Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Physiol Genomics ; 55(3): 113-131, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36645671

ABSTRACT

Solute carrier 12 (Slc12) is a family of electroneutral cation-coupled chloride (Cl-) cotransporters. Na+/K+/2Cl- (Nkcc) and Na+/Cl- cotransporters (Ncc) belong to the Nkcc/Ncc subfamily. Human and mouse possess one gene for the Na+/Cl- cotransporter (ncc gene: slc12a3), whereas teleost fishes possess multiple ncc genes, slc12a3 (ncc1) and slc12a10 (ncc2), in addition to their species-specific paralogs. Amphibians and squamates have two ncc genes: slc12a3 (ncc1) and ncc3. However, the evolutionary relationship between slc12a10 and ncc3 remains unresolved, and the presence of slc12a10 (ncc2) in mammals has not been clarified. Synteny and phylogenetic analyses of vertebrate genome databases showed that ncc3 is the ortholog of slc12a10, and slc12a10 is present in most ray-finned fishes, coelacanths, amphibians, reptiles, and a few mammals (e.g., platypus and horse) but pseudogenized or deleted in birds, most mammals, and some ray-finned fishes (pufferfishes). This shows that slc12a10 is widely present among bony vertebrates and pseudogenized or deleted independently in multiple lineages. Notably, as compared with some fish that show varied slc12a10 tissue expression profile, spotted gar, African clawed frog, red-eared slider turtle, and horse express slc12a10 in the ovaries or premature gonads. In horse tissues, an unexpectedly large number of splicing variants for Slc12a10 have been cloned, many of which encode truncated forms of Slc12a10, suggesting that the functional constraints of horse slc12a10 are weakened, which may be in the process of becoming a pseudogene. Our results elaborate on the evolution of Nkcc/Ncc subfamily of Slc12 in vertebrates.NEW & NOTEWORTHY slc12a10 is not a fish-specific gene and is present in a few mammals (e.g., platypus and horse), non-avian reptiles, amphibians, but was pseudogenized or deleted in most mammals (e.g., human, mouse, cat, cow, and rhinoceros), birds, and some ray-finned fishes (pufferfishes).


Subject(s)
Platypus , Female , Cattle , Animals , Humans , Horses , Mice , Solute Carrier Family 12, Member 3 , Phylogeny , Fishes/genetics , Reptiles/genetics , Birds , Amphibians/genetics
2.
Front Physiol ; 13: 939114, 2022.
Article in English | MEDLINE | ID: mdl-35812342

ABSTRACT

The kidney is an important organ that maintains body fluid homeostasis in seawater and freshwater teleost fishes. Seawater teleosts excrete sulfate and magnesium in small amounts of isotonic urine, and freshwater teleosts excrete water in large amounts of hypo-osmotic urine. The volume, osmolality, and ionic compositions of the urine are regulated mainly by membrane transport proteins expressed in the renal tubular epithelial cells. Gene expression, immunohistochemical, and functional analyses of the fish kidney identified membrane transport proteins involved in the secretion of sulfate and magnesium ions by the proximal tubules and reduction of urine volume by the collecting ducts in seawater teleosts, and excretion of water as hypotonic urine by the distal tubules and collecting ducts in freshwater teleosts. These studies promote an understanding of how the kidney contributes to the seawater and freshwater acclimation of teleosts at the molecular level.

3.
Elife ; 92020 08 12.
Article in English | MEDLINE | ID: mdl-32783809

ABSTRACT

Recent studies in mice demonstrate that a subset of neurons in the medial preoptic area (MPOA) that express galanin play crucial roles in regulating parental behavior in both sexes. However, little information is available on the function of galanin in social behaviors in other species. Here, we report that, in medaka, a subset of MPOA galanin neurons occurred nearly exclusively in males, resulting from testicular androgen stimulation. Galanin-deficient medaka showed a greatly reduced incidence of male-male aggressive chases. Furthermore, while treatment of female medaka with androgen induced male-typical aggressive acts, galanin deficiency in these females attenuated the effect of androgen on chases. Given their male-biased and androgen-dependent nature, the subset of MPOA galanin neurons most likely mediate androgen-dependent male-male chases. Histological studies further suggested that variability in the projection targets of the MPOA galanin neurons may account for the species-dependent functional differences in these evolutionarily conserved neural substrates.


Subject(s)
Androgens/metabolism , Galanin/metabolism , Oryzias/metabolism , Sex Characteristics , Animals , Animals, Genetically Modified , Cell Line , Female , Galanin/genetics , Gene Expression , HEK293 Cells , Humans , Male , Neurons/metabolism , Preoptic Area/cytology , Preoptic Area/metabolism , Receptors, Galanin/metabolism , Reproduction , Signal Transduction , Steroids/metabolism
4.
Elife ; 82019 08 06.
Article in English | MEDLINE | ID: mdl-31383257

ABSTRACT

Male and female animals display innate sex-specific mating behaviors. In teleost fish, altering the adult sex steroid milieu can effectively reverse sex-typical mating behaviors, suggesting remarkable sexual lability of their brains as adults. In the teleost medaka, neuropeptide B (NPB) is expressed female-specifically in the brain nuclei implicated in mating behavior. Here, we demonstrate that NPB is a direct mediator of estrogen action on female mating behavior, acting in a female-specific but reversible manner. Analysis of regulatory mechanisms revealed that the female-specific expression of NPB is dependent on direct transcriptional activation by estrogen via an estrogen-responsive element and is reversed in response to changes in the adult sex steroid milieu. Behavioral studies of NPB knockouts revealed that female-specific NBP mediates female receptivity to male courtship. The female-specific NPB signaling identified herein is presumably a critical element of the neural circuitry underlying sexual dimorphism and lability of mating behaviors in teleosts.


Subject(s)
Neuropeptides/metabolism , Oryzias/physiology , Sexual Behavior, Animal/drug effects , Animals , Estrogens/metabolism , Female , Gene Expression Regulation/drug effects
5.
Sci Rep ; 8(1): 16855, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30442908

ABSTRACT

The neuroplastic mechanisms in the fish brain that underlie sex reversal remain unknown. Gonadotropin-releasing hormone 3 (GnRH3) neurons control male reproductive behaviours in Mozambique tilapia and show sexual dimorphism, with males having a greater number of GnRH3 neurons. Treatment with androgens such as 11-ketotestosterone (KT), but not 17ß-estradiol, increases the number of GnRH3 neurons in mature females to a level similar to that observed in mature males. Compared with oestrogen, the effect of androgen on neurogenesis remains less clear. The present study examined the effects of 11-KT, a non-aromatizable androgen, on cellular proliferation, neurogenesis, generation of GnRH3 neurons and expression of cell cycle-related genes in mature females. The number of proliferating cell nuclear antigen-positive cells was increased by 11-KT. Simultaneous injection of bromodeoxyuridine and 11-KT significantly increased the number of newly-generated (newly-proliferated) neurons, but did not affect radial glial cells, and also resulted in newly-generated GnRH3 neurons. Transcriptome analysis showed that 11-KT modulates the expression of genes related to the cell cycle process. These findings suggest that tilapia could serve as a good animal model to elucidate the effects of androgen on adult neurogenesis and the mechanisms for sex reversal in the fish brain.


Subject(s)
Androgens/pharmacology , Brain/cytology , Brain/metabolism , Gonadotropin-Releasing Hormone/metabolism , Neurogenesis/drug effects , Neurons/metabolism , Tilapia/metabolism , Animals , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Proliferation/drug effects , Cerebral Ventricles/cytology , Female , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , Image Processing, Computer-Assisted , Neuroglia/cytology , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/drug effects , Proliferating Cell Nuclear Antigen/metabolism , Testosterone/analogs & derivatives , Testosterone/pharmacology
6.
Gen Comp Endocrinol ; 223: 47-53, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26433060

ABSTRACT

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.


Subject(s)
Brain/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Oryzias/metabolism , Receptors, Glucocorticoid/genetics , Animals , Female , In Situ Hybridization , Male , Neurons/cytology , Oryzias/genetics , Oryzias/growth & development , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Glucocorticoid/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sex Factors
7.
Endocrinology ; 156(8): 2949-57, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26030477

ABSTRACT

The CRH family of neuropeptides, including CRH and urocortins, plays pivotal roles in the regulation of physiological and behavioral stress responses in vertebrates. In this study, we identified a previously undescribed member of the CRH family of peptides in a teleost fish species (medaka; Oryzias latipes) and named this peptide teleocortin (Tcn). Medaka Tcn is a 41-amino acid polypeptide derived from the C terminus of a larger precursor protein that is encoded by a 2-exon gene, thus sharing common structural features with known CRH family peptides. tcn was found exclusively in teleost fish. Phylogenetic analysis suggested that tcn probably has an ancient origin but was lost from the tetrapod lineage shortly after the divergence of the teleost and tetrapod lineages. In the medaka brain, tcn was expressed in nuclei of the telencephalon, preoptic area, hypothalamus, tegmentum, and isthmic region. Because none of these nuclei have been implicated in the control of ACTH secretion from the pituitary, Tcn may exert its effects centrally in the brain rather than via stimulation of the pituitary-adrenal/interrenal axis. Most, if not all, tcn-expressing neurons also expressed crh, suggesting that Tcn and Crh share common physiological functions. Moreover, Tcn activated Crh receptors 1 and 2 with equivalent or slightly higher potency than Crh, further suggesting that these peptides share common functions. Taken together, these data identified Tcn as a novel, teleost-specific member of the CRH family of peptides that may act centrally with Crh to regulate physiological and behavioral stress responses.


Subject(s)
Corticotropin-Releasing Hormone/genetics , Fish Proteins/physiology , Multigene Family , Neuropeptides/genetics , Neuropeptides/physiology , Oryzias/genetics , Amino Acid Sequence , Animals , Brain/metabolism , Cloning, Molecular , Fish Proteins/genetics , Fish Proteins/isolation & purification , Molecular Sequence Data , Neuropeptides/isolation & purification , Phylogeny , Sequence Homology, Amino Acid , Tissue Distribution , Urocortins/genetics
8.
Biochem Biophys Res Commun ; 445(1): 113-9, 2014 Feb 28.
Article in English | MEDLINE | ID: mdl-24491545

ABSTRACT

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.


Subject(s)
Brain/metabolism , Ciliary Neurotrophic Factor/genetics , Fish Proteins/genetics , Oryzias/genetics , Y Chromosome/genetics , Amino Acid Sequence , Animals , Ciliary Neurotrophic Factor/classification , Estradiol/pharmacology , Female , Gene Expression/drug effects , Gene Expression Profiling , Genetic Linkage , Male , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Phylogeny , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Sex Factors , Testosterone/analogs & derivatives , Testosterone/pharmacology , Time Factors
9.
Endocrinology ; 155(3): 1021-32, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24424038

ABSTRACT

In the brain of medaka (Oryzias latipes), a teleost fish, we recently found that the supracommissural/posterior nuclei of the ventral telencephalic area (Vs/Vp) and the magnocellular/gigantocellular portions of the magnocellular preoptic nucleus (PMm/PMg) express estrogen receptor (ER) and androgen receptor (AR) specifically in females. This finding led us to postulate that sex steroid hormones might induce gene expression unique to females in these nuclei. In the present study, we searched for genes differentially expressed between the sexes in the medaka brain and identified the gene encoding neuropeptide B (npb) as being female-specifically expressed in Vs/Vp and PMm/PMg. As expected, the neurons expressing npb female-specifically constituted a significant proportion of the ER/AR-expressing neurons in these nuclei. Subsequent analyses provided evidence that the female-specific expression of npb in Vs/Vp and PMm/PMg results from the reversible and transient action of estrogens secreted from the ovary and that this estrogenic action is most likely mediated by the direct transcriptional activation of npb through an estrogen-responsive element in its proximal promoter region. Vs/Vp and PMm/PMg are generally recognized in teleost fish as the sites where neurons expressing 2 other neuropeptides, isotocin and vasotocin, are present, but the female-specific npb/ER/AR-expressing neurons were distinct from, although adjacent to, isotocin and vasotocin neurons. Taken together, these data demonstrate that npb is female-specifically expressed in novel, as-yet undefined populations of Vs/Vp and PMm/PMg neurons, resulting from the direct stimulatory action of ovarian estrogens via female-specific ER in these neurons.


Subject(s)
Gene Expression Regulation , Neuropeptides/metabolism , Preoptic Area/metabolism , Telencephalon/metabolism , Animals , Brain Mapping/methods , Estrogens/metabolism , Female , Male , Neurons/metabolism , Neurotransmitter Agents/metabolism , Oryzias , Oxytocin/analogs & derivatives , Oxytocin/metabolism , Promoter Regions, Genetic , Receptors, Androgen/metabolism , Receptors, Estrogen/metabolism , Sex Factors , Time Factors , Vasotocin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...