Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 12(3): e0174452, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28346489

RESUMO

Gonadotropin-releasing hormone (GnRH) is important in the control of reproduction, but its actions in non-reproductive processes are less well known. In this study we examined the effect of disrupting the GnRH receptor in mice to determine if growth, metabolism or behaviors that are not associated with reproduction were affected. To minimize the effects of other hormones such as FSH, LH and sex steroids, the neonatal-prepubertal period of 2 to 28 days of age was selected. The study shows that regardless of sex or phenotype in the Gnrhr gene knockout line, there was no significant difference in the daily development of motor control, sensory detection or spatial orientation among the wildtype, heterozygous or null mice. This included a series of behavioral tests for touch, vision, hearing, spatial orientation, locomotory behavior and muscle strength. Neither the daily body weight nor the final weight on day 28 of the kidney, liver and thymus relative to body weight varied significantly in any group. However by day 28, metabolic changes in the GnRH null females compared with wildtype females showed a significant reduction in inguinal fat pad weight normalized to body weight; this was accompanied by an increase in glucose compared with wildtype females shown by Student-Newman-Keuls Multiple Comparison test and Student's unpaired t tests. Our studies show that the GnRH-GnRHR system is not essential for growth or motor/sensory/orientation behavior during the first month of life prior to puberty onset. The lack of the GnRH-GnRHR axis, however, did affect females resulting in reduced subcutaneous inguinal fat pad weight and increased glucose with possible insulin resistance; the loss of the normal rise of estradiol at postnatal days 15-28 may account for the altered metabolism in the prepubertal female pups.


Assuntos
Comportamento Animal/fisiologia , Metabolismo Energético/genética , Locomoção/genética , Atividade Motora/genética , Orientação Espacial/fisiologia , Receptores LHRH/genética , Tecido Adiposo/metabolismo , Animais , Feminino , Resistência à Insulina/genética , Masculino , Camundongos , Camundongos Knockout , Receptores LHRH/metabolismo , Reflexo/genética
2.
J Neurosci ; 34(46): 15297-305, 2014 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-25392497

RESUMO

Sex differences in brain function underlie robust differences between males and females in both normal and disease states. Although alternative mechanisms exist, sexual differentiation of the male mammalian brain is initiated predominantly by testosterone secreted by the testes during the perinatal period. Despite considerable advances in understanding how testosterone and its metabolite estradiol sexually differentiate the brain, little is known about the mechanism that generates the male-specific perinatal testosterone surge. In mice, we show that a male-specific activation of GnRH neurons occurs 0-2 h following birth and that this correlates with the male-specific surge of testosterone occurring up to 5 h after birth. The necessity of GnRH signaling for the sexually differentiating effects of the perinatal testosterone surge was demonstrated by the persistence of female-like brain characteristics in adult male, GnRH receptor knock-out mice. Kisspeptin neurons have recently been identified to be potent, direct activators of GnRH neurons. We demonstrate that a population of kisspeptin neurons appears in the preoptic area of only the male between E19 and P1. The importance of kisspeptin inputs to GnRH neurons for the process of sexual differentiation was demonstrated by the lack of a normal neonatal testosterone surge, and disordered brain sexual differentiation of male mice in which the kisspeptin receptor was deleted selectively from GnRH neurons. These observations demonstrate the necessity of perinatal GnRH signaling for driving brain sexual differentiation and indicate that kisspeptin inputs to GnRH neurons are essential for this process to occur.


Assuntos
Hormônio Liberador de Gonadotropina/fisiologia , Neurônios/fisiologia , Área Pré-Óptica/metabolismo , Receptores Acoplados a Proteínas G/fisiologia , Diferenciação Sexual/fisiologia , Transdução de Sinais , Animais , Animais Recém-Nascidos , Feminino , Hormônio Liberador de Gonadotropina/genética , Masculino , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Gravidez , Receptores Acoplados a Proteínas G/genética , Receptores de Kisspeptina-1 , Receptores LHRH/genética , Receptores LHRH/fisiologia , Caracteres Sexuais , Testosterona/sangue , Tirosina 3-Mono-Oxigenase/metabolismo , Vasopressinas/metabolismo
3.
Gen Comp Endocrinol ; 209: 118-34, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25107740

RESUMO

Gonadotropin-releasing hormone (GnRH) and its receptor are essential for reproduction in vertebrates. Although there are three major types of GnRH peptides and two major types of receptors in vertebrates, the pattern of distribution is unusual. Evidence is presented from genome mining that type I GnRHRs are not restricted to mammals, but can be found in the lobe-finned and cartilaginous fishes. This implies that this tail-less GnRH receptor emerged early in vertebrate evolution, followed by several independent losses in different lineages. Also, we have identified representatives from the three major GnRH peptide types (mammalian GnRH1, vertebrate GnRH2 and dogfish GnRH3) in a single cartilaginous fish, the little skate. Skate and coelacanth are the only examples of animals with both type I and II GnRH receptors and all three peptide types, suggesting this was the ancestral condition in vertebrates. Our analysis of receptor synteny in combination with phylogeny suggests that there were three GnRH receptor types present before the two rounds of whole genome duplication in early vertebrates. To further understand the origin of the GnRH peptide-receptor system, the relationship of vertebrate and invertebrate homologs was examined. Our evidence supports the hypothesis of a GnRH superfamily with a common ancestor for the vertebrate GnRHs, invertebrate (inv)GnRHs, corazonins and adipokinetic hormones. The invertebrate deuterostomes (echinoderms, hemichordates and amphioxus) have derived GnRH-like peptides, although one amphioxus GnRH with a syntenic relationship to human GnRHs has been shown to be functional. Phylogenetic analysis suggests that gene duplications in the ancestral bilaterian produced two receptor types, one of which became adipokinetic hormone receptor/GnRHR and the other corazonin receptor/invGnRHR. It appears that the ancestral deuterostome had both a GnRHR and invGnRHR, and this is still the case in amphioxus. During the transition to vertebrates both the invertebrate-type peptide and receptor were lost, leaving only the vertebrate-type system that presently exists.


Assuntos
Evolução Molecular , Hormônio Liberador de Gonadotropina/genética , Receptores LHRH/genética , Sequência de Aminoácidos , Animais , Deleção de Genes , Hormônio Liberador de Gonadotropina/classificação , Hormônio Liberador de Gonadotropina/metabolismo , Humanos , Invertebrados/genética , Filogenia , Receptores LHRH/metabolismo , Reprodução/fisiologia , Vertebrados/genética
4.
Genome Biol Evol ; 6(6): 1466-79, 2014 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-24904013

RESUMO

The cystine knot growth factor (CKGF) superfamily includes important secreted developmental regulators, including the families of transforming growth factor beta, nerve growth factor, platelet-derived growth factor, and the glycoprotein hormones (GPHs). The evolutionary origin of the GPHs and the related invertebrate bursicon hormone, and their characteristic receptors, contributes to an understanding of the endocrine system in metazoans. Using a sensitive search method with hidden Markov models, we identified homologs of the hormones and receptors, along with the closely related bone morphogenetic protein (BMP) antagonists in basal metazoans. In sponges and a comb jelly, cystine knot hormones (CKHs) with mixed features of GPHs, bursicon, and BMP antagonists were identified using primary sequence and phylogenetic analysis. Also, we identified potential receptors for these CKHs, leucine-rich repeat-containing G protein-coupled receptors (LGRs), in the same species. Cnidarians, such as the sea anemone, coral, and hydra, diverged later in metazoan evolution and appear to have duplicated and differentiated CKH-like peptides resulting in bursicon/GPH-like peptides and several BMP antagonists: Gremlin (Grem), sclerostin domain containing (SOSD), neuroblastoma suppressor of tumorigenicity 1 (NBL1), and Norrie disease protein. An expanded cnidarian LGR group also evolved, including receptors for GPH and bursicon. With the appearance of bilaterians, a separate GPH (thyrostimulin) along with bursicon and BMP antagonists were present. Synteny indicates that the GPHs, Grem, and SOSD have been maintained in a common gene neighborhood throughout much of metazoan evolution. The stable and highly conserved CKGFs are not identified in nonmetazoan organisms but are established with their receptors in the basal metazoans, becoming critical to growth, development, and regulation in all animals.


Assuntos
Hormônio Foliculoestimulante/genética , Glicoproteínas/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Hormônios de Invertebrado/genética , Filogenia , Receptores Acoplados a Proteínas G/genética , Sequência de Aminoácidos , Animais , Evolução Molecular , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/química , Dados de Sequência Molecular , Receptores Acoplados a Proteínas G/química , Alinhamento de Sequência
5.
Mol Biol Evol ; 31(4): 765-78, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24361996

RESUMO

Gonadotropin-releasing hormone (GnRH) is a critical reproductive regulator in vertebrates. Homologous peptides are also found in invertebrates, with a variety of characterized functions. In the amphioxus, an invertebrate that provides the best model for the transition to vertebrates, four GnRH receptors (GnRHRs) were previously described, but their native ligands were not identified. Using a more sensitive search methodology with hidden Markov models, we identified the first GnRH-like peptide confirmed in the amphioxus Branchiostoma floridae. This peptide specifically activated one of the four GnRHRs. Although the primary structure of this peptide was divergent from any previously isolated GnRH peptide, the minimal conserved residues found in all other GnRH superfamily members were retained. The peptide was immunolocalized in proximity of the central canal of the anterior nerve cord, a region where other neuropeptides and receptors have been found. Additionally, the amphioxus GnRH-like gene was positioned in a locus surrounded by syntenic homologs of the human GnRH paralogon. The amphioxus GnRH-like peptide, with its distinct primary structure, activated a receptor with equal potency to multiple ligands that span the GnRH superfamily.


Assuntos
Hormônio Liberador de Gonadotropina/fisiologia , Anfioxos/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Células COS , Chlorocebus aethiops , Sequência Conservada , Evolução Molecular , Hormônio Liberador de Gonadotropina/química , Humanos , Dados de Sequência Molecular , Sistema Nervoso/metabolismo , Especificidade de Órgãos , Filogenia , Receptores LHRH/metabolismo , Transdução de Sinais , Sintenia
6.
Genome Biol Evol ; 3: 284-94, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21402861

RESUMO

Vertebrates have a large glycoprotein hormone, stanniocalcin, which originally was shown to inhibit calcium uptake from the environment in teleost fish gills. Later, humans, other mammals, and teleost fish were shown to have two forms of stanniocalcin (STC1 and STC2) that were widely distributed in many tissues. STC1 is associated with calcium and phosphate homeostasis and STC2 with phosphate, but their receptors and signaling pathways have not been elucidated. We undertook a phylogenetic investigation of stanniocalcin beyond the vertebrates using a combination of BLAST and HMMER homology searches in protein, genomic, and expressed sequence tag databases. We identified novel STC homologs in a diverse array of multicellular and unicellular organisms. Within the eukaryotes, almost all major taxonomic groups except plants and algae have STC homologs, although some groups like echinoderms and arthropods lack STC genes. The critical structural feature for recognition of stanniocalcins was the conserved pattern of ten cysteines, even though the amino acid sequence identity was low. Signal peptides in STC sequences suggest they are secreted from the cell of synthesis. The role of glycosylation signals and additional cysteines is not yet clear, although the 11th cysteine, if present, has been shown to form homodimers in some vertebrates. We predict that large secreted stanniocalcin homologs appeared in evolution as early as single-celled eukaryotes. Stanniocalcin's tertiary structure with five disulfide bonds and its primary structure with modest amino acid conservation currently lack an established receptor-signaling system, although we suggest possible alternatives.


Assuntos
Células Eucarióticas/metabolismo , Evolução Molecular , Glicoproteínas/genética , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Sequência Conservada/genética , Cisteína/genética , Glicoproteínas/química , Humanos , Dados de Sequência Molecular , Peptídeos/química , Filogenia , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Especificidade da Espécie
7.
Gen Comp Endocrinol ; 171(1): 1-16, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21185290

RESUMO

Gonadotropin-releasing hormone (GnRH) plays a central role in vertebrate reproduction. The evolutionary origin of this neuropeptide and its receptor is not obvious, but the advent of genomics makes it possible to examine the roots of GnRH and delve deeper into its ancestral relationships. New peptide sequences identified in invertebrates from annelids to tunicates reveal GnRH-like peptides of 10-12 amino acids. Structural conservation suggests homology between the 15 known invertebrate peptides and the 15 known vertebrate GnRHs. The functions of the invertebrate GnRH-like peptides are not necessarily related to reproduction. We suggest that structurally related families of invertebrate peptides including corazonin and adipokinetic hormone (AKH) form a superfamily of neuropeptides with the GnRH family. GnRH receptors have also been identified in invertebrates from annelids to tunicates suggesting that the origin of GnRH and its receptor extends deep in evolution to the origin of bilaterian animals. To resolve the relationship of invertebrate and vertebrate receptors, we conducted large-scale phylogenetic analysis using maximum likelihood. The data support a superfamily that includes GnRH, AKH and corazonin receptors derived from both published sequences and unpublished gene model predictions. Closely related to the GnRHR superfamily is the vasopressin/oxytocin superfamily of receptors. Phylogenetic analysis suggests a shared ancestry with deep roots. A functional role for GnRH in vertebrates or invertebrates leads to questions about the evolutionary origin of the pituitary. Our analysis suggests a functioning pituitary was the result of genomic duplications in early vertebrates.


Assuntos
Evolução Molecular , Hormônio Liberador de Gonadotropina/metabolismo , Sequência de Aminoácidos , Animais , Hormônio Liberador de Gonadotropina/química , Hormônio Liberador de Gonadotropina/classificação , Hormônio Liberador de Gonadotropina/genética , Humanos , Dados de Sequência Molecular , Filogenia , Receptores LHRH/metabolismo , Homologia de Sequência de Aminoácidos
8.
Endocrinology ; 151(3): 1142-52, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20068010

RESUMO

Mutations in the GnRH receptor gene (GNRHR) can result in hypogonadotropic hypogonadism in humans. Unlike most mammals, mice lack a second form of GnRH (GnRH2) and a type 2 GnRH receptor. To determine whether the GnRH receptor is critical at all stages of reproduction and whether this receptor has additional physiological functions in developing and adult mice, we have generated mice from an embryonic stem cell line containing a retroviral vector with multiple stop codons inserted into intron 1 of the Gnrhr gene. This gene trap insertion resulted in the disruption of exon 2 and exon 3 of the Gnrhr gene. The insertion also contained a lacZ gene that was used as a reporter for GnRH receptor expression in these mice. This model has a similar phenotype to the clinical syndrome of hypogonadotropic hypogonadism. Null Gnrhr mice had small sexual organs, low levels of FSH, LH, and steroid hormones, failure of sexual maturation, infertility, and inability to respond to exogenous GnRH. However, the defective GnRH receptor did not prevent morula/blastocyst development, implantation, masculinization of fetal male mice, or maintenance of early pregnancy. The phenotype of this null Gnrhr mouse was more severe than models in the literature, including the N-ethyl-N-nitrosourea-induced Gnrhr mutant, the kisspeptin (Kiss1) knockout, and the kisspeptin receptor (Gpr54) knockout. In terms of gonadal morphology, adult gene trap-Gnrhr null mice demonstrate a complete cessation of reproduction and serve as an important model for understanding GnRH/GnRHR physiology.


Assuntos
Hormônio Liberador de Gonadotropina/fisiologia , Hipogonadismo/genética , Ovário/embriologia , Receptores LHRH/genética , Testículo/embriologia , Alquilantes , Animais , Etilnitrosoureia , Feminino , Genes Reporter , Kisspeptinas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo , Gravidez , Proteínas/genética , Receptores Acoplados a Proteínas G/genética , Receptores de Kisspeptina-1
9.
Integr Comp Biol ; 50(1): 86-97, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21558190

RESUMO

Stanniocalcin (STC) is present throughout vertebrates, including humans, but a structure for STC has not been identified in animals that evolved before bony fish. The origin of this pleiotropic hormone known to regulate calcium is not clear. In the present study, we have cloned three stanniocalcins from two invertebrates, the tunicate Ciona intestinalis and the amphioxus Branchiostoma floridae. Both species are protochordates with the tunicates as the closest living relatives to vertebrates. Amphioxus are basal to both tunicates and vertebrates. The genes and predicted proteins of tunicate and amphioxus share several key structural features found in all previously described homologs. Both the invertebrate and vertebrate genes have four conserved exons. The predicted length of the single pro-STC in Ciona is 237 amino acids and the two pro-hormones in amphioxus are 207 and 210 residues, which is shorter than human pro-STCs at 247 and 302 residues due to expansion of the C-terminal region in vertebrate forms. The conserved pattern of 10 cysteines in all chordate STCs is crucial for identification as amphioxus and tunicate amino acids are only 14-23% identical with human STC1 and STC2. The 11th cysteine, which is the cysteine shown to form a homodimer in vertebrates, is present only in amphioxus STCa, but not in amphioxus STCb or tunicate STC, suggesting the latter two are monomers. The expression of stanniocalcin in Ciona is widespread as shown by RT-PCR and by quantitative PCR. The latter method shows that the highest amount of STC mRNA is in the heart with lower amounts in the neural complex, branchial basket, and endostyle. A widespread distribution is present also in mammals and fish for both STC1 and STC2. Stanniocalcin is a presumptive regulator of calcium in both Ciona and amphioxus, although the structure of a STC receptor remains to be identified in any organism. Our data suggest that amphioxus STCa is most similar to the common ancestor of vertebrate STCs because it has an 11th cysteine necessary for dimerization, an N-glycosylation motif, although not the canonical one in vertebrate STCs, and similar gene organization. Tunicate and amphioxus STCs are more similar in structure to vertebrate STC1 than to vertebrate STC2. The unique features of STC2, including 14 instead of 11 cysteines and a cluster of histidines in the C-terminal region, appear to be found exclusively in vertebrates.


Assuntos
Ciona intestinalis/genética , Genômica , Glicoproteínas/genética , Filogenia , Urocordados/genética , Sequência de Aminoácidos , Animais , Cálcio/metabolismo , Humanos , Dados de Sequência Molecular , Isoformas de Proteínas/genética , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Peixe-Zebra/genética
10.
Endocrinology ; 150(6): 2847-56, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19264870

RESUMO

In vertebrates, activation of the GnRH receptor is necessary to initiate the reproductive cascade. However, little is known about the characteristics of GnRH receptors before the vertebrates evolved. Recently genome sequencing was completed for amphioxus, Branchiostoma floridae. To understand the GnRH receptors (GnRHR) from this most basal chordate, which is also classified as an invertebrate, we cloned and characterized four GnRHR cDNAs encoded in the amphioxus genome. We found that incubation of GnRH1 (mammalian GnRH) and GnRH2 (chicken GnRH II) with COS7 cells heterologously expressing the amphioxus GnRHRs caused potent intracellular inositol phosphate turnover in two of the receptors. One of the two receptors displayed a clear preference for GnRH1 over GnRH2, a characteristic not previously seen outside the type I mammalian GnRHRs. Phylogenetic analysis grouped the four receptors into two paralogous pairs, with one pair grouping basally with the vertebrate GnRH receptors and the other grouping with the octopus GnRHR-like sequence and the related receptor for insect adipokinetic hormone. Pharmacological studies showed that octopus GnRH-like peptide and adipokinetic hormone induced potent inositol phosphate turnover in one of these other two amphioxus receptors. These data demonstrate the functional conservation of two distinct types of GnRH receptors at the base of chordates. We propose that one receptor type led to vertebrate GnRHRs, whereas the other type, related to the mollusk GnRHR-like receptor, was lost in the vertebrate lineage. This is the first report to suggest that distinct invertebrate and vertebrate GnRHRs are present simultaneously in a basal chordate, amphioxus.


Assuntos
Evolução Molecular , Invertebrados , Filogenia , Receptores LHRH/análise , Receptores LHRH/genética , Vertebrados , Sequência de Aminoácidos , Animais , Cordados , DNA Complementar/genética , Humanos , Dados de Sequência Molecular , Receptores LHRH/fisiologia , Reprodução/fisiologia , Transdução de Sinais/fisiologia
11.
J Comp Neurol ; 513(6): 685-701, 2009 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-19235874

RESUMO

The distribution of growth hormone-releasing hormone-like peptides (GHRH-LP) in the central nervous system of the zebrafish was investigated by using immunohistochemical techniques with polyclonal antibodies. ELISAs showed that the antiserum raised against salmon (s)GHRH-LP recognized both zebrafish GHRH-LP1 and -2, whereas the antiserum raised against carp (c)GHRH-LP was more sensitive but detected only zebrafish GHRH-LP1. Neither antiserum detected the true GHRH. Large cells in the nucleus lateralis tuberis were immunoreactive with both antisera, which suggests that they contained zebrafish GHRH-LP1, but not excluding GHRH-LP2. Also, immunoreactive fibers, which putatively originated from these hypothalamic neurons, were present in the hypophysis; both antisera detected fibers, although only sGHRH-LP antiserum stained fibers in the neurointermediate lobe. These fibers may have a neuroendocrine role. Candidates for a role in feeding include several areas in which both antisera labeled cells and fibers, implying a strong reaction for GHRH-LP1 and possibly GHRH-LP2. These areas include the isthmus with cells in the secondary gustatory/visceral nucleus, which were also calretinin immunoreactive. Numerous GHRH-LP-immunoreactive fibers (also labeled by both antisera) probably originate from the gustatory/visceral nucleus to innervate the ventral area of the telencephalon, preglomerular nuclei, torus lateralis and hypothalamic diffuse nucleus, habenula, torus semicircularis, and dorsolateral funiculus of the spinal cord. Present results in the zebrafish brain suggest involvement of GHRH-LP in both neuroendocrine and feeding-associated nervous circuits. The present data on the location of the two GHRH-LPs are the first clue to the possible functions of these two hormones.


Assuntos
Encéfalo/metabolismo , Hormônio Liberador de Hormônio do Crescimento/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/anatomia & histologia , Sequência de Aminoácidos , Animais , Carpas , Sequência Conservada , Ensaio de Imunoadsorção Enzimática , Hormônio Liberador de Hormônio do Crescimento/genética , Humanos , Imuno-Histoquímica , Dados de Sequência Molecular , Neurônios/metabolismo , Proteínas de Peixe-Zebra/genética
12.
Gen Comp Endocrinol ; 161(1): 3-12, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19007784

RESUMO

Modern fish are the result of major changes in evolution including three possible duplications of the whole genome. Retained duplicate genes are often involved with metabolism, transcription, neurogenic processes and development. Here we examine the consequences of the most recent (350 mya) teleost-specific duplication in five fishes (zebrafish, fugu, medaka, stickleback and rainbow trout) in regard to duplicate copies of hormones and receptors in the secretin superfamily. This subset of genes was selected as the superfamily is limited to ten hormones and their receptors and includes some important members: glucagon, growth hormone-releasing hormone (GHRH), pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal polypeptide (VIP). We used reports from the literature and an extensive database search of the fish genomes to evaluate the status of the superfamily and its duplicate genes. We found that all five fish species have an almost complete set of orthologs with the human superfamily of hormones, although they lack secretin and its receptor. Receptor orthologs are present in zebrafish, fugu, medaka, stickleback and to a lesser extent in salmonids. Zebrafish retain duplicate copies for seven hormones and five receptors. Duplicated genes in fugu, medaka, stickleback and salmonids are also present, based mainly on genome annotation or mRNA transcription. Separate chromosome locations and synteny support zebrafish duplicates as the result of large-scale duplications. Novel changes in fish include the modification of a duplicate glucagon receptor to a GLP-1 receptor and, unlike humans, the presence of bioactive and specific PHI and GHRH-like peptide receptors. We conclude that fish duplicates in the secretin superfamily are a rich, mostly unexplored area for endocrine research.


Assuntos
Peixes/genética , Genes Duplicados , Receptores Acoplados a Proteínas G/genética , Receptores dos Hormônios Gastrointestinais/genética , Secretina/genética , Animais , Oncorhynchus mykiss/genética , Oryzias/genética , Smegmamorpha/genética , Takifugu/genética , Peixe-Zebra/genética
13.
J Mol Endocrinol ; 41(5): 343-66, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18757498

RESUMO

A group of ten hormones in humans are structurally related and known as the secretin superfamily. These hormones bind to G-protein-coupled receptors that activate the cAMP pathway and are clustered as the secretin or B family. We used an evolutionary approach with zebrafish as a model to understand why some of these hormones, such as peptide histidine-methionine (PHM) and pituitary adenylate cyclase-activating polypeptide (PACAP)-related peptide (PRP) in humans lack a receptor. We used molecular techniques to clone two full-length receptor cDNAs in zebrafish, which were analyzed for amino acid sequence and ligand-binding motifs, phylogenetic position, synteny, tissue expression, functional response, and signaling pathway. Evidence is provided that the two cDNAs encoded the peptide histidine-isoleucine (PHI) receptor and PRP receptor, which is known as GHRH-like peptide (GHRH-LP) receptor in non-mammals. Further, we cloned a zebrafish cDNA encoding the peptides PHI and vasoactive intestinal peptide (VIP). The PHIR had been previously labeled as one type of a VIP-PACAP (VPAC2R) shared receptor based only on sequence data. The PHIR cDNA, transfected into COS7 cells, responded to zebrafish PHI in a sensitive and dose-dependent manner (EC(50)=1.8x10(-9) M) but not to PACAP and VIP. The GHRH-LP receptor responded to both zebrafish GHRH-LP1 and GHRH with a 3.5-fold greater response to the former. For comparison, two zebrafish receptors (PAC1R and VPAC1R) and two human receptors (VPAC2R and GHRHR) were tested with human and/or zebrafish peptides. Unexpectedly, zebrafish VIP activated its PAC1R suggesting that in evolution, PAC1R is not always a specific receptor for PACAP. We conclude that zebrafish, like goldfish, have a specific receptor for PHI and GHRH-LP. Our evidence that zebrafish PHI is more potent than human PHM in activating the human VPAC2R (EC(50)=7.4x10(-9) M) supports our suggestion that the VPAC2R and PHIR shared a common ancestral receptor.


Assuntos
Hormônio Liberador de Hormônio do Crescimento/metabolismo , Peptídeo PHI/metabolismo , Receptores de Superfície Celular/metabolismo , Secretina/metabolismo , Peixe-Zebra/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Hormônio Liberador de Hormônio do Crescimento/genética , Humanos , Dados de Sequência Molecular , Peptídeo PHI/genética , Peptídeos/genética , Peptídeos/metabolismo , Filogenia , Receptores de Superfície Celular/classificação , Receptores de Superfície Celular/genética , Secretina/classificação , Secretina/genética , Alinhamento de Sequência , Distribuição Tecidual , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/classificação , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
14.
Endocrinology ; 149(4): 1571-80, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18162530

RESUMO

Disruption of the pituitary adenylate cyclase-activating polypeptide (PACAP) gene in mice has demonstrated a role for this highly conserved neuropeptide in the regulation of metabolism and temperature control. Localization of PACAP neurons within hypothalamic nuclei that regulate appetite suggest PACAP may affect feeding and thus energy balance. We used PACAP-null mice to address this question, examining both food intake and energy expenditure. PACAP-null mice were leaner than wild-type littermates due to decreased adiposity and displayed increased insulin sensitivity. The lean phenotype in the PACAP-null mice was completely eliminated if animals were fed a high-fat diet or housed near thermoneutrality (28 C). Further metabolic analyses of PACAP-null mice housed at 21 C indicated that the reduced body weight could not be explained by decreased food intake, increased metabolic rate, or increased locomotor activity. The thyroid hormone axis of PACAP-null mice was affected, because mRNA levels of hypothalamic TRH and brown adipose tissue type 2 deiodinase were reduced in PACAP-null mice housed at room temperature, and brain deiodinase activity was lower in PACAP-null mice after an acute cold challenge compared with wild-type controls. These results demonstrate that PACAP is not required for the regulation of food intake yet is necessary to maintain normal energy homeostasis, likely playing a role in central cold-sensing mechanisms.


Assuntos
Ingestão de Alimentos , Metabolismo Energético , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/fisiologia , Adipogenia , Animais , Regulação da Temperatura Corporal , Temperatura Baixa , Insulina/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/metabolismo , Hormônios Tireóideos/sangue , Hormônio Liberador de Tireotropina/metabolismo
15.
Mol Cell Endocrinol ; 280(1-2): 13-9, 2008 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-17945412

RESUMO

Mice lacking pituitary adenylate cyclase-activating polypeptide (PACAP) show high mortality during the postnatal period, as well as impaired reproduction in females. This study characterizes the reproductive phenotype in female mice lacking PACAP due to targeted disruption (knockout) of the single copy pacap gene (Adcyap1) to determine the site(s) of action of PACAP in the cascade of reproductive events. PACAP null females showed normal puberty onset, estrous cycles, and seminal plugs when paired with a male of proven fertility. However, significantly fewer PACAP null females (21%) than wild-type females (100%) gave birth following mating. Although a defect was not detected in ovulation, ovarian histology or fertilization of released eggs in PACAP null females, only 13% had implanted embryos 6.5 days after mating. Associated with the decrease in implantation, prolactin and progesterone levels were significantly lower in females lacking PACAP than in wild types on day 6.5 after mating. Our evidence suggests that impaired implantation is the defect responsible for decreased fertility in PACAP null female mice.


Assuntos
Implantação do Embrião , Fertilidade , Infertilidade Feminina/fisiopatologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Animais , Peso ao Nascer , Implantação do Embrião/genética , Estro/metabolismo , Feminino , Fertilidade/genética , Genótipo , Infertilidade Feminina/genética , Infertilidade Feminina/metabolismo , Tamanho da Ninhada de Vivíparos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ovulação , Fenótipo , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/deficiência , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Gravidez , Progesterona/sangue , Prolactina/sangue , Pseudogravidez/metabolismo , Pseudogravidez/fisiopatologia , Maturidade Sexual , Fatores de Tempo
16.
Integr Comp Biol ; 48(5): 570-87, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21669817

RESUMO

Reproduction in all vertebrates requires the brain hormone gonadotropin-releasing hormone (GnRH) to activate a cascade of events leading to gametogenesis. All vertebrates studied to date have one to three forms of GnRH in specific but different neurons in the brain. In addition, at least one type of GnRH receptor is present in each vertebrate for activation of specific physiological events within a target cell. Humans possess two types of GnRH (GnRH1 and GnRH2) but only one functional GnRH receptor. Zebrafish, Danio rerio, also have two types of GnRH (GnRH2 and GnRH3), although in contrast to humans, zebrafish appear to have four different GnRH receptors in their genome. To characterize the biological significance of multiple GnRH receptors within a single species, we cloned four GnRH receptor cDNAs from zebrafish and compared their structures, expression, and cell physiology. The zebrafish receptors are 7-transmembrane G-protein coupled receptors with amino-acid sequence identities ranging from 45 to 71% among the four receptors. High sequence similarity was observed among the seven helices of zebrafish GnRHRs compared with the human GnRHR, the green monkey type II GnRHR, and the two goldfish GnRHRs. Also, key amino acids for putative ligand binding, disulfide bond formation, N-glycosylation, and G-protein coupling were present in the extracellular and intracellular domains. The four zebrafish receptors were expressed in a variety of tissues including the brain, eye, and gonads. In an inositol phosphate assay, each receptor was functional as shown by its response to physiological doses of native GnRH peptides; two receptors showed selectivity between GnRH2 and GnRH3. Each of the four receptor genes was mapped to distinct chromosomes. Our phylogenetic and syntenic analysis segregated the four zebrafish GnRH receptors into two distinct phylogenetic groups that are separate gene lineages conserved throughout vertebrate evolution. We suggest the maintenance of four functional GnRH receptors in zebrafish compared with only one in humans may depend either on subfunctionalization or neofunctionalization in fish compared with mammalian GnRH receptors. The differences in structure, location, and response to GnRH forms strongly suggests that the four zebrafish GnRH receptors have novel functions in addition to the conventional activation of the pituitary gland in the reproductive axis.

17.
Eur J Neurosci ; 25(9): 2604-18, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17561835

RESUMO

Previous studies have demonstrated that pituitary adenylate cyclase-activating polypeptide (PACAP) exerts trophic effects during neurodevelopment. In particular, the occurrence of PACAP and its receptors in the cerebellum during pre- and postnatal periods suggests that it could play a crucial role in ontogenesis of this structure. To test this hypothesis, we compared the histogenesis of cerebellar cortex in wild-type and PACAP-knockout (PACAP-/-) mice at postnatal days (P)4 and 7. Morphometric analysis of PACAP-/- mice revealed a significant reduction in the thickness of the external granule cell layer at P4 and of the internal granule cell layer at P7. Expression of nestin, a neural precursor marker, and synaptophysin, a mature neuronal marker, was quantified by real-time PCR and Western blot. No modification of nestin expression was noticed between wild-type and PACAP-/- mice, but a substantial decrease in synaptophysin expression was observed in PACAP-/- mice at P4 and P7. Immunohistochemistry revealed a reduction in synaptophysin labelling in the molecular and internal granule cell layers of PACAP-/- mice at P7. Caspase-3 activation was significantly increased in PACAP-/- mice at P4 and P7. Autoradiographic studies revealed no difference in PACAP binding site distributions and PACAP was effective at stimulating cAMP production in both wild-type and PACAP-/- cultured granule cells. This study demonstrates that disruption of the PACAP gene induces alteration of the immature cerebellum. Neuronal differentiation of granule cells was delayed whereas cell death that naturally occurs during ontogeny was increased in PACAP-/- mice. These data provide the first evidence of a physiological role for PACAP during cerebellar development.


Assuntos
Cerebelo/anormalidades , Cerebelo/crescimento & desenvolvimento , Regulação da Expressão Gênica no Desenvolvimento/genética , Neurônios/metabolismo , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Células-Tronco/metabolismo , Animais , Animais Recém-Nascidos , Biomarcadores/metabolismo , Caspase 3/metabolismo , Morte Celular/genética , Diferenciação Celular/genética , Cerebelo/metabolismo , AMP Cíclico/metabolismo , Ativação Enzimática/fisiologia , Proteínas de Filamentos Intermediários/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/metabolismo , Malformações do Sistema Nervoso/genética , Malformações do Sistema Nervoso/metabolismo , Malformações do Sistema Nervoso/fisiopatologia , Nestina , Neurônios/citologia , Células-Tronco/citologia , Sinaptofisina/metabolismo
18.
Peptides ; 28(9): 1680-7, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17467121

RESUMO

One approach to understanding the role of PACAP in vivo is to knockdown the translation of PACAP mRNA to protein or to knock out the PACAP gene by targeted disruption. In this paper, we review the effect of PACAP knockdown with morpholinos on early brain development in zebrafish. Also reviewed is the role of PACAP at several stages of reproduction as assessed in mice with a disrupted PACAP gene. New data are presented to analyze PACAP's action in energy homeostasis (body mass, food intake, endocrine parameters) using female PACAP-null mice. The evidence suggests PACAP is important for brain development in zebrafish and is required for normal reproduction, but not for body mass or food intake in mice maintained near thermoneutrality.


Assuntos
Encéfalo/embriologia , Deleção de Genes , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/fisiologia , Animais , Encéfalo/metabolismo , Ingestão de Alimentos/fisiologia , Comportamento Alimentar/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Reprodução/fisiologia , Peixe-Zebra
19.
Ann N Y Acad Sci ; 1070: 602-21, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16888233

RESUMO

To study the role of pituitary adenylate cyclase-activating polypeptide (PACAP) in early brain development, we examined PACAP and its receptors for first expression and then separately knocked down the two forms of PACAP in zebrafish where development is rapid and observable. We injected morpholinos (antisense oligonucleotides) into fertilized eggs to block PACAP. Morphological changes in the brain were observed in embryos at 27 h post fertilization (hpf). Using in situ hybridization of early brain marker genes, we found that the most striking effects were an increase in pax2.1 expression in eye stalks associated with absence of either form of PACAP or an increase in eng2 and fgf8 in the midbrain-hindbrain boundary after loss of PACAP2. These marker genes are among the earliest factors in the formation of the midbrain-hindbrain boundary, an early organizing center. We suggest that PACAP is a target gene with feedback inhibition on pax2.1, eng2, or fgf8 in specific brain areas. In the hindbrain, the absence of either form of PACAP had little effect, as shown by expression of ephA4 and meis1.1. During midbrain development, our evidence suggests that PACAP1 can activate mbx. In both the diencephalon and/or forebrain, lack of PACAP1 or PACAP2 led to an increase in fgf8, again suggesting a suppressive effect of PACAP during development on these important genes that help to define cells in the forebrain. The early expression of transcripts for PACAP and its receptors by 0.5-6 hpf make both PACAP1 and PACAP2 candidates for factors that influence brain development.


Assuntos
Encéfalo/embriologia , Encéfalo/metabolismo , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Animais , Biomarcadores , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Olho/embriologia , Olho/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Fatores de Tempo , Transcrição Gênica/genética , Peixe-Zebra/metabolismo
20.
Mol Cell Endocrinol ; 257-258: 47-64, 2006 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-16934393

RESUMO

Gonadotropin-releasing hormone (GnRH) is a highly conserved peptide that is expressed early in brain development in vertebrates. In zebrafish, we detected GnRH mRNA within 2h post fertilization by RT-PCR. To determine if GnRH is involved in development, we used gene knockdown techniques to block translation of gnrh2 or gnrh3 mRNA after which the expression patterns for gene markers were examined at 24h post fertilization with in situ hybridization. First, loss of either GnRH2 or GnRH3 affected regionalization of the brain as shown by a change in expression of fgf8 or pax2.1 genes in the midbrain-hindbrain boundary or diencephalon-midbrain boundary. Second, lack of GnRH2 and/or GnRH3 altered gene markers expressed in the formation of the eye cup (pax2.1, pax6.1, mab21l2 and meis1.1) or eye stalk (fgf8 and pax2.1). Third, knockdown of GnRH2 affected the size and shape of the midbrain and expression of gene markers therein. Results from assays with the TUNEL method and caspase-3 and -9 activity showed the brain and eye changes were unlikely to result from secondary apoptotic cell death before 24h post fertilization. These experiments suggest that GnRH loss-of-function affects early brain and eye formation during development.


Assuntos
Encéfalo/embriologia , Olho/embriologia , Hormônio Liberador de Gonadotropina/fisiologia , Sequência de Aminoácidos , Animais , Apoptose/genética , Sequência de Bases , Padronização Corporal , Encéfalo/metabolismo , Diencéfalo/embriologia , Diencéfalo/metabolismo , Embrião não Mamífero , Olho/metabolismo , Proteínas do Olho/metabolismo , Fator 8 de Crescimento de Fibroblasto/metabolismo , Hormônio Liberador de Gonadotropina/análogos & derivados , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/metabolismo , Proteínas de Homeodomínio/metabolismo , Imuno-Histoquímica , Mesencéfalo/embriologia , Mesencéfalo/metabolismo , Dados de Sequência Molecular , Proteína Meis1 , Oligonucleotídeos Antissenso/farmacologia , Oligopeptídeos/genética , Oligopeptídeos/metabolismo , Oligopeptídeos/fisiologia , Organogênese/fisiologia , Fator de Transcrição PAX2/metabolismo , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/metabolismo , Ácido Pirrolidonocarboxílico/análogos & derivados , Ácido Pirrolidonocarboxílico/metabolismo , Interferência de RNA , Receptor EphA4/metabolismo , Proteínas Repressoras/metabolismo , Rombencéfalo/embriologia , Rombencéfalo/metabolismo , Fatores de Transcrição/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...