Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(22): e2316459121, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38781215

RESUMO

Adult male animals typically court and attempt to mate with females, while attacking other males. Emerging evidence from mice indicates that neurons expressing the estrogen receptor ESR1 in behaviorally relevant brain regions play a central role in mediating these mutually exclusive behavioral responses to conspecifics. However, the findings in mice are unlikely to apply to vertebrates in general because, in many species other than rodents and some birds, androgens-rather than estrogens-have been implicated in male behaviors. Here, we report that male medaka (Oryzias latipes) lacking one of the two androgen receptor subtypes (Ara) are less aggressive toward other males and instead actively court them, while those lacking the other subtype (Arb) are less motivated to mate with females and conversely attack them. These findings indicate that, in male medaka, the Ara- and Arb-mediated androgen signaling pathways facilitate appropriate behavioral responses, while simultaneously suppressing inappropriate responses, to males and females, respectively. Notably, males lacking either receptor retain the ability to discriminate the sex of conspecifics, suggesting a defect in the subsequent decision-making process to mate or fight. We further show that Ara and Arb are expressed in intermingled but largely distinct populations of neurons, and stimulate the expression of different behaviorally relevant genes including galanin and vasotocin, respectively. Collectively, our results demonstrate that male teleosts make adaptive decisions to mate or fight as a result of the activation of one of two complementary androgen signaling pathways, depending on the sex of the conspecific that they encounter.


Assuntos
Androgênios , Oryzias , Receptores Androgênicos , Comportamento Sexual Animal , Transdução de Sinais , Animais , Masculino , Oryzias/metabolismo , Oryzias/fisiologia , Comportamento Sexual Animal/fisiologia , Feminino , Receptores Androgênicos/metabolismo , Receptores Androgênicos/genética , Androgênios/metabolismo , Agressão/fisiologia
2.
Zoolog Sci ; 40(2): 91-104, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37042689

RESUMO

For adaptation to a high salinity marine environment, cartilaginous fishes have evolved a ureosmotic strategy. They have a highly elaborate "four-loop nephron" in the kidney, which is considered to be important for reabsorption of urea from the glomerular filtrate to maintain a high concentration of urea in the body. However, the function and regulation, generally, of the "four-loop nephron" are still largely unknown due to the complicated configuration of the nephron and its many subdivided segments. Laser microdissection (LMD) followed by RNA-sequencing (RNA-seq) analysis is a powerful technique to obtain segment-dependent gene expression profiles. In the present study, using the kidney of cloudy catshark, Scyliorhinus torazame, we tested several formaldehyde-free and formaldehyde-based fixatives to optimize the fixation methods. Fixation by 1% neutral buffered formalin for 15 min resulted in sufficient RNA and structural integrities, which allowed LMD clipping of specific nephron segments and subsequent RNA-seq analysis. RNA-seq from the LMD samples of the second-loop, the fourth-loop, and the five tubular segments in the bundle zone revealed a number of specific membrane transporter genes that can characterize each segment. Among them, we examined expressions of the Na + -coupled cotransporters abundantly expressed in the second loop samples. Although the proximal II segment of the second loop is known for the elimination of excess solutes, the present results imply that the PII segment is also crucial for reabsorption of valuable solutes. Looking ahead to future studies, the segment-dependent gene expression profiling will be a powerful technique for unraveling the renal mechanisms and regulation in euryhaline elasmobranchs.


Assuntos
Microdissecção , Néfrons , Animais , Peixes , Perfilação da Expressão Gênica , RNA , Ureia/metabolismo
3.
Proc Biol Sci ; 287(1928): 20200713, 2020 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-32517612

RESUMO

Serotonin is a biogenic monoamine conserved across phyla that is implicated in diverse physiological and behavioural functions. On examining the expression of the rate-limiting enzymes in serotonin synthesis, tryptophan hydroxylases (TPHs), in the teleost medaka (Oryzias latipes), we found that males have much higher levels of tph1 expression as compared with females. This robust sexual dimorphism was found to probably result from the direct stimulation of tph1 transcription by androgen/androgen receptor binding to canonical bipartite androgen-responsive elements in its proximal promoter region. Our results further revealed that tph1 expression occurs exclusively in pro-opiomelanocortin (pomc)-expressing cells and that the resulting serotonin and its derivative melatonin inhibit the expression of the pituitary hormone genes, fshb, sl and tshb. This suggests that serotonin and/or melatonin synthesized in pomc-expressing cells act in a paracrine manner to suppress pituitary hormone levels. Consistent with these findings and the male-biased expression of tph1, the expression levels of fshb, sl and tshb were all higher in females than in males. Taken together, the male bias in tph1 expression and consequent serotonin/melatonin production presumably contribute to sex differences in the expression of pituitary hormones and ultimately in the physiological functions mediated by them.


Assuntos
Oryzias/fisiologia , Hormônios Hipofisários/metabolismo , Caracteres Sexuais , Animais , Feminino , Masculino , Melatonina/metabolismo , Oryzias/metabolismo , Serotonina/metabolismo , Triptofano Hidroxilase/metabolismo
4.
Gen Comp Endocrinol ; 284: 113129, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30825478

RESUMO

Vertebrate brains are sexually differentiated, giving rise to differences in various physiological and behavioral phenotypes between the sexes. In developing mammals and birds, the neural substrate underlying sex-dependent physiology and behavior undergoes an irreversible process of sexual differentiation due to the effects of perinatal gonadal steroids and sex chromosome complement. The differentiated neural substrate is then activated in the adult by the sex-specific steroid milieu to facilitate the expression of sex-typical phenotypes. However, this well-established concept does not hold for teleost fish, whose sexual phenotypes (behavioral or otherwise) are highly labile throughout life and can be reversed even in adulthood. Indeed, the available evidence suggests that, in teleosts, neither gonadal steroids early in development nor the sex chromosome complement contribute much to brain sexual differentiation; instead, steroids in adulthood serve to both differentiate the neural substrate and activate it to elicit sex-typical phenotypes in a transient and reversible manner. Evidence further suggests that marked sexual dimorphisms and adult steroid-dependent lability in the neural expression of sex steroid receptors constitute the primary molecular basis for sexual differentiation and lability of the teleost brain. The consequent sexually dimorphic but reversible steroid sensitivity in response to the adult steroid milieu may enable the teleost brain to maintain lifelong sexual lability and to undergo phenotypic sex reversal.


Assuntos
Encéfalo/fisiologia , Peixes/fisiologia , Diferenciação Sexual , Animais , Aves/fisiologia , Feminino , Masculino , Mamíferos/fisiologia , Cromossomos Sexuais/genética
6.
Fish Physiol Biochem ; 45(2): 753-771, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30617941

RESUMO

The localization of gonadotropin-releasing hormone (GnRH) in the brain and pituitary of the self-fertilizing mangrove killifish Kryptolebias marmoratus was examined by immunohistochemistry and in situ hybridization to understand its neuroendocrine system. The genome assembly of K. marmoratus did not have any sequence encoding GnRH1, but sequences encoding GnRH2 (chicken GnRH-II) and GnRH3 (salmon GnRH) were found. Therefore, GnRH1 was identified by in silico cloning. The deduced amino acid sequence of the K. marmoratus GnRH1 (mature peptide) was identical to that of the medaka GnRH. GnRH1 neurons were detected in the ventral part of the preoptic nucleus by immunohistochemistry and in situ hybridization, and GnRH1-immunoreactive (ir) fibers were observed throughout the brain. GnRH1-ir fibers were in close contact with luteinizing hormone (LH)-ir cells in the pituitary using double immunohistochemistry. GnRH2 neurons were detected in the midbrain tegmentum by immunohistochemistry and in situ hybridization. Although GnRH2-ir fibers were observed throughout the brain, they were not detected in the pituitary. GnRH3 neurons were detected in the lateral part of the ventral telencephalic area by both methods. GnRH3-ir fibers were observed throughout the brain, and a few GnRH3-ir fibers were in close contact with LH-ir cells in the pituitary. These results indicate that GnRH1 and possibly GnRH3 are responsible for gonadal maturation through LH secretion and that all three forms of GnRH function as neurotransmitters or neuromodulators in the brain of K. marmoratus.


Assuntos
Encéfalo/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Peixes Listrados/metabolismo , Hipófise/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Hormônio Liberador de Gonadotropina/química , Organismos Hermafroditas/fisiologia , Humanos , Imuno-Histoquímica , Filogenia , Reprodução/fisiologia
7.
Gen Comp Endocrinol ; 223: 47-53, 2015 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-26433060

RESUMO

The differential impact of stress on brain functions of males and females has been widely observed in vertebrates. Recent evidence suggests that stress-induced glucocorticoid signaling affects sexual differentiation and sex changes in teleost fish. These facts led us to postulate that there were sex differences in glucocorticoid signaling in the teleost brain that underlie some sex differences in their physiological and behavioral traits. Here we found sexually dimorphic expression of a glucocorticoid receptor gene (gr1) in the brain of medaka fish (Oryzias latipes), with females having greater expression in several preoptic and thalamic nuclei. Further, gr1 exhibits female-biased expression in neurons of the anterior parvocellular preoptic nucleus that produce the neuropeptides vasotocin and gonadotropin-releasing hormone 1 (these neuropeptides have been implicated in the regulation of neuroendocrine and behavioral functions). These findings suggest that glucocorticoids have a greater influence on physiology and behavior mediated by these neuropeptides in females than in males, which may contribute to sex differences in the brain's response to stress.


Assuntos
Encéfalo/metabolismo , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Oryzias/metabolismo , Receptores de Glucocorticoides/genética , Animais , Feminino , Hibridização In Situ , Masculino , Neurônios/citologia , Oryzias/genética , Oryzias/crescimento & desenvolvimento , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Glucocorticoides/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores Sexuais
8.
Biochem Biophys Res Commun ; 445(1): 113-9, 2014 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-24491545

RESUMO

In vertebrates, sex differences in the brain have been attributed to differences in gonadal hormone secretion; however, recent evidence in mammals and birds shows that sex chromosome-linked genes, independent of gonadal hormones, also mediate sex differences in the brain. In this study, we searched for genes that were differentially expressed between the sexes in the brain of a teleost fish, medaka (Oryzias latipes), and identified two sex chromosome genes with male-biased expression, cntfa (encoding ciliary neurotrophic factor a) and pdlim3a (encoding PDZ and LIM domain 3 a). These genes were found to be located 3-4 Mb from and on opposite sides of the Y chromosome-specific region containing the sex-determining gene (the medaka X and Y chromosomes are genetically identical, differing only in this region). The male-biased expression of both genes was evident prior to the onset of sexual maturity. Sex-reversed XY females, as well as wild-type XY males, had more pronounced expression of these genes than XX males and XX females, indicating that the Y allele confers higher expression than the X allele for both genes. In addition, their expression was affected to some extent by sex steroid hormones, thereby possibly serving as focal points of the crosstalk between the genetic and hormonal pathways underlying brain sex differences. Given that sex chromosomes of lower vertebrates, including teleost fish, have evolved independently in different genera or species, sex chromosome genes with sexually dimorphic expression in the brain may contribute to genus- or species-specific sex differences in a variety of traits.


Assuntos
Encéfalo/metabolismo , Fator Neurotrófico Ciliar/genética , Proteínas de Peixes/genética , Oryzias/genética , Cromossomo Y/genética , Sequência de Aminoácidos , Animais , Fator Neurotrófico Ciliar/classificação , Estradiol/farmacologia , Feminino , Expressão Gênica/efeitos dos fármacos , Perfilação da Expressão Gênica , Ligação Genética , Masculino , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , Filogenia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Fatores Sexuais , Testosterona/análogos & derivados , Testosterona/farmacologia , Fatores de Tempo
9.
Cell Tissue Res ; 356(1): 243-51, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24477796

RESUMO

The stress-related corticotropin-releasing hormone (CRH) was first identified by isolation of its cDNA from the brain of the Japanese eel Anguilla japonica. CRH cDNA encodes a signal peptide, a cryptic peptide and CRH (41 amino acids). The sequence homology to mammalian CRH is high. Next, the distribution of CRH-immunoreactive (ir) cell bodies and fibers in the brain and pituitary were examined by immunohistochemistry. CRH-ir cell bodies were detected in several brain regions, e.g., nucleus preopticus pars magnocellularis, nucleus preopticus pars gigantocellularis and formatio reticularis superius. In the brain, CRH-ir fibers were distributed not only in the hypothalamus but also in various regions. Some CRH-ir fibers projected to adrenocorticotropic hormone (ACTH) cells in the rostral pars distalis of the pituitary and also the α-melanocyte-stimulating hormone (α-MSH) cells in the pars intermedia of the pituitary. Finally, the neuroanatomical relationship between the CRH neurons and gonadotropin-releasing hormone (GnRH) neurons was examined by dual-label immunohistochemistry. CRH-ir fibers were found to be in close contact with GnRH-ir cell bodies in the hypothalamus and in the midbrain tegmentum and GnRH-ir fibers were in close contact with CRH-ir cell bodies in the nucleus preopticus pars magnocellularis. These results suggest that CRH has some physiological functions other than the stimulation of ACTH and α-MSH secretion and that reciprocal connections may exist between the CRH neurons and GnRH neurons in the brain of the Japanese eel.


Assuntos
Encéfalo/metabolismo , Hormônio Liberador da Corticotropina/genética , DNA Complementar/genética , Enguias/genética , Hormônio Liberador de Gonadotropina/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , Hormônio Liberador da Corticotropina/química , Hormônio Liberador da Corticotropina/metabolismo , Feminino , Imuno-Histoquímica , Japão , Masculino , Dados de Sequência Molecular , Hipófise/citologia , Hipófise/metabolismo , Homologia de Sequência de Aminoácidos
10.
Mol Cell Endocrinol ; 580: 112101, 2024 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-37923055

RESUMO

Terrestrial vertebrates have a population of androgen-dependent vasotocin (VT)-expressing neurons in the extended amygdala that are more abundant in males and mediate male-typical social behaviors, including aggression. Teleosts lack these neurons but instead have novel male-specific VT-expressing neurons in the tuberal hypothalamus. Here we found in medaka that vt expression in these neurons is dependent on post-pubertal gonadal androgens and that androgens can act on these neurons to directly stimulate vt transcription via the androgen receptor subtype Ara. Furthermore, administration of exogenous VT induced aggression in females and alterations in the androgen milieu led to correlated changes in the levels of tuberal hypothalamic vt expression and aggression in both sexes. However, genetic ablation of vt failed to prevent androgen-induced aggression in females. Collectively, our results demonstrate a marked androgen dependence of male-specific vt expression in the teleost tuberal hypothalamus, although its relevance to male-typical aggression needs to be further validated.


Assuntos
Agressão , Oryzias , Animais , Feminino , Masculino , Agressão/fisiologia , Androgênios/farmacologia , Androgênios/metabolismo , Comportamento Sexual Animal/fisiologia , Vasotocina/metabolismo , Oryzias/metabolismo , Hipotálamo/metabolismo
11.
Nat Commun ; 15(1): 5342, 2024 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-38937445

RESUMO

In vertebrates, folliculogenesis and ovulation are regulated by two distinct pituitary gonadotropins: follicle-stimulating hormone (FSH) and luteinizing hormone (LH). Currently, there is an intriguing consensus that a single hypothalamic neurohormone, gonadotropin-releasing hormone (GnRH), regulates the secretion of both FSH and LH, although the required timing and functions of FSH and LH are different. However, recent studies in many non-mammalian vertebrates indicated that GnRH is dispensable for FSH function. Here, by using medaka as a model teleost, we successfully identify cholecystokinin as the other gonadotropin regulator, FSH-releasing hormone (FSH-RH). Our histological and in vitro analyses demonstrate that hypothalamic cholecystokinin-expressing neurons directly affect FSH cells through the cholecystokinin receptor, Cck2rb, thereby increasing the expression and release of FSH. Remarkably, the knockout of this pathway minimizes FSH expression and results in a failure of folliculogenesis. Here, we propose the existence of the "dual GnRH model" in vertebrates that utilize both FSH-RH and LH-RH.


Assuntos
Hormônio Foliculoestimulante , Hormônio Liberador de Gonadotropina , Hipotálamo , Oryzias , Animais , Hormônio Liberador de Gonadotropina/metabolismo , Hormônio Liberador de Gonadotropina/genética , Hormônio Foliculoestimulante/metabolismo , Hormônio Foliculoestimulante/genética , Feminino , Oryzias/metabolismo , Oryzias/genética , Hipotálamo/metabolismo , Neurônios/metabolismo , Hormônio Luteinizante/metabolismo , Folículo Ovariano/metabolismo , Ovulação/genética
12.
Am J Physiol Regul Integr Comp Physiol ; 304(6): R423-34, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23364529

RESUMO

Cardiac natriuretic peptides (atrial natriuretic peptide, ANP; b-type natriuretic peptide, BNP; ventricular natriuretic peptide, VNP) and their direct ancestor C-type natriuretic peptide 3 (CNP3) exert potent osmoregulatory actions in fish. However, very little is known about their roles in embryonic osmoregulation. In this study, we performed loss-of-function analysis using euryhaline medaka (Oryzias latipes), which has lost ANP and VNP during evolution and thus possesses only BNP and CNP3. We found that the maintenance of whole-body osmolality in seawater embryos was impaired by the knockdown of BNP+OLGC7 (BNP receptor) or CNP3 alone from 1 day postfertilization, and the CNP3 knockdown was accompanied by greater water loss. The impaired osmoregulation in the knockdown embryos was not due to the suppressed expression of major transporters for NaCl excretion via ionocytes or of key enzyme genes for metabolic water production, but to the impaired blood circulation to the yolk-sac membrane caused by abnormal heart development. We detected a strong positive correlation between impaired blood circulation and increased body fluid osmolality and pharmacological blockade of blood flow increased body fluid osmolality in seawater embryos. We also found that the exaggerated water loss in CNP3 knockdown embryos is related to the failure to suppress aquaporin (AQP3, AQP4, and AQP9) gene expression. These results show that CNP3 decrease water permeability of body surfaces and that both BNP and CNP3 ensure sufficient blood flow to the yolk-sac membrane for efficient salt excretion by ionocytes and sufficient water production by yolk metabolism to promote seawater adaptation during early development in medaka.


Assuntos
Adaptação Fisiológica/fisiologia , Peptídeos Natriuréticos/metabolismo , Oryzias/embriologia , Oryzias/metabolismo , Equilíbrio Hidroeletrolítico/fisiologia , Animais , Técnicas de Silenciamento de Genes , Oryzias/genética , Água do Mar , Equilíbrio Hidroeletrolítico/genética
13.
Dev Dyn ; 241(11): 1665-77, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22972610

RESUMO

BACKGROUND: Luteinizing hormone (LH) and follicle stimulating hormone (FSH), produced in gonadotrope cells in the adenohypophysis are key regulators of vertebrate reproduction. The differential regulation of these hormones, however, is poorly understood and little is known about gonadotrope embryonic development. We developed a stable transgenic line of medaka with the LH beta subunit gene (lhb) promotor driving green fluorescent protein (gfp) expression to characterize development of LH-producing gonadotropes in whole larvae and histological sections. Additionally, developmental and tissue-specific gene expression was examined. RESULTS: The lhb gene is maternally expressed during early embryogenesis. Transcript levels increase by stage 21 (36 hours post fertilization [hpf]) and then decrease during continued larval development. Examination of the expression of pituitary marker genes show that LH-producing cells are initially localized outside the primordial pituitary, and they were localized to the developing gut tube by 32 hpf. At hatching, lhb-GFP is clearly detected in the gut epithelium and in the anterior digestive tract. lhb-GFP expression later consolidate in the developing pituitary by 2 weeks postfertilization. CONCLUSIONS: During embryonic development, lhb is primarily expressed outside the central nervous system and pituitary. The novel expression of lhb in the embryonic gut suggests that LH has a hitherto unidentified developmental function.


Assuntos
Hormônio Luteinizante/metabolismo , Oryzias/metabolismo , Animais , Animais Geneticamente Modificados , Regulação da Expressão Gênica no Desenvolvimento/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hibridização In Situ , Hormônio Luteinizante/genética , Oryzias/genética , Reação em Cadeia da Polimerase
14.
PNAS Nexus ; 2(12): pgad413, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38111823

RESUMO

Secretogranin 2 (Scg2) is a member of the secretogranin/chromogranin family of proteins that is involved in neuropeptide and hormone packaging to secretory granules and serves as a precursor for several secreted pleiotropic peptides. A recent study in zebrafish showed that the teleost Scg2 orthologs, scg2a and scg2b, play an important role in mating behavior, but its modes of action and regulatory mechanisms remain unclear. In this study, we identify scg2a in another teleost species, medaka, by transcriptomic analysis as a gene that is expressed in an ovarian secretion-dependent manner in a group of neurons relevant to female sexual receptivity, termed FeSP neurons. Investigation of scg2a expression in the FeSP neurons of estrogen receptor (Esr)-deficient medaka revealed that it is dependent on estrogen signaling through Esr2b, the major determinant of female-typical mating behavior. Generation and characterization of scg2a-deficient medaka showed no overt changes in secretory granule packaging in FeSP neurons. This, along with the observation that Scg2a and neuropeptide B, a major neuropeptide produced by FeSP neurons, colocalize in a majority of secretory granules, suggests that Scg2a mainly serves as a precursor for secreted peptides that act in conjunction with neuropeptide B. Further, scg2a showed sexually biased expression in several brain nuclei implicated in mating behavior. However, we found no significant impact of scg2a deficiency on the performance of mating behavior in either sex. Collectively, our results indicate that, although perhaps not essential for mating behavior, scg2a acts in an estrogen/Esr2b signaling-dependent manner in neurons that are relevant to female sexual receptivity.

15.
Nat Commun ; 14(1): 1428, 2023 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-36918573

RESUMO

Teleost fishes exhibit complex sexual characteristics in response to androgens, such as fin enlargement and courtship display. However, the molecular mechanisms underlying their evolutionary acquisition remain largely unknown. To address this question, we analyse medaka (Oryzias latipes) mutants deficient in teleost-specific androgen receptor ohnologs (ara and arb). We discovered that neither ar ohnolog was required for spermatogenesis, whilst they appear to be functionally redundant for the courtship display in males. However, both were required for reproductive success: ara for tooth enlargement and the reproductive behaviour eliciting female receptivity, arb for male-specific fin morphogenesis and sexual motivation. We further showed that differences between the two ar ohnologs in their transcription, cellular localisation of their encoded proteins, and their downstream genetic programmes could be responsible for the phenotypic diversity between the ara and arb mutants. These findings suggest that the ar ohnologs have diverged in two ways: first, through the loss of their roles in spermatogenesis and second, through gene duplication followed by functional differentiation that has likely resolved the pleiotropic roles derived from their ancestral gene. Thus, our results provide insights into how genome duplication impacts the massive diversification of sexual characteristics in the teleost lineage.


Assuntos
Oryzias , Receptores Androgênicos , Animais , Masculino , Feminino , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Antagonistas de Receptores de Angiotensina , Inibidores da Enzima Conversora de Angiotensina , Peixes/genética , Peixes/metabolismo , Evolução Biológica , Evolução Molecular , Oryzias/genética , Oryzias/metabolismo
16.
Proc Biol Sci ; 279(1749): 5014-23, 2012 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-23075834

RESUMO

To dissect the molecular and cellular basis of sexual differentiation of the teleost brain, which maintains marked sexual plasticity throughout life, we examined sex differences in neural expression of all subtypes of nuclear oestrogen and androgen receptors (ER and AR) in medaka. All receptors were differentially expressed between the sexes in specific nuclei in the forebrain. The most pronounced sex differences were found in several nuclei in the ventral telencephalic and preoptic areas, where ER and AR expression were prominent in females but almost completely absent in males, indicating that these nuclei represent female-specific target sites for both oestrogen and androgen in the brain. Subsequent analyses revealed that the female-specific expression of ER and AR is not under the direct control of sex-linked genes but is instead regulated positively by oestrogen and negatively by androgen in a transient and reversible manner. Taken together, the present study demonstrates that sex-specific target sites for both oestrogen and androgen occur in the brain as a result of the activational effects of gonadal steroids. The consequent sex-specific but reversible steroid sensitivity of the adult brain probably contributes substantially to the process of sexual differentiation and the persistent sexual plasticity of the teleost brain.


Assuntos
Encéfalo/metabolismo , Proteínas de Peixes/metabolismo , Regulação da Expressão Gênica , Oryzias/metabolismo , Receptores Androgênicos/metabolismo , Receptores de Estrogênio/metabolismo , Androgênios/metabolismo , Animais , Estrogênios/metabolismo , Feminino , Proteínas de Peixes/genética , Masculino , Receptores Androgênicos/genética , Receptores de Estrogênio/genética , Caracteres Sexuais , Esteroides/metabolismo
17.
Zoological Lett ; 8(1): 10, 2022 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-35879745

RESUMO

Generally, successful testis transplantation has been considered to require immune suppression in the recipient to avoid rejection of the transplanted tissue. In the present study, we demonstrate in medaka that allogeneic adult testicular tissue will engraft in adult recipients immediately after partial castration without the use of immunosuppressive drugs. The allografted testes are retained in the recipient's body for at least 3 months and are able to produce viable sperm that yield offspring after natural mating. Some recipients showed a high frequency (over 60%) of offspring derived from spermatozoa produced by the transplanted testicular tissue. Histological analyses showed that allografted testicular tissues included both germ cells and somatic cells that had become established within an immunocompetent recipient testis. The relative simplicity of this testis transplantation approach will benefit investigations of the basic processes of reproductive immunology and will improve the technique of gonadal tissue transplantation.

18.
Endocrinology ; 163(2)2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34962983

RESUMO

Animals properly perform sexual behaviors by using multiple sensory cues. However, neural mechanisms integrating multiple sensory cues and regulating motivation for sexual behaviors remain unclear. Here, we focused on peptidergic neurons, terminal nerve gonadotropin-releasing hormone (TN-GnRH) neurons, which receive inputs from various sensory systems and co-express neuropeptide FF (NPFF) in addition to GnRH. Our behavioral analyses using knockout medaka of GnRH (gnrh3) and/or NPFF (npff) demonstrated that some sexual behavioral repertoires were delayed, not disrupted, in gnrh3 and npff single knockout males, while the double knockout appeared to alleviate the significant defects that were observed in single knockouts. We also found anatomical evidence to show that both neuropeptides modulate the sexual behavior-controlling brain areas. Furthermore, we demonstrated that NPFF activates neurons in the preoptic area via indirect pathway, which is considered to induce the increase in motivation for male sexual behaviors. Considering these results, we propose a novel mechanism by which co-existing peptides of the TN-GnRH neurons, NPFF, and GnRH3 coordinately modulate certain neuronal circuit for the control of behavioral motivation. Our results may go a long way toward understanding the functional significance of peptidergic neuromodulation in response to sensory information from the external environments.


Assuntos
Hormônio Liberador de Gonadotropina/fisiologia , Oligopeptídeos/fisiologia , Oryzias , Ácido Pirrolidonocarboxílico/análogos & derivados , Comportamento Sexual Animal/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Encéfalo/metabolismo , Química Encefálica , Feminino , Técnicas de Inativação de Genes , Hormônio Liberador de Gonadotropina/análise , Hormônio Liberador de Gonadotropina/genética , Masculino , Neurônios/química , Neurônios/fisiologia , Oligopeptídeos/análise , Oligopeptídeos/genética , Filogenia , Ácido Pirrolidonocarboxílico/análise , Alinhamento de Sequência
19.
Commun Biol ; 5(1): 1215, 2022 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-36357668

RESUMO

In vertebrates, female receptivity to male courtship is highly dependent on ovarian secretion of estrogens and prostaglandins. We recently identified female-specific neurons in the medaka (Oryzias latipes) preoptic area that express Npba, a neuropeptide mediating female sexual receptivity, in response to ovarian estrogens. Here we show by transcriptomic analysis that these neurons express a multitude of neuropeptides, in addition to Npba, in an ovarian-dependent manner, and we thus termed them female-specific, sex steroid-responsive peptidergic (FeSP) neurons. Our results further revealed that FeSP neurons express a prostaglandin E2 receptor gene, ptger4b, in an ovarian estrogen-dependent manner. Behavioral and physiological examination of ptger4b-deficient female medaka found that they exhibit increased sexual receptivity while retaining normal ovarian function and that their FeSP neurons have reduced firing activity and impaired neuropeptide release. Collectively, this work provides evidence that prostaglandin E2/Ptger4b signaling mediates the estrogenic regulation of FeSP neuron activity and female sexual receptivity.


Assuntos
Neuropeptídeos , Oryzias , Animais , Feminino , Masculino , Oryzias/genética , Receptores de Prostaglandina E , Estrogênios , Neurônios , Neuropeptídeos/genética , Prostaglandinas
20.
Gen Comp Endocrinol ; 173(2): 253-8, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21663743

RESUMO

The pituitary gonadotropins, follicle-stimulating hormone (FSH) and luteinizing hormone (LH), are essential for the control of vertebrate reproduction. Although the molecular structures of these two hormones are well conserved from teleosts to mammals, some studies report differences in their regulatory mechanisms of gene expression between teleosts and tetrapods. In the present study, we examined the molecular evolution of the gonadotropin gene loci in vertebrates and found that there is a syntenic conservation among the teleost fshb and tetrapod fshb and lhb loci. However, the teleost lhb locus has no syntenic homology to either tetrapod lhb or teleost fshb; this fact suggests that an extensive genome-wide rearrangement of the lhb locus, caused by an accelerated genome evolution speed after the third round of genome-wide duplication, occurred in the teleost lineage. We subsequently demonstrated by double labeling in situ hybridization using a teleost medaka that the fshb and lhb genes in teleosts are expressed in completely separate cellular populations in the pituitary, which is different in tetrapods. Furthermore, the expression analysis in ovariectomized and steroid-treated medaka revealed that, under breeding conditions, the expression of the medaka LHß was down-regulated by ovariectomy and recovered by treatment with gonadal steroids; this result is also completely opposite in mammals, where the steroids have negative-feedback effects on LHß expression. We suggest that these differences between teleosts and mammals in the cellular expression pattern and dynamic expressional changes of the lhb gene are the result of the drastic changes in the genomic environment of the lhb gene that occurred early in teleost evolution.


Assuntos
Peixes/genética , Gonadotropinas/genética , Hormônio Luteinizante/genética , Animais , Evolução Molecular , Hormônio Foliculoestimulante/genética , Hibridização In Situ , Oryzias/genética , Hipófise/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA