRb through the activation of the AC/cAMP/PKA and PLC/IP3/Ca2+/CaM/CaMK-II cascades.


Assuntos
Proteínas de Peixes/biossíntese , Glucagon/metabolismo , Glucagon/farmacologia , Hipófise/metabolismo , Hormônios Hipofisários/biossíntese , Transdução de Sinais/fisiologia , Sequência de Aminoácidos , Animais , Células Cultivadas , Ciclídeos , Proteínas de Peixes/genética , Expressão Gênica , Células HEK293 , Humanos , Hipófise/citologia , Hormônios Hipofisários/genética , Transdução de Sinais/efeitos dos fármacos
9.
Anim Reprod Sci ; 203: 1-9, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30797596

RESUMO

Results of previous studies indicated the existence of LPXRFa, the piscine ortholog of gonadotropin-inhibitory hormone (GnIH), and kisspeptin (Kiss2) in tongue sole (Cynoglossus semilaevis), and that LPXRFa exerts an inhibitory effect on Kiss2 activation in the protein kinase A (PKA) pathway. The functions in the control of reproduction and whether LPXRFa antagonizes the action of Kiss2 by inhibiting the protein kinase C (PKC) pathway, however, are still unknown. In the present study, there was an initial investigation of the direct effects of LPXRFa and Kiss2 on relative abundance of pituitary hormone mRNA transcripts using a whole pituitary culture system. Results indicated that LPXRFa-1 specifically functioned to increase relative abundance of lhß mRNA when there were comparisons with the control, without any effect on relative abundance of gh, gthα and fshß mRNA. Treatment with LPXRFa-2 resulted in a reduction in relative abundance of gthα and lhß mRNA, and did not alter relative abundance of fshß mRNA. Treatment of LPXRFa-2 resulted in a greater relative abundance of gh mRNA. Treatment with Kiss2, however, resulted in an increase in relative abundance of gthα and fshß mRNA transcripts, without altering relative abundances of gh and lhß mRNA. Subsequently, there was valuation of the potential interaction between LPXRFa and kisspeptin in COS-7 cells transfected with the cognate receptors. Both LPXRFa-1 and LPXRFa-2 suppressed serum responsive element-dependent luciferase (SRE-luc) activity when compared to stimulation with Kiss2 alone, indicating an inhibitory effect of LPXRFa on kisspeptin activation on the PKC pathway. Overall, data from the present study provide novel evidence for differential actions of LPXRFa and kisspeptin on pituitary hormone synthesis as well as for the interaction between LPXRFa and kisspeptin systems in teleosts.


Assuntos
Proteínas de Peixes/metabolismo , Linguados/fisiologia , Hormônio Liberador de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Hormônios Hipofisários/metabolismo , Proteína Quinase C/metabolismo , RNA Mensageiro/metabolismo , Animais , Células COS , Chlorocebus aethiops , Proteínas de Peixes/genética , Regulação da Expressão Gênica , Hormônio Liberador de Gonadotropina/genética , Kisspeptinas/genética , Hipófise/citologia , Hipófise/metabolismo , Hormônios Hipofisários/genética , Proteína Quinase C/genética , RNA Mensageiro/genética , Transdução de Sinais
10.
Int J Mol Sci ; 20(1)2018 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-30587833

RESUMO

Epidermal growth factor (EGF) is a potent regulator of cell function in many cell types. In mammals, the EGF/EGFR system played an important role in both pituitary physiology and pathology. However, it is not clear about the pituitary action of EGF in lower vertebrates. In this study, using grass carp as a model, we found that EGF could stimulate NK3R mRNA and protein expression through pituitary ErbB1 and ErbB2 coupled to MEK/ERK and PI3K/Akt/mTOR pathways. In addition, EGF could also induce pituitary somatolactin α (SLα) secretion and mRNA expression in a dose- and time-dependent manner in vivo and in vitro. The stimulatory actions of EGF on SLα mRNA expression were also mediated by PI3K/Akt/mTOR and MEK/ERK pathways coupled to ErbB1 and ErbB2 activation. Our previous study has reported that neurokinin B (NKB) could also induce SLα secretion and mRNA expression in carp pituitary cells. In the present study, interestingly, we found that EGF could significantly enhance NKB-induced SLα mRNA expression. Further studies found that NK3R antagonist SB222200 could block EGF-induced SLα mRNA expression, indicating an NK3R requirement. Furthermore, cAMP/PKA inhibitors and PLC/PKC inhibitors could both abolish EGF- and EGF+NKB-induced SLα mRNA expression, which further supported that EGF-induced SLα mRNA expression is NK3R dependent.


Assuntos
Carpas/metabolismo , Fator de Crescimento Epidérmico/farmacologia , Proteínas de Peixes/metabolismo , Hipófise/efeitos dos fármacos , Hormônios Hipofisários/metabolismo , Receptores da Neurocinina-3/metabolismo , Animais , AMP Cíclico/metabolismo , Sinergismo Farmacológico , Receptores ErbB/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas de Peixes/genética , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Hipófise/metabolismo , Hormônios Hipofisários/genética , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , RNA Mensageiro/metabolismo , Receptores da Neurocinina-3/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Fosfolipases Tipo C/antagonistas & inibidores , Fosfolipases Tipo C/metabolismo
11.
Neurobiol Dis ; 120: 12-20, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30149182

RESUMO

The lateral hypothalamus contains neurons producing orexins that promote wakefulness and suppress REM sleep as well as neurons producing melanin-concentrating hormone (MCH) that likely promote REM sleep. Narcolepsy with cataplexy is caused by selective loss of the orexin neurons, and the MCH neurons appear unaffected. As the orexin and MCH systems exert opposing effects on REM sleep, we hypothesized that imbalance in this REM sleep-regulating system due to activity in the MCH neurons may contribute to the striking REM sleep dysfunction characteristic of narcolepsy. To test this hypothesis, we chemogenetically activated the MCH neurons and pharmacologically blocked MCH signaling in a murine model of narcolepsy and studied the effects on sleep-wake behavior and cataplexy. To chemoactivate MCH neurons, we injected an adeno-associated viral vector containing the hM3Dq stimulatory DREADD into the lateral hypothalamus of orexin null mice that also express Cre recombinase in the MCH neurons (MCH-Cre::OX-KO mice) and into control MCH-Cre mice with normal orexin expression. In both lines of mice, activation of MCH neurons by clozapine-N-oxide (CNO) increased rapid eye movement (REM) sleep without altering other states. In mice lacking orexins, activation of the MCH neurons also increased abnormal intrusions of REM sleep manifest as cataplexy and short latency transitions into REM sleep (SLREM). Conversely, a MCH receptor 1 antagonist, SNAP 94847, almost completely eliminated SLREM and cataplexy in OX-KO mice. These findings affirm that MCH neurons promote REM sleep under normal circumstances, and their activity in mice lacking orexins likely triggers abnormal intrusions of REM sleep into non-REM sleep and wake, resulting in the SLREM and cataplexy characteristic of narcolepsy.


Assuntos
Hormônios Hipotalâmicos/metabolismo , Melaninas/metabolismo , Narcolepsia/metabolismo , Neurônios/fisiologia , Hormônios Hipofisários/metabolismo , Sono REM/fisiologia , Animais , Feminino , Hormônios Hipotalâmicos/antagonistas & inibidores , Hormônios Hipotalâmicos/genética , Masculino , Melaninas/antagonistas & inibidores , Melaninas/genética , Camundongos , Camundongos Knockout , Narcolepsia/genética , Neurônios/efeitos dos fármacos , Piperidinas/farmacologia , Hormônios Hipofisários/antagonistas & inibidores , Hormônios Hipofisários/genética
12.
PLoS One ; 13(7): e0198877, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29979686

RESUMO

The aim of the present study is to check whether we can replicate, in an independent series, previous results showing that the molecular study of pituitary-specific gene expression complements the inmunohistochemical identification of pituitary neuroendocrine tumours. We selected 112 patients (51 (46.4%) women; mean age 51.4±16 years; 102 macroadenomas (91.9%), 9 microadenomas (8.1%)) with complete clinical, radiological, immunohistochemical and molecular data from our data set of pituitary neuroendocrine tumours. Patients were different from those previously studied. We measured the expression of the pituitary-specific hormone genes and type 1 corticotrophin-releasing hormone and arginine vasopressin 1b receptors, by quantitative real-time polymerase chain reaction using TaqMan probes. Afterwards, we identified the different pituitary neuroendocrine tumour subtypes following the 2017 World Health Organization classification of pituitary tumours, calculating the concordance between their molecular and immuhistochemical identification. The concordance between molecular and immunohistochemical identification of functioning pituitary neuroendocrine tumours with the clinical diagnosis was globally similar to the previous series, where the SYBR Green technique was used instead of TaqMan probes. Our results also corroborated the poor correlation between molecular and immunohistochemical detection of the silent pituitary neuroendocrine tumour variants. This discrepancy was more remarkable in lactotroph, null-cell and plurihormonal pituitary neuroendocrine tumours. In conclusion, this study validates the results previously published by our group, highlighting a complementary role for the molecular study of the pituitary-specific hormone genes in the typification of pituitary neuroendocrine tumours subtypes.


Assuntos
Tumores Neuroendócrinos/diagnóstico , Hormônios Hipofisários/genética , Neoplasias Hipofisárias/diagnóstico , Receptores de Hormônio Liberador da Corticotropina/genética , Receptores de Vasopressinas/genética , Adulto , Idoso , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Tumores Neuroendócrinos/genética , Tumores Neuroendócrinos/patologia , Hipófise/patologia , Neoplasias Hipofisárias/genética , Neoplasias Hipofisárias/patologia
13.
ACS Chem Neurosci ; 9(6): 1235-1246, 2018 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-29714060

RESUMO

Many physiological pathways are involved in appetite, food intake, and the maintenance of energy homeostasis. In particular, neuropeptides within the central nervous system have been demonstrated to be critical signaling molecules for modulating appetite. Both anorexigenic (appetite-decreasing) and orexigenic (appetite-stimulating) neuropeptides have been described. The biological effects of these neuropeptides can be observed following central administration in animal models. This review focuses on single nucleotide polymorphisms (SNPs) in six orexigenic neuropeptides: agouti-related protein (AGRP), galanin, melanin concentrating hormone (MCH), neuropeptide Y (NPY), orexin A, and orexin B. Following a brief summary of the neuropeptides and their orexigenic activities, reports associating SNPs within the orexigenic neuropeptides to energy homeostasis, food intake, obesity, and BMI in humans are reviewed. Additionally, the NIH tool Variation Viewer was utilized to identify missense SNPs within the mature, biologically active neuropeptide sequences. For SNPs found through Variation Viewer, a concise discussion on relevant pharmacological structure-activity relationship studies for select SNPs is included. This review is meant to update reported orexigenic neuropeptide SNPs and demonstrate the potential utility of genomic sequence databases for finding SNPs that may result in altered receptor signaling for neuropeptide pathways associated with appetite.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/genética , Neuropeptídeos/genética , Polimorfismo de Nucleotídeo Único/genética , Receptores Acoplados a Proteínas G/genética , Animais , Humanos , Hormônios Hipotalâmicos/genética , Hormônios Hipotalâmicos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Melaninas/genética , Melaninas/metabolismo , Neuropeptídeos/metabolismo , Orexinas/genética , Orexinas/metabolismo , Hormônios Hipofisários/genética , Hormônios Hipofisários/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
14.
Fish Physiol Biochem ; 44(3): 983-995, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29550894

RESUMO

Using reverse transcription-polymerase chain reaction (RT-PCR) and RACE (rapid amplification of cDNA ends), somatolactin-α (rmSLα) and -ß (rmSLß) were identified from the pituitary gland of rare minnows (Gobiocypris rarus). The full-length cDNAs of these two genes were 1288 and 801 bp, encoding prepeptides of 250 and 228 amino acids residues, respectively. rmSLß can be detected in the brain (including the pituitary), ovary, testis, and gill, while rmSLα was mainly expressed in the brain. On the other hand, rmSLα was expressed in all the fetal developmental stages; however, rmSLß can just be detected in the stages since from 14 h post-fertilization (hpf). After exposure to acute waterborne cadmium (Cd), rmSLα was distinctly upregulated in juvenile rare minnows at all detected time points, from 24 to 96 h and 10 days, while rmSLß was significantly altered only in 96 h or 10-day treatment groups. As for adults, acute Cd exposure caused alterations of both rmSLα and rmSLß in the brain (containing the pituitary) at the 24 h; subchronic waterborne Cd treatment led to upregulation of rmSLα, while decrease of mSLß in the brain. Alteration of rmSL transcripts following waterborne Cd exposure further confirmed the endocrine disruption of this heavy metal. Besides, exposure to as low as 5 µg/L Cd caused alteration of rmSLα, which suggested that rmSLα might be a potential biomarker for risk assessment of aquatic Cd.


Assuntos
Cádmio/toxicidade , Cyprinidae/genética , Proteínas de Peixes/genética , Glicoproteínas/genética , Hormônios Hipofisários/genética , Poluentes Químicos da Água/toxicidade , Sequência de Aminoácidos , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , DNA Complementar/genética , Feminino , Brânquias/efeitos dos fármacos , Brânquias/metabolismo , Masculino , Ovário/efeitos dos fármacos , Ovário/metabolismo , Filogenia , Testículo/efeitos dos fármacos , Testículo/metabolismo
15.
Orv Hetil ; 159(7): 278-284, 2018 Feb.
Artigo em Húngaro | MEDLINE | ID: mdl-29429351

RESUMO

Developmental disorders affecting the hypothalamic-pituitary system can result in pituitary hormone deficiency showing a diverse clinical presentation. A significant majority of these disorders are closely linked to defects in transcription factor genes which play a major role in pituitary development. Those affecting the early phase of organogenesis typically lead to complex conditions affecting the pituitary as well as structures in the central nervous system. Transcription factors involved in the late phase can result in combined but rarely isolated pituitary hormone deficiency without extra-pituitary manifestation. Identifying the defects in these pituitary transcription factor genes may provide a useful tool in predicting disease progression as well as screening family members. Several pituitary transcription factors can be detected in the adult gland as well which is strongly emphasized in the World Health Organization's most recent guideline for pituitary tumor classification. Our review summarizes the current essential knowledge relevant for clinical endocrinologists. Orv Hetil. 2018; 159(7): 278-284.


Assuntos
Hipopituitarismo/genética , Sistema Hipotálamo-Hipofisário , Hormônios Hipofisários/genética , Hormônios Hipofisários/metabolismo , Análise Mutacional de DNA , Humanos , Hipófise/crescimento & desenvolvimento , Hipófise/metabolismo
16.
J Endocrinol ; 236(1): R1-R13, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28855316

RESUMO

Evidence for the presence of pituitary gland stem cells has been provided over the last decade using a combination of approaches including in vitro clonogenicity assays, flow cytometric side population analysis, immunohistochemical analysis and genetic approaches. These cells have been demonstrated to be able to self-renew and undergo multipotent differentiation to give rise to all hormonal lineages of the anterior pituitary. Furthermore, evidence exists for their contribution to regeneration of the organ and plastic responses to changing physiological demand. Recently, stem-like cells have been isolated from pituitary neoplasms raising the possibility that a cytological hierarchy exists, in keeping with the cancer stem cell paradigm. In this manuscript, we review the evidence for the existence of pituitary stem cells, their role in maintaining organ homeostasis and the regulation of their differentiation. Furthermore, we explore the emerging concept of stem cells in pituitary tumours and their potential roles in these diseases.


Assuntos
Transformação Celular Neoplásica , Células-Tronco Neoplásicas/patologia , Hipófise/citologia , Neoplasias Hipofisárias/patologia , Células-Tronco/citologia , Animais , Expressão Gênica , Homeostase , Humanos , Células-Tronco Neoplásicas/metabolismo , Hipófise/metabolismo , Hormônios Hipofisários/genética , Hormônios Hipofisários/metabolismo , Neoplasias Hipofisárias/genética , Neoplasias Hipofisárias/metabolismo , Células-Tronco/metabolismo
17.
Nat Neurosci ; 21(1): 29-32, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29180747

RESUMO

Excitation of accumbal D2 cells governs vital actions, including avoidance of learned risks, but the origins of this excitation and roles of D2 cells in innate risk-avoidance are unclear. Hypothalamic neurons producing orexins (also called hypocretins) enhance innate risk-avoidance via poorly understood neurocircuits. We describe a direct orexin→D2 excitatory circuit and show that D2 cell activity is necessary for orexin-dependent innate risk-avoidance in mice, thus revealing an unsuspected hypothalamus-accumbens interplay in action selection.


Assuntos
Aprendizagem da Esquiva/fisiologia , Instinto , Neurônios/fisiologia , Orexinas/metabolismo , Transdução de Sinais/fisiologia , 2-Amino-5-fosfonovalerato/farmacologia , 6-Ciano-7-nitroquinoxalina-2,3-diona/farmacologia , Animais , Channelrhodopsins/genética , Channelrhodopsins/metabolismo , Antagonistas de Aminoácidos Excitatórios/farmacologia , Hormônios Hipotalâmicos/genética , Hormônios Hipotalâmicos/metabolismo , Hipotálamo/citologia , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Melaninas/genética , Melaninas/metabolismo , Camundongos , Camundongos Transgênicos , Rede Nervosa/efeitos dos fármacos , Rede Nervosa/fisiologia , Neurônios/efeitos dos fármacos , Neuropeptídeo Y/genética , Neuropeptídeo Y/metabolismo , Orexinas/genética , Hormônios Hipofisários/genética , Hormônios Hipofisários/metabolismo , Receptor A2A de Adenosina/genética , Receptor A2A de Adenosina/metabolismo , Receptores de Dopamina D1/genética
18.
Artigo em Inglês | MEDLINE | ID: mdl-28821466

RESUMO

Evidence suggests that pharmaceuticals and personal care products reach urban watersheds, bioconcentrate in fish, and potentially disrupt physiological homeostasis. These impairments often affect hormone functions. Selective serotonin reuptake inhibitors (SRRIs) are increasingly studied with regards to their endocrine disrupting effects on teleost physiological processes, including reproduction. To examine whether FLX effects on the endocrine regulation of reproductive physiology in goldfish are sex-specific, we exposed sexually recrudescent female goldfish to two waterborne concentrations of FLX (0.54µg/L and 54µg/L) using an experimental design previously used for sexually mature male goldfish. To evaluate possible endocrine disrupting effects, we quantified the gonadosomatic index, circulating hormone concentrations (luteinizing hormone, LH; growth hormone, GH; 17-ß estradiol, E2; and testosterone, T), and the expression of isotocin and vasotocin in the telencephalon, gonadotropin subunits and GH in the pituitary, and gonadotropin receptors, GH receptors, and aromatase in the ovary. Female goldfish exposed to 0.54µg/L FLX exhibited a significant decrease in circulating E2, and conversely, a significant increase in circulating LH and ovarian aromatase mRNA levels, suggesting disruption of E2-mediated feedback on LH release. These results, when compared with those previously observed in males, reveal that waterborne exposure to environmentally relevant levels of FLX sex-specifically disrupts the reproductive endocrine axis in goldfish, characterized by a decrease in E2 in females, and conversely, estrogen-like effects in males. These data emphasize the importance of studying the effect of endocrine disrupting chemicals on both sexes.


Assuntos
Fluoxetina/toxicidade , Carpa Dourada/fisiologia , Ovário/efeitos dos fármacos , Inibidores Seletivos de Recaptação de Serotonina/toxicidade , Animais , Aromatase/genética , Aromatase/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Ovário/fisiologia , Hormônios Hipofisários/genética , Hormônios Hipofisários/metabolismo , Subunidades Proteicas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Maturidade Sexual/fisiologia , Telencéfalo/efeitos dos fármacos , Receptor 5 Toll-Like/genética , Receptor 5 Toll-Like/metabolismo , Poluentes Químicos da Água/toxicidade
19.
J Cell Mol Med ; 21(12): 3626-3632, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28707430

RESUMO

Pituitary stalk interruption syndrome (PSIS) is a rare type of hypopituitarism manifesting various degrees of pituitary hormone deficiency. Although mutations have been identified in some familial cases, the underpinning mechanisms of sporadic patients with PSIS who are in a vast majority remain elusive, necessitating a comprehensive study using systemic approaches. We postulate that other genetic mechanisms may be responsible for the sporadic PSIS. To test this hypothesis, we conducted a study in 24 patients with PSIS of Han Chinese with no family history using whole-exome sequencing (WES) and bioinformatic analysis. We identified a group of heterozygous mutations in 92% (22 of 24) of the patients, and these genes are mostly associated with Notch, Shh, Wnt signalling pathways. Importantly, 83% (20 of 24) of the patients had more than one mutation in those pathways suggesting synergy of compound mutations underpin the pathogenesis of sporadic PSIS.


Assuntos
Genoma Humano , Proteínas Hedgehog/genética , Hipopituitarismo/genética , Mutação , Hormônios Hipofisários/genética , Receptores Notch/genética , Proteínas Wnt/genética , Adolescente , Adulto , Povo Asiático , Criança , Biologia Computacional , Feminino , Expressão Gênica , Proteínas Hedgehog/metabolismo , Humanos , Hipopituitarismo/etnologia , Hipopituitarismo/metabolismo , Hipopituitarismo/patologia , Masculino , Hipófise/anormalidades , Hipófise/metabolismo , Hormônios Hipofisários/deficiência , Receptores Notch/metabolismo , Transdução de Sinais , Síndrome , Sequenciamento Completo do Genoma , Proteínas Wnt/metabolismo
20.
Endocrinology ; 158(6): 1776-1797, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28323939

RESUMO

Tachykinin-1 (TAC1) is known to have diverse functions in mammals, but similar information is scarce in fish species. Using grass carp as a model, the pituitary actions, receptor specificity and postreceptor signaling of TAC1 gene products, namely substance P (SP) and neurokinin A (NKA), were examined. TAC1 encoding SP and NKA as well as tachykinin receptors NK1R and NK2R were cloned in the carp pituitary. The newly cloned receptors were shown to be functional with properties similar to mammalian counterparts. In carp pituitary cells, SP and NKA could trigger luteinizing hormone (LH), prolactin (PRL), and somatolactin α (SLα) secretion, with parallel rises in PRL and SLα transcripts. Short-term SP treatment (3 hours) induced LH release, whereas prolonged induction (24 hours) could attenuate LHß messenger RNA (mRNA) expression. At pituitary cell level, LH, PRL, and SLα regulation by TAC1 gene products were mediated by NK1R, NK2R, and NK3R, respectively. Apparently, SP- and NKA-induced LH and SLα secretion and transcript expression were mediated by adenylyl cyclase/cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA), phospholiphase C (PLC)/inositol 1,4,5-triphosphate/protein kinase C (PKC), and Ca2+/calmodulin (CaM)/CaM-dependent protein kinase-II pathways. The signal transduction for PRL responses was similar, except for the absence of a PKC component. Regarding SP inhibition of LHß mRNA expression, the cAMP/PKA- and PLC/PKC-dependent (but not Ca2+/CaM-dependent) cascades were involved. These results, as a whole, suggest that TAC1 gene products play a role in LH, PRL, and SLα regulation via overlapping postreceptor signaling coupled to different subtypes of tachykinin receptor expressed in the carp pituitary.


Assuntos
Carpas , Hormônios Hipofisários/genética , Hormônios Hipofisários/metabolismo , Taquicininas/fisiologia , Animais , Carpas/genética , Carpas/metabolismo , Clonagem Molecular , Feminino , Proteínas de Peixes/genética , Proteínas de Peixes/fisiologia , Expressão Gênica , Células HEK293 , Humanos , Masculino , Hipófise/citologia , Hipófise/metabolismo , Maturidade Sexual/fisiologia , Transdução de Sinais/genética , Taquicininas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA