Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Gen Comp Endocrinol ; 344: 114372, 2023 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-37652166

RESUMO

SH3 domain binding kinase 1 (SBK1) is a serine/threonine kinase that belongs to the new kinase family (NFK) with limited information on its function. Previous studies reported that SBK1 plays a role in memory formation, lipid metabolism, and cancer cell progression. Nevertheless, the regulatory mechanism of Sbk1 expression in various tissues remains unknown. We report here that Sbk1 expression in mouse hepatocytes was downregulated by glucocorticoid, whereas saturated and unsaturated fatty acids were stimulators of Sbk1 expression. The regulatory role of glucocorticoid and fatty acid was further confirmed by the Sbk1 promoter assay, which aligned with the presence of several glucocorticoid-response elements (GRE) and peroxisome proliferator responsive elements (PPRE) in the mouse Sbk1 promoter. The inhibitory effect of glucocorticoids on hepatic Sbk1 expression and protein content could also be demonstrated in vivo after prednisolone injection. Moreover, the expression of SBK1 in goldfish (gfSBK1) was also sensitive to glucocorticoid suppression as their mouse orthologues. In contrast, insulin had a differential action on SBK1 expression that it promoted the expression of all SBK1 isoforms in the goldfish hepatocytes but inhibited Sbk1 expression in the mouse hepatocytes. Together, our findings indicate that SBK1 expression is hormone- and nutrient-sensitive with a species-specific response.


Assuntos
Carpa Dourada , Fatores de Transcrição , Camundongos , Animais , Fatores de Transcrição/metabolismo , Carpa Dourada/genética , Carpa Dourada/metabolismo , Glucocorticoides/metabolismo , Domínios de Homologia de src , Fígado/metabolismo
2.
Gen Comp Endocrinol ; 265: 90-96, 2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29355530

RESUMO

Spexin (SPX), a novel peptide coevolved with the galanin/kisspeptin family, was first identified by bioinformatics prior to its protein purification/functional studies. Its mature peptide is highly conserved among different vertebrate classes. Based on the studies in mammals and fish models, SPX was found to be widely distributed at tissue level, secreted into systemic circulation, identified at notable levels in central nervous system and peripheral tissues, and has been confirmed/implicated in multiple functions in different tissues/organs, suggesting that SPX may serve as a neuroe​ndocrine signal with pleotropic functions. In this article, different isoforms of SPX and their binding with their cognate receptors GalR2 and GalR3, the biological functions of SPX reported in mammals including GI tract movement, energy balance and weight loss, fatty acid uptake, glucose homeostasis, nociception and cardiovascular/renal functions, as well as the recent findings in fish models regarding the role of SPX in reproduction and feeding control will be reviewed with interesting questions for future investigations.


Assuntos
Sistemas Neurossecretores/metabolismo , Hormônios Peptídicos/metabolismo , Sequência de Aminoácidos , Animais , Peixes/metabolismo , Galanina/metabolismo , Humanos , Mamíferos/metabolismo , Hormônios Peptídicos/química , Filogenia , Ligação Proteica
3.
Am J Physiol Endocrinol Metab ; 307(10): E872-84, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25184991

RESUMO

Although the importance of kisspeptin in the pituitary is firmly established, the signaling mechanisms for the pituitary actions of kisspeptin are still largely unknown. Somatolactin (SL), a member of the growth hormone (GH)/prolactin (PRL) family, is a pituitary hormone with pleiotropic functions in fish, but its regulation by kisspeptin has not been examined. To investigate the functional role of kisspeptin in SL regulation, expression of two paralogues of goldfish Kiss1 receptors (Kiss1ra and Kiss1rb) were confirmed in immunoidentified SLα but not SLß cells isolated by RT-PCR coupled with laser capture microdissection. In goldfish pituitary cells prepared from neurointermediate lobe (NIL), synthetic goldfish Kiss decapeptides (gKiss1-10 and gKiss2-10) could increase SLα release. Consistent with the lack of Kiss1r expression in SLß cells, SLß release was not altered by kisspeptin stimulation. In parallel experiments, goldfish gKiss1-10 could elevate cyclic adenosine monophosphate (cAMP) production, upregulate protein kinase A (PKA) and protein kinase C (PKC) activities, and trigger a rapid rise in intracellular Ca(2+) levels in goldfish NIL cells. Using a pharmacological approach, cAMP/PKA and phospholipase C (PLC)/PKC pathways and subsequent activation of Ca(2+)/calmodulin (CaM)-dependent cascades were shown to be involved in SLα release induced by gKiss1-10. Apparently, the Ca(2+)-dependent cascades were triggered by extracellular Ca(2+) entry via voltage-sensitive Ca(2+) channels and mobilization of inositol trisphosphate-sensitive intracellular Ca(2+) stores. Our results demonstrate that gKiss1-10 can act directly at the pituitary level to trigger SLα release via a complex network of post-receptor signaling mechanisms.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Proteínas de Peixes/metabolismo , Glicoproteínas/metabolismo , Kisspeptinas/metabolismo , Adeno-Hipófise Parte Intermédia/metabolismo , Hormônios Hipofisários/metabolismo , Proteína Quinase C/metabolismo , Fosfolipases Tipo C/metabolismo , Animais , Carpa Dourada , Hipófise/citologia , Hipófise/metabolismo , Adeno-Hipófise Parte Intermédia/citologia , Transdução de Sinais
4.
Am J Physiol Regul Integr Comp Physiol ; 307(6): R755-68, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25009216

RESUMO

Somatostain (SS) is known to inhibit growth hormone (GH) and prolactin (PRL) secretion. Somatolactin (SL) is a member of the GH/PRL family, but its regulation by goldfish brain somatostatin-28 (gbSS-28) has not been examined. To this end, the structural identity of goldfish SLα was established by 5'/3'-rapid amplification of cDNA ends. As revealed by in situ hybridization and immunohistochemical staining, the expression of SL isoforms was detected in pituitary cells located in the neurointermediate lobe (NIL). The transcripts of goldfish SS receptor 5a (Sst5a) but not Sst1b, Sst2, or Sst3a were detected in the goldfish NIL cells by RT-PCR. In goldfish pituitary cells, gbSS-28 not only had an inhibitory effect on basal SLα and SLß mRNA levels but also could abolish insulin-like growth factor-stimulated SL gene expression. In primary cultures of goldfish NIL cells, gbSS-28 reduced forskolin-stimulated total cAMP production. With the use of a pharmacological approach, the adenylate cyclase (AC)/cAMP and phospholipase C (PLC)/inositol trisphosphate (IP3)/protein kinase C (PKC) cascades were shown to be involved in gbSS-28-inhibited SLα mRNA expression. Similar postreceptor signaling cascades were also observed for gbSS-28-reduced SLß mRNA expression, except that PKC coupling to PLC was not involved. These results provide evidence that gbSS-28 can inhibit SLα and SLß gene expression at the goldfish pituitary level via Sst5 through differential coupling of AC/cAMP and PLC/IP3/PKC cascades.


Assuntos
Proteínas de Peixes/metabolismo , Glicoproteínas/metabolismo , Carpa Dourada/metabolismo , Hipófise/efeitos dos fármacos , Hormônios Hipofisários/metabolismo , Somatostatina-28/farmacologia , Adenilil Ciclases/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Células Cultivadas , Clonagem Molecular , AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Regulação para Baixo , Proteínas de Peixes/genética , Glicoproteínas/genética , Carpa Dourada/genética , Inositol 1,4,5-Trifosfato/metabolismo , Dados de Sequência Molecular , Hipófise/metabolismo , Hormônios Hipofisários/genética , Cultura Primária de Células , Proteína Quinase C/metabolismo , RNA Mensageiro/metabolismo , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Fosfolipases Tipo C/metabolismo
5.
Gen Comp Endocrinol ; 208: 94-108, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-25172151

RESUMO

In mammals, neurokinin B (NKB), the gene product of the tachykinin family member TAC3, is known to be a key regulator for episodic release of luteinizing hormone (LH). Its regulatory actions are mediated by a subpopulation of kisspeptin neurons within the arcuate nucleus with co-expression of NKB and dynorphin A (commonly called the "KNDy neurons"). By forming an "autosynaptic feedback loop" within the hypothalamus, the KNDy neurons can modulate gonadotropin-releasing hormone (GnRH) pulsatility and subsequent LH release in the pituitary. NKB regulation of LH secretion has been recently demonstrated in zebrafish, suggesting that the reproductive functions of NKB may be conserved from fish to mammals. Interestingly, the TAC3 genes in fish not only encode the mature peptide of NKB but also a novel tachykinin-like peptide, namely NKB-related peptide (or neurokinin F). Recent studies in zebrafish also reveal that the neuroanatomy of TAC3/kisspeptin system within the fish brain is quite different from that of mammals. In this article, the current ideas of "KNDy neuron" model for GnRH regulation and steroid feedback, other reproductive functions of NKB including its local actions in the gonad and placenta, the revised model of tachykinin evolution from invertebrates to vertebrates, as well as the emerging story of the two TAC3 gene products in fish, NKB and NKB-related peptide, will be reviewed with stress on the areas with interesting questions for future investigations.


Assuntos
Dinorfinas/metabolismo , Peixes/metabolismo , Mamíferos/metabolismo , Neurocinina B/metabolismo , Neurônios/metabolismo , Reprodução , Animais
6.
Front Endocrinol (Lausanne) ; 15: 1399274, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38894746

RESUMO

Gonadotropin-releasing hormone (GnRH) is a key stimulator for gonadotropin secretion in the pituitary and its pivotal role in reproduction is well conserved in vertebrates. In fish models, GnRH can also induce prolactin (PRL) release, but little is known for the corresponding effect on PRL gene expression as well as the post-receptor signalling involved. Using grass carp as a model, the functional role of GnRH and its underlying signal transduction for PRL regulation were examined at the pituitary level. Using laser capture microdissection coupled with RT-PCR, GnRH receptor expression could be located in carp lactotrophs. In primary cell culture prepared from grass carp pituitaries, the native forms of GnRH, GnRH2 and GnRH3, as well as the GnRH agonist [D-Arg6, Pro9, NEt]-sGnRH were all effective in elevating PRL secretion, PRL mRNA level, PRL cell content and total production. In pituitary cells prepared from the rostral pars distalis, the region in the carp pituitary enriched with lactotrophs, GnRH not only increased cAMP synthesis with parallel CREB phosphorylation and nuclear translocation but also induced a rapid rise in cytosolic Ca2+ by Ca2+ influx via L-type voltage-sensitive Ca2+ channel (VSCC) with subsequent CaM expression and NFAT2 dephosphorylation. In carp pituitary cells prepared from whole pituitaries, GnRH-induced PRL secretion was reduced/negated by inhibiting cAMP/PKA, PLC/PKC and Ca2+/CaM/CaMK-II pathways but not the signalling events via IP3 and CaN/NFAT. The corresponding effect on PRL mRNA expression, however, was blocked by inhibiting cAMP/PKA/CREB/CBP and Ca2+/CaM/CaN/NFAT2 signalling but not PLC/IP3/PKC pathway. At the pituitary cell level, activation of cAMP/PKA pathway could also induce CaM expression and Ca2+ influx via VSCC with parallel rises in PRL release and gene expression in a Ca2+/CaM-dependent manner. These findings, as a whole, suggest that the cAMP/PKA-, PLC/PKC- and Ca2+/CaM-dependent cascades are differentially involved in GnRH-induced PRL secretion and PRL transcript expression in carp lactotrophs. During the process, a functional crosstalk between the cAMP/PKA- and Ca2+/CaM-dependent pathways may occur with PRL release linked with CaMK-II and PKC activation and PRL gene transcription caused by nuclear action of CREB/CBP and CaN/NFAT2 signalling.


Assuntos
Cálcio , Carpas , Proteínas Quinases Dependentes de AMP Cíclico , AMP Cíclico , Hormônio Liberador de Gonadotropina , Hipófise , Prolactina , Proteína Quinase C , Fosfolipases Tipo C , Animais , Carpas/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Prolactina/metabolismo , Hipófise/metabolismo , Hipófise/citologia , Proteína Quinase C/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Cálcio/metabolismo , Fosfolipases Tipo C/metabolismo , Fosfolipases Tipo C/genética , AMP Cíclico/metabolismo , Transdução de Sinais/efeitos dos fármacos , Calmodulina/metabolismo , Células Cultivadas , Expressão Gênica/efeitos dos fármacos
7.
Am J Physiol Endocrinol Metab ; 304(2): E176-86, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23193053

RESUMO

Pituitary hormones can act locally via autocrine/paracrine mechanisms to modulate pituitary functions, which represents an interesting aspect of pituitary regulation other than the traditional hypothalamic input and feedback signals from the periphery. Somatolactin, a member of the growth hormone (GH)/prolactin (PL) family, is a pleiotropic hormone with diverse functions, but its pituitary actions are still unknown. Recently, two SL isoforms, SLα and SLß, have been cloned in grass carp. Based on the sequences obtained, recombinant proteins of carp SLα and SLß with similar bioactivity in inducing pigment aggregation in carp melanophores were produced. In carp pituitary cells, SLα secretion and cell content were elevated by static incubation with recombinant carp SLα and SLß, respectively. These stimulatory actions occurred with a parallel rise in SLα mRNA level with no changes in SLß secretion, cell content, and gene expression. In contrast, SLα mRNA expression could be reduced by removing endogenous SLα and SLß with immunoneutralization. At the pituitary cell level, SLα release, cell content, and mRNA expression induced by carp SLα and SLß could be blocked by inhibiting JAK2/STAT5, PI3K/Akt, MEK1/2, and p38 MAPK, respectively. Furthermore, SLα and SLß induction also triggered rapid phosphorylation of STAT5, Akt, MEK1/2, ERK1/2, MKK3/6, and p38 MAPK. These results suggest that 1) SLα and SLß produced locally in the carp pituitary can serve as novel autocrine/paracrine stimulators for SLα secretion and synthesis and 2) SLα production induced by local release of SLα and SLß probably are mediated by the JAK2/STAT5, PI3K/Akt, and MAPK signaling pathways.


Assuntos
Carpas , Proteínas de Peixes/biossíntese , Proteínas de Peixes/metabolismo , Proteínas de Peixes/farmacologia , Glicoproteínas/biossíntese , Glicoproteínas/metabolismo , Glicoproteínas/farmacologia , Hipófise/metabolismo , Hormônios Hipofisários/biossíntese , Hormônios Hipofisários/metabolismo , Hormônios Hipofisários/farmacologia , Animais , Comunicação Autócrina/efeitos dos fármacos , Comunicação Autócrina/genética , Comunicação Autócrina/fisiologia , Carpas/genética , Carpas/metabolismo , Carpas/fisiologia , Células Cultivadas , Proteínas de Peixes/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Glicoproteínas/genética , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Sistema de Sinalização das MAP Quinases/fisiologia , Modelos Biológicos , Comunicação Parácrina/efeitos dos fármacos , Comunicação Parácrina/genética , Comunicação Parácrina/fisiologia , Hipófise/efeitos dos fármacos , Hormônios Hipofisários/genética , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/farmacologia , Via Secretória/efeitos dos fármacos , Via Secretória/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
8.
Am J Physiol Endocrinol Metab ; 305(3): E348-66, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23715729

RESUMO

Spexin (SPX) is a neuropeptide identified recently by bioinformatic approach. At present not much is known about its biological actions, and comparative studies of SPX in nonmammalian species are still lacking. To examine the structure and function of SPX in fish model, SPX was cloned in goldfish and found to be highly comparable with its mammalian counterparts. As revealed by NMR spectroscopies, goldfish SPX is composed of an α-helix from Gln(5) to Gln(14) with a flexible NH2 terminus from Asn(1) to Pro(4), and its molecular surface is largely hydrophobic except for Lys(11) as the only charged residue in the helical region. In goldfish, SPX transcripts were found to be widely expressed in various tissues, and protein expression of SPX was also detected in the brain. In vivo feeding studies revealed that SPX mRNA levels in the telencephalon, optic tectum, and hypothalamus of goldfish brain could be elevated by food intake. However, brain injection of goldfish SPX inhibited both basal and NPY- or orexin-induced feeding behavior and food consumption. Similar treatment also reduced transcript expression of NPY, AgRP, and apelin, with concurrent rises in CCK, CART, POMC, MCH, and CRH mRNA levels in different brain areas examined. The differential effects of SPX treatment on NPY, CCK, and MCH transcript expression could also be noted in vitro in goldfish brain cell culture. Our studies for the first time unveil the solution structure of SPX and its novel function as a satiety factor through differential modulation of central orexigenic and anorexigenic signals.


Assuntos
Ingestão de Alimentos/fisiologia , Carpa Dourada/fisiologia , Resposta de Saciedade/fisiologia , Sequência de Aminoácidos , Animais , Estimulantes do Apetite/farmacologia , Encéfalo/citologia , Química Encefálica/genética , Química Encefálica/fisiologia , Células Cultivadas , Dicroísmo Circular , Clonagem Molecular , Primers do DNA , DNA Complementar/biossíntese , DNA Complementar/genética , Interpretação Estatística de Dados , Espectroscopia de Ressonância Magnética , Dados de Sequência Molecular , Neuropeptídeos/metabolismo , Neuropeptídeos/farmacologia , Reação em Cadeia da Polimerase , Período Pós-Prandial/fisiologia , Distribuição Tecidual
9.
Front Endocrinol (Lausanne) ; 14: 1283298, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38027109

RESUMO

Adiponectin (AdipoQ) is an adipokine involved in glucose homeostasis and lipid metabolism. In mammals, its role in appetite control is highly controversial. To shed light on the comparative aspects of AdipoQ in lower vertebrates, goldfish was used as a model to study feeding regulation by AdipoQ in fish species. As a first step, goldfish AdipoQ was cloned and found to be ubiquitously expressed at the tissue level. Using sequence alignment, protein modeling, phylogenetic analysis and comparative synteny, goldfish AdipoQ was shown to be evolutionarily related to its fish counterparts and structurally comparable with AdipoQ in higher vertebrates. In our study, recombinant goldfish AdipoQ was expressed in E. coli, purified by IMAC, and confirmed to be bioactive via activation of AdipoQ receptors expressed in HepG2 cells. Feeding in goldfish revealed that plasma levels of AdipoQ and its transcript expression in the liver and brain areas involved in appetite control including the telencephalon, optic tectum, and hypothalamus could be elevated by food intake. In parallel studies, IP and ICV injection of recombinant goldfish AdipoQ in goldfish was effective in reducing foraging behaviors and food consumption. Meanwhile, transcript expression of orexigenic factors (NPY, AgRP, orexin, and apelin) was suppressed with parallel rises in anorexigenic factors (POMC, CART, CCK, and MCH) in the telencephalon, optic tectum and/or hypothalamus. In these brain areas, transcript signals for leptin receptor were upregulated with concurrent drops in the NPY receptor and ghrelin receptors. In the experiment with IP injection of AdipoQ, transcript expression of leptin was also elevated with a parallel drop in ghrelin mRNA in the liver. These findings suggest that AdipoQ can act as a novel satiety factor in goldfish. In this case, AdipoQ signals (both central and peripheral) can be induced by feeding and act within the brain to inhibit feeding behaviors and food intake via differential regulation of orexigenic/anorexigenic factors and their receptors. The feeding inhibition observed may also involve the hepatic action of AdipoQ by modulation of feeding regulators expressed in the liver.


Assuntos
Ingestão de Alimentos , Carpa Dourada , Animais , Ingestão de Alimentos/fisiologia , Carpa Dourada/genética , Adiponectina/metabolismo , Distribuição Tecidual , Escherichia coli/metabolismo , Filogenia , Clonagem Molecular , Proteínas Recombinantes/metabolismo , Mamíferos/metabolismo
10.
Gen Comp Endocrinol ; 179(1): 38-46, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22885559

RESUMO

It has been established that kisspeptin regulates reproduction via stimulation of hypothalamic gonadotropin-releasing hormone (GnRH) secretion, which then induces pituitary luteinizing hormone (LH) release. Kisspeptin also directly stimulates pituitary hormone release in some mammals. However, in goldfish, whether kisspeptin directly affects pituitary hormone release is controversial. In this study, synthetic goldfish kisspeptin-1((1-10)) (gKiss1) enhances LH and growth hormone (GH) release from primary cultures of goldfish pituitary cells in column perifusion. gKiss1 stimulation of LH and GH secretion were still manifested in the presence of the two native goldfish GnRHs, salmon (s)GnRH (goldfish GnRH-3) and chicken (c)GnRH-II (goldfish GnRH-2), but were attenuated by two voltage-sensitive calcium channel blockers, verapamil and nifedipine. gKiss-induced increases in intracellular Ca(2+) in Fura-2AM pre-loaded goldfish pars distalis cells were also inhibited by nifedipine. These results indicate that, in goldfish, (1) direct gKiss1 actions on pituitary LH and GH secretion exist, (2) these actions are independent of GnRH and (3) they involve Ca(2+) signalling.


Assuntos
Sinalização do Cálcio , Carpa Dourada/metabolismo , Hormônio do Crescimento/metabolismo , Kisspeptinas/farmacologia , Hormônio Luteinizante/metabolismo , Hipófise/efeitos dos fármacos , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Células Cultivadas , Nifedipino/farmacologia , Hipófise/citologia , Hipófise/metabolismo , Verapamil/farmacologia
11.
Gigascience ; 112022 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-36480030

RESUMO

Japanese eels (Anguilla japonica) are commercially important species, harvested extensively for food. Currently, this and related species (American and European eels) are challenging to breed on a commercial basis. As a result, the wild stock is used for aquaculture. Moreover, climate change, habitat loss, water pollution, and altered ocean currents affect eel populations negatively. Accordingly, the International Union for Conservation of Nature lists Japanese eels as endangered and on its red list. Here we presented a high-quality genome assembly for Japanese eels and demonstrated that large chromosome reorganizations occurred in the events of third-round whole-genome duplications (3R-WRDs). Several chromosomal fusions and fissions have reduced the ancestral protochromosomal number of 25 to 19 in the Anguilla lineage. A phylogenetic analysis of the expanded gene families showed that the olfactory receptors (group δ and ζ genes) and voltage-gated Ca2+ channels expanded significantly. Both gene families are crucial for olfaction and neurophysiology. Additional tandem and proximal duplications occurred following 3R-WGD to acquire immune-related genes for an adaptive advantage against various pathogens. The Japanese eel assembly presented here can be used to study other Anguilla species relating to evolution and conservation.


Assuntos
Duplicação Gênica , Cromossomos/genética , Filogenia
12.
Am J Physiol Endocrinol Metab ; 301(6): E1208-19, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21862722

RESUMO

Somatolactin (SL), a member of the growth hormone/prolactin family, is a pituitary hormone unique to fish models. Although SL is known to have diverse functions in fish, the mechanisms regulating its secretion and synthesis have not been fully characterized. Using grass carp pituitary cells as a model, here we examined the role of insulin-like growth factor (IGF) in SL regulation at the pituitary level. As a first step, the antisera for the two SL isoforms expressed in the carp pituitary, SLα and SLß, were produced, and their specificity was confirmed by antiserum preabsorption and immunohistochemical staining in the carp pituitary. Western blot using these antisera revealed that grass carp SLα and SLß could be N-linked glycosylated and their basal secretion and cell content in carp pituitary cells could be elevated by IGF-I and -II treatment. These stimulatory effects occurred with parallel rises in SLα and SLß mRNA levels, and these SL gene expression responses were not mimicked by insulin but blocked by IGF-I receptor inactivation. In carp pituitary cells, IGF-I and -II could induce rapid phosphorylation of IGF-I receptor, MEK1/2, ERK1/2, MKK3/6, and p38 MAPK; and SLα and SLß secretion, protein production, and mRNA expression caused by IGF-I and -II stimulation were negated by inactivating MEK1/2 and p38 MAPK. Parallel inhibition of PI3K and Akt, however, were not effective in these regards. These results, taken together, provide evidence that IGF can upregulate SL secretion and synthesis at the pituitary level via stimulation of MAPK- but not PI3K/Akt-dependent pathways.


Assuntos
Carpas , Proteínas de Peixes/biossíntese , Proteínas de Peixes/metabolismo , Glicoproteínas/biossíntese , Glicoproteínas/metabolismo , Fator de Crescimento Insulin-Like I/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Hipófise/efeitos dos fármacos , Hormônios Hipofisários/biossíntese , Hormônios Hipofisários/metabolismo , Animais , Carpas/metabolismo , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Feminino , Proteínas de Peixes/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Glicoproteínas/genética , Imuno-Histoquímica , Fator de Crescimento Insulin-Like I/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Hipófise/citologia , Hipófise/metabolismo , Hormônios Hipofisários/genética , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Estimulação Química , Regulação para Cima/efeitos dos fármacos
13.
Front Endocrinol (Lausanne) ; 12: 681646, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34276561

RESUMO

Spexin (SPX), a highly conserved neuropeptide, is known to have diverse functions and has been implicated/associated with pathological conditions, including obesity, diabetes, anorexia nervosa, and anxiety/mood disorders. Although most of the studies on SPX involved the mouse model, the solution structure of mouse SPX, structural aspects for SPX binding with its receptors GalR2/3, and its cellular expression/distribution in mouse tissues are largely unknown. Using CD and NMR spectroscopies, the solution structure of mouse SPX was shown to be in the form of a helical peptide with a random coil from Asn1 to Pro4 in the N-terminal followed by an α-helix from Gln5 to Gln14 in the C-terminus. The molecular surface of mouse SPX is largely hydrophobic with Lys11 as the only charged residue in the α-helix. Based on the NMR structure obtained, docking models of SPX binding with mouse GalR2 and GalR3 were constructed by homology modeling and MD simulation. The models deduced reveal that the amino acids in SPX, especially Asn1, Leu8, and Leu10, could interact with specific residues in ECL1&2 and TMD2&7 of GalR2 and GalR3 by H-bonding/hydrophobic interactions, which provides the structural evidence to support the idea that the two receptors can act as the cognate receptors for SPX. For tissue distribution of SPX, RT-PCR based on 28 tissues/organs harvested from the mouse demonstrated that SPX was ubiquitously expressed at the tissue level with notable signals detected in the brain, GI tract, liver, gonad, and adrenal gland. Using immunohistochemical staining, protein signals of SPX could be located in the liver, pancreas, white adipose tissue, muscle, stomach, kidney, spleen, gonad, adrenal, and hypothalamo-pituitary axis in a cell type-specific manner. Our results, as a whole, not only can provide the structural information for ligand/receptor interaction for SPX but also establish the anatomical basis for our on-going studies to examine the physiological functions of SPX in the mouse model.


Assuntos
Hormônios Peptídicos/metabolismo , Receptor Tipo 2 de Galanina/metabolismo , Receptor Tipo 3 de Galanina/metabolismo , Animais , Espectroscopia de Ressonância Magnética , Camundongos , Simulação de Acoplamento Molecular
14.
Front Endocrinol (Lausanne) ; 12: 681647, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34276562

RESUMO

Spexin (SPX) is a pleiotropic peptide with highly conserved protein sequence from fish to mammals and its biological actions are mediated by GalR2/GalR3 receptors expressed in target tissues. Recently, SPX has been confirmed to be a novel satiety factor in fish species but whether the peptide has a similar function in mammals is still unclear. Using the mouse as a model, the functional role of SPX in feeding control and the mechanisms involved were investigated. After food intake, serum SPX in mice could be up-regulated with elevations of transcript expression and tissue content of SPX in the glandular stomach but not in other tissues examined. As revealed by immunohistochemical staining, food intake also intensified SPX signals in the major cell types forming the gastric glands (including the foveolar cells, parietal cells, and chief cells) within the gastric mucosa of glandular stomach. Furthermore, IP injection of SPX was effective in reducing food intake with parallel attenuation in transcript expression of NPY, AgRP, NPY type 5 receptor (NPY5R), and ghrelin receptor (GHSR) in the hypothalamus, and these inhibitory effects could be blocked by GalR3 but not GalR2 antagonism. In agreement with the central actions of SPX, similar inhibition on feeding and hypothalamic expression of NPY, AgRP, NPY5R, and GHSR could also be noted with ICV injection of SPX. In the same study, in contrast to the drop in NPY5R and GHSR, SPX treatment could induce parallel rises of transcript expression of leptin receptor (LepR) and melanocortin 4 receptor (MC4R) in the hypothalamus. These findings, as a whole, suggest that the role of SPX as a satiety factor is well conserved in the mouse. Apparently, food intake can induce SPX production in glandular stomach and contribute to the postprandial rise of SPX in circulation. Through GalR3 activation, this SPX signal can act within the hypothalamus to trigger feedback inhibition on feeding by differential modulation of feeding regulators (NPY and AgRP) and their receptors (NPY5R, GHSR, LepR, and MC4R) involved in the feeding circuitry within the CNS.


Assuntos
Ingestão de Alimentos/fisiologia , Hipotálamo/metabolismo , Hormônios Peptídicos/metabolismo , Saciação/fisiologia , Animais , Camundongos , Receptores de Grelina/metabolismo , Regulação para Cima
15.
Front Endocrinol (Lausanne) ; 12: 681648, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34025589

RESUMO

Spexin (SPX), a neuropeptide with diverse functions, is a novel satiety factor in fish models and its role in feeding control has been recently confirmed in mammals. In mouse, food intake was shown to trigger SPX expression in glandular stomach with parallel rise in serum SPX and these SPX signals could inhibit feeding via central actions within the hypothalamus. However, the mechanisms for SPX regulation by food intake are still unclear. To examine the role of insulin signal caused by glucose uptake in SPX regulation, the mice were IP injected with glucose and insulin, respectively. In this case, serum SPX was elevated by glucose but not altered by insulin. Meanwhile, SPX transcript expression in the glandular stomach was up-regulated by glucose but the opposite was true for insulin treatment. Using in situ hybridization, the differential effects on SPX gene expression were located in the gastric mucosa of glandular stomach. Co-injection experiments also revealed that glucose stimulation on serum SPX and SPX mRNA expressed in glandular stomach could be blocked by insulin. In gastric mucosal cells prepared from glandular stomach, the opposite effects on SPX transcript expression by glucose and insulin could still be noted with similar blockade of the stimulatory effects of glucose by insulin. In this cell model, SPX gene expression induced by glucose was mediated by glucose uptake via GLUT, ATP synthesis by glycolysis/respiratory chain, and subsequent modulation of KATP channel activity, but the voltage-sensitive Ca2+ channels were not involved. The corresponding inhibition by insulin, however, was mediated by PI3K/Akt, MEK1/2/ERK1/2, and P38MAPK cascades coupled to insulin receptor but not IGF-1 receptor. Apparently, glucose uptake in mice can induce SPX expression in the glandular stomach through ATP synthesis via glucose metabolism and subsequent modification of KATP channel activity, which may contribute to SPX release into circulation to act as the satiety signal after food intake. The insulin rise caused by glucose uptake, presumably originated from the pancreas, may serve as a negative feedback to inhibit the SPX response by activating MAPK and PI3K/Akt pathways in the stomach.


Assuntos
Glucose/metabolismo , Insulina/metabolismo , Hormônios Peptídicos/metabolismo , Estômago/metabolismo , Animais , Células Cultivadas , Ingestão de Alimentos , Expressão Gênica , Masculino , Camundongos Endogâmicos C57BL , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Hormônios Peptídicos/sangue , Hormônios Peptídicos/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Estômago/citologia
16.
Gen Comp Endocrinol ; 165(1): 60-71, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19501591

RESUMO

Kisspeptin, the product of Kiss1 gene, is a novel regulator of the gonadotropic axis. In mammals, its stimulatory effect on gonadotropin secretion is well documented and mediated mainly by hypothalamic release of gonadotropin-releasing hormone. Although the pituitary actions of kisspeptin have been reported, the effects of kisspeptin on gonadotropin release via direct action on pituitary cells are still controversial. Using goldfish as a model, here we examined the direct actions of kisspeptin on pituitary functions in modern-day bony fish. As a first step, the structural identity of goldfish Kiss1 was established by 5'/3'RACE and Kiss1 transcript was shown to be widely expressed in various tissues in goldfish. At the pituitary level, Kiss1 receptor (Kiss1r) expression was detected in immuno-identified gonadotrophs, lactotrophs, and somatotrophs. Kiss1 transcript was also located in goldfish somatotrophs but not in lactotrophs or gonadotrophs. In parallel studies, goldfish kisspeptin-10 was synthesized and used to test the pituitary actions of kisspeptin in vitro. In goldfish pituitary cell cultures, 30-min incubation with kisspeptin-10 increased basal release of luteinizing hormone (LH), prolactin (PRL), and growth hormone (GH). Transcript expression of LH, PRL, and GH were also elevated by prolonging kisspeptin-10 treatment to 24h. These results taken together suggest that kisspeptin via Kiss1r activation can act directly at the pituitary level to trigger LH, PRL, and GH secretion and gene expression in goldfish. Our finding of Kiss1 expression in somatotrophs also rises the possibility that kisspeptin may be produced locally in the fish pituitary and serve as an autocrine/paracrine regulator.


Assuntos
Proteínas de Peixes , Regulação da Expressão Gênica , Carpa Dourada , Oligopeptídeos , Hipófise/efeitos dos fármacos , Hipófise/metabolismo , Animais , Células Cultivadas , Clonagem Molecular , Proteínas de Peixes/genética , Proteínas de Peixes/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Carpa Dourada/genética , Carpa Dourada/metabolismo , Hormônio do Crescimento/metabolismo , Kisspeptinas , Hormônio Luteinizante/metabolismo , Oligopeptídeos/genética , Oligopeptídeos/metabolismo , Oligopeptídeos/farmacologia , Hipófise/citologia , Prolactina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
Artigo em Inglês | MEDLINE | ID: mdl-32082258

RESUMO

In mammals, local production of tumor necrosis factor α (TNFα) inhibits growth hormone (GH)-induced IGF-I expression at tissue level and contributes to GH resistance caused by sepsis/endotoxemia and inflammation. Although the loss of GH responsiveness can be mediated by a parallel rise in SOCS expression, the signaling mechanisms for TNFα-induced SOCS expression at the hepatic level have not been characterized and the comparative aspects of the phenomenon, especially in lower vertebrates, are still unknown. Recently, type II SOCS, including SOCS1-3 and CISH, have been cloned in grass carp and shown to act as the feedback repressors for GH signaling via JAK2/STAT5 pathway. To shed light on the mechanisms for TNFα-induced GH resistance in fish model, grass carp TNFα was cloned and confirmed to be a single-copy gene expressed in various tissues including the liver. In carp hepatocytes, incubation with the endotoxin LPS induced TNFα expression with parallel rises in SOCS1-3 and CISH mRNA levels. Similar to LPS, TNFα treatment could block GH-induced IGF-I/-II mRNA expression and elevate SOCS1, SOCS3, and CISH transcript levels. However, TNFα was not effective in altering SOCS2 expression. In parallel experiment, LPS blockade of IGF-I/-II signals caused by GH could be partially reverted by TNFα receptor antagonism. At hepatocyte level, TNFα induction also triggered rapid phosphorylation of IκBα, MEK1/2, ERK1/2, MKK3/6, P38MAPK, Akt, JAK2, and STAT1,3,5, and TNFα-induced SOCS1, SOCS3, and CISH mRNA expression could be negated by inhibiting the IKK/NFκB, MAPK, PI3K/Akt, and JAK/STAT cascades. Our findings, as a whole, suggest that local production of TNFα may interfere with IGF-I/-II induction by GH in the carp liver by up-regulation of SOCS1, SOCS3, and CISH via IKK/NFκB, MAPK, PI3K/Akt, and JAK/STAT-dependent mechanisms, which may contribute to GH resistance induced by endotoxin in carp species.


Assuntos
Resistência a Medicamentos , Hormônio do Crescimento/farmacologia , Hepatócitos/efeitos dos fármacos , Proteínas Supressoras da Sinalização de Citocina/genética , Fator de Necrose Tumoral alfa/farmacologia , Animais , Carpas/genética , Carpas/crescimento & desenvolvimento , Carpas/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Resistência a Medicamentos/efeitos dos fármacos , Resistência a Medicamentos/genética , Endotoxinas/toxicidade , Feminino , Proteínas de Peixes/genética , Proteínas de Peixes/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Transtornos do Crescimento/induzido quimicamente , Transtornos do Crescimento/genética , Transtornos do Crescimento/metabolismo , Hepatócitos/metabolismo , Lipopolissacarídeos/toxicidade , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
18.
Biochem Biophys Res Commun ; 390(3): 827-33, 2009 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-19853581

RESUMO

A fish calmodulin (CaM) gene was characterized for the first time in grass carp. The CaM gene is about 12-Kb in size with identical intron/exon organization as that of mammalian CaM genes. When compared to mammalian counterparts, the 5'-promoter region of grass carp CaM gene contains a TATA box and has a much lower GC content and CpG dinucleotide frequency. Interestingly, the 5'-promoter of carp CaM gene is AT-rich with multiple IRS elements and putative binding sites for Pit-1, Sp1/Sp3 and AP1. Using luciferase reporter assay, a potent silencer region was identified in the distal region of grass carp CaM promoter. Besides, the CaM promoter activity could be upregulated by IGF but suppressed by PACAP, forskolin and over-expression of Sp1 and Sp3. These findings, taken together, indicate that grass carp CaM gene does not exhibit the typical features of housekeeping genes and its expression is under the control of hormone factors, presumably by coupling with the appropriate signaling pathways/transcription factors.


Assuntos
Calmodulina/genética , Carpas/genética , Regulação da Expressão Gênica , Transcrição Gênica , Região 5'-Flanqueadora , Animais , Sequência de Bases , Colforsina/farmacologia , Dados de Sequência Molecular , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Sítio de Iniciação de Transcrição
19.
Artigo em Inglês | MEDLINE | ID: mdl-31551932

RESUMO

Glucagon, a key hormone for glucose homeostasis, can exert functional crosstalk with somatotropic axis via modification of IGF-I expression. However, its effect on IGF-I regulation is highly variable in different studies and the mechanisms involved are largely unknown. Using grass carp as a model, the signal transduction and transcriptional mechanisms for IGF-I regulation by glucagon were examined in Cyprinid species. As a first step, the carp HNF1α, a liver-enriched transcription factor, was cloned and confirmed to be a single-copy gene expressed in the liver. In grass carp hepatocytes, glucagon treatment could elevate IGF-I, HNF1α, and CREB mRNA levels, induce CREB phosphorylation, and up-regulate HNF1α and CREB protein expression. The effects on IGF-I, HNF1α, and CREB gene expression were mediated by cAMP/PKA and PLC/IP3/PKC pathways with differential coupling with the MAPK and PI3K/Akt cascades. During the process, protein:protein interaction between HNF1α and CREB and recruitment of RNA Pol-II to IGF-I promoter also occurred with a rise in IGF-I primary transcript level. In parallel study to examine grass carp IGF-I promoter activity expressed in αT3 cells, similar pathways for post-receptor signaling were also confirmed in glucagon-induced IGF-I promoter activation and the trans-activating effect by glucagon was mediated by the binding sites for HNF1α and CREB located in the proximal region of IGF-I promoter. Our findings, as a whole, shed light on a previously undescribed mechanism for glucagon-induced IGF-I gene expression by increasing HNF1α and CREB production via functional crosstalk of post-receptor signaling. Probably, by protein:protein interaction between the two transcription factors and subsequent transactivation via their respective cis-acting elements in the IGF-I promoter, IGF-I gene transcription can be initiated by glucagon at the hepatic level.

20.
Artigo em Inglês | MEDLINE | ID: mdl-30899246

RESUMO

In fish models, seasonal change in feeding is under the influence of water temperature. However, the effects of temperature on appetite control can vary among fish species and the mechanisms involved have not been fully characterized. Using goldfish (Carassius auratus) as a model, seasonal changes in feeding behavior and food intake were examined in cyprinid species. In our study, foraging activity and food consumption in goldfish were found to be reduced with positive correlation to the gradual drop in water temperature occurring during the transition from summer (28.4 ± 2.2°C) to winter (15.1 ± 2.6°C). In goldfish with a 4-week acclimation at 28°C, their foraging activity and food consumption were notably higher than their counterparts with similar acclimation at 15°C. When compared to the group at 28°C during summer, the attenuation in feeding responses at 15°C during the winter also occurred with parallel rises of leptin I and II mRNA levels in the liver. Meanwhile, a drop in orexin mRNA along with concurrent elevations of CCK, MCH, POMC, CART, and leptin receptor (LepR) transcript expression could be noted in brain areas involved in feeding control. In short-term study, goldfish acclimated at 28°C were exposed to 15°C for 24 h and the treatment was effective in reducing foraging activity and food intake. The opposite was true in reciprocal experiment with a rise in water temperature to 28°C for goldfish acclimated at 15°C. In parallel time-course study with lowering of water temperature from 28 to 15°C, short-term exposure (6-12 h) of goldfish to 15°C could also increase leptin I and II mRNA levels in the liver. Similar to our seasonality study, transcript level of orexin was reduced along with up-regulation of CCK, MCH, POMC, CART, and LepR gene expression in different brain areas. Our results, as a whole, suggest that temperature-driven regulation of leptin output from the liver in conjunction with parallel modulations of orexigenic/anorexigenic signals and leptin responsiveness in the brain may contribute to the seasonal changes of feeding behavior and food intake observed in goldfish.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA