Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros











Intervalo de año de publicación
1.
J Reprod Dev ; 70(5): 343-347, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39135241

RESUMEN

The adenohypophysis is composed of the anterior and intermediate lobes (AL and IL, respectively), and secretes hormones that play an important role in reproduction. CD9- and SOX2-double (CD9/SOX2) positive cells located in the marginal cell layer (MCL) facing the Rathke's cleft in the AL and IL form the primary stem cell niche in the adult adenohypophysis of rats. In this study, we successfully obtained 3-dimensional (3D) cell aggregates that closely resembled the primary niche of MCL in vivo. After incubation in a Matrigel containing several growth factors, approximately 20% of the cells in the CD9/SOX2-positive cell aggregates were differentiated into hormone-producing cells. The cell aggregates generated in this study may provide insight into the regulation of the pituitary stem/progenitor cell niche and the turnover of hormone-producing cells.


Asunto(s)
Diferenciación Celular , Factores de Transcripción SOXB1 , Nicho de Células Madre , Tetraspanina 29 , Animales , Tetraspanina 29/metabolismo , Ratas , Factores de Transcripción SOXB1/metabolismo , Adenohipófisis/metabolismo , Adenohipófisis/citología , Masculino , Técnicas de Cultivo Tridimensional de Células/métodos , Células Cultivadas , Proteoglicanos/metabolismo , Laminina/metabolismo , Colágeno , Combinación de Medicamentos
2.
Histochem Cell Biol ; 155(4): 503-512, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33398434

RESUMEN

Hypertension leads to structural remodeling of cerebral blood vessels, which has been implicated in the pathophysiology of cerebrovascular diseases. The remodeling and progression of arteriolosclerosis under hypertension involve fibrosis along with the production of type I collagen around cerebral arterioles. However, the source and regulatory mechanisms of this collagen production remain elusive. In this study, we examined if perivascular macrophages (PVMs) are involved in collagen production around cerebral small vessels in hypertensive SHRSP/Izm rats. Immunoreactivity for type I collagen around cerebral small vessels in 12-week-old hypertensive rats tended to higher than those in 4-week-old hypertensive and 12-week-old control rats. In ultrastructural analyses using transmission electron microscopy, the substantial deposition of collagen fibers could be observed in the intercellular spaces around PVMs near the arterioles of rats with prolonged hypertension. In situ hybridization analyses revealed that cells positive for mRNA of Col1a1, which comprises type I collagen, were observed near cerebral small vessels. The Col1a1-positive cells around cerebral small vessels were colocalized with immunoreactivity for CD206, a marker for PVMs, but not with those for glial fibrillary acidic protein or desmin, markers for other perivascular cells such as astrocytes and vascular smooth muscle cells. These results demonstrated that enhanced production of type I collagen is observed around cerebral small vessels in rats with prolonged hypertension and Col1a1 is expressed by PVMs, and support the concept that PVMs are involved in collagen production and vascular fibrosis under hypertensive conditions.


Asunto(s)
Arterias Cerebrales/metabolismo , Colágeno Tipo I/biosíntesis , Hipertensión/metabolismo , Macrófagos/metabolismo , Animales , Modelos Animales de Enfermedad , Masculino , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY
3.
Acta Histochem Cytochem ; 53(3): 43-53, 2020 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-32624629

RESUMEN

Leukemias are refractory hematopoietic malignancies, for which the development of new therapeutic agents requires in vivo studies using tumor-bearing mouse models. Although several organs are commonly examined in such studies to evaluate the disease course, the effectiveness of interventions and the localization of tumor cells in the affected organs are still unclear. In this study, we histologically examined the distribution of leukemia cells in several organs using two leukemic mouse models produced by the administration of two cell lines (THP-1, a human myelomonocytic leukemia, and A20, a mouse B cell leukemia/lymphoma) to severe immunodeficient mice. Survival of the mice depended on the tumor burden. Although A20 and THP-1 tumor cells massively infiltrated the parenchyma of the liver and spleen at 21 days after transplantation, A20 cells were hardly found in connective tissues in Glisson's capsule in the liver as compared with THP-1 cells. In the bone marrow, there was more severe infiltration of A20 cells than THP-1 cells. THP-1 and A20 cells were widely spread in the lungs, but were rarely observed in the small intestine. These findings suggest that each leukemia model has a unique localization of tumor cells in several affected organs, which could critically affect the disease course and the efficacy of therapeutic agents, including cellular immunotherapies.

4.
Acta Histochem Cytochem ; 51(5): 145-152, 2018 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-30510328

RESUMEN

Laminin, a major basement membrane protein, comprises three subunit chains: α, ß, and γ chains. Among these chains, only the laminin α chain is capable of signaling via laminin receptors. Although laminin isoforms containing the α5 chain were reported to be the first laminin produced during rat anterior pituitary gland development, the functions of these isoforms are unknown. We used immunohistochemical techniques to localize the laminin α5 chain and its specific receptor, basal cell adhesion molecule (BCAM), in fetal and adult pituitary gland. Laminin α5 chain immunoreactivity was observed in the basement membrane of the primordial adenohypophysis at embryonic days 12.5 to 19.5. Double immunostaining showed that BCAM was present and co-localized with the laminin α5 chain in the tissue. Quantitative analysis showed that the laminin α5 chain and BCAM were expressed in the anterior pituitary gland during postnatal development and in adulthood (postnatal day 60). In the adult gland, co-localization of the laminin α5 chain and BCAM was observed, and BCAM was detected in both the folliculo-stellate cells and endothelial cells. These results suggest that laminin α5 chain signaling via BCAM occurs in both the fetal adenohypophysis and adult anterior pituitary gland.

5.
Acta Histochem Cytochem ; 50(2): 63-69, 2017 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-28522881

RESUMEN

After publication of reports describing the presence of stem/progenitor cells among non-hormone-producing cells in the pituitary, the mechanism responsible for proliferation and differentiation generated considerable interest. Several studies have suggested that Notch signaling is involved. In the present study, we examined the histochemical relationship between Notch signaling molecules and the transcription factor SOX2 in rat pituitary. Combined in situ hybridization and immunohistochemistry showed that Notch2 mRNA and SOX2 were co-expressed at embryonic day 14.5 in most cells in the adenohypophyseal primordium. In adult rat pituitary, double immunohistochemistry showed that SOX2 and either Notch2 or the Notch signaling target HES1 were co-localized within cells with large oval nuclei in both the marginal cell layer and cell aggregates in the main part of the anterior lobe, which are believed to be stem cell niches. Furthermore, when the Notch signaling inhibitor DAPT was added to a primary culture of adult rat anterior pituitary cells, the proportion of SOX2-expressing cells within Notch2-positive cells was approximately 30% lower. These findings suggest that Notch signaling has a role in maintaining the stemness of precursor cells in the adult rat pituitary gland.

6.
Endocr J ; 64(6): 633-638, 2017 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-28392548

RESUMEN

Retinoic acid (RA) is converted from retinal by retinaldehyde dehydrogenases (RALDHs) and is an essential signaling molecule in embryonic and adult tissue. We previously reported that RALDH1 was produced in the rat anterior pituitary gland and hypothesized that RA was generated in the gland. Midkine (MK) is an RA-inducible growth factor, and MK production in the rat anterior pituitary gland was recently reported. However, the mechanism that regulates gene expression of MK in the pituitary gland has not been determined. To investigate regulation of MK production in the anterior pituitary gland, we analyzed changes in MK mRNA in cultured rat anterior pituitary cells. We identified MK-expressing cells by double-staining with in situ hybridization and immunohistochemical techniques for RALDH1. MK mRNA was expressed in RALDH1-producing cells in the anterior pituitary gland. Using isolated anterior pituitary cells of rats, we examined the effect of RA on gene expression of MK. Quantitative real-time PCR revealed that 72 h exposure to a concentration of 10-6 M of retinal and all-trans retinoic acid increased MK mRNA levels by about 2-fold. Moreover, the stimulatory effect of all-trans retinoic acid was mimicked by the RA receptor agonist Am80. This is the first report to show that RA is important in regulating MK expression in rat anterior pituitary gland.


Asunto(s)
Regulación de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/agonistas , Factores de Crecimiento Nervioso/agonistas , Adenohipófisis/metabolismo , Retinal-Deshidrogenasa/metabolismo , Tretinoina/metabolismo , Regulación hacia Arriba , 2-Naftilamina/análogos & derivados , 2-Naftilamina/farmacología , Familia de Aldehído Deshidrogenasa 1 , Animales , Benzoatos/farmacología , Células Cultivadas , Regulación de la Expresión Génica/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Cinética , Masculino , Midkina , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Adenohipófisis/citología , Adenohipófisis/efectos de los fármacos , Pirimidinas/farmacología , ARN Mensajero/metabolismo , Ratas Wistar , Receptores de Ácido Retinoico/agonistas , Receptores de Ácido Retinoico/metabolismo , Retinal-Deshidrogenasa/genética , Retinaldehído/metabolismo , Transducción de Señal/efectos de los fármacos , Tetrahidronaftalenos/farmacología , Regulación hacia Arriba/efectos de los fármacos
7.
Med Mol Morphol ; 50(3): 145-154, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28353090

RESUMEN

Extracellular matrix (ECM) is essential in tissue physiology and pathologic conditions such as tumorigenesis. It affects tumor cell behavior, proliferation, and metastasis. Pituitary adenomas differ in their clinical characteristics, including ECM deposition, and we recently reported that the characteristics of collagen-producing cells differed between control human anterior pituitary gland and pituitary adenomas. ECM deposition is not defined solely by production; degradation and maintenance are also important. Tissue inhibitors of metalloproteinases (TIMPs) help maintain ECM by inhibiting degradation caused by matrix metalloproteases. The present study attempted to characterize TIMP-expressing cells in the human anterior pituitary. Specimens of human pituitary adenomas and control pituitary were obtained during surgery, and in situ hybridization for TIMP1, TIMP2, TIMP3, and TIMP4, followed by immunohistochemistry, was used to characterize TIMP-expressing cells. TIMP expression exhibited a distinct pattern in the human anterior pituitary. Azan staining showed that fibrous matrix deposition varied among pituitary adenomas and that the area of fibrosis was associated with the number and number of types of TIMP3-expressing cells. These results suggest that TIMPs are important in the maintenance of ECM in human pituitary and that TIMP expressions are altered in fibrosis associated with pituitary adenoma.


Asunto(s)
Adenoma/metabolismo , Adenoma/patología , Neoplasias Hipofisarias/metabolismo , Neoplasias Hipofisarias/patología , Adenoma/genética , Matriz Extracelular/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Hipófisis/metabolismo , Hipófisis/patología , Neoplasias Hipofisarias/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Inhibidores Tisulares de Metaloproteinasas/genética , Inhibidores Tisulares de Metaloproteinasas/metabolismo
8.
Cell Tissue Res ; 368(1): 125-133, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27942853

RESUMEN

The rat anterior pituitary is composed of hormone-producing cells, non-hormone-producing cells (referred to as folliculostellate cells) and marginal layer cells. In the adult rat, progenitor cells of hormone-producing cells have recently been reported to be maintained within this non-hormone-producing cell population. In tissue, non-hormone-producing cells construct homophilic cell aggregates by the differential expression of the cell adhesion molecule E-cadherin. We have previously shown that Notch signaling, a known regulator of progenitor cells in a number of organs, is activated in the cell aggregates. We now investigate the relationship between Notch signaling and E-cadherin-mediated cell adhesion in the pituitary gland. Immunohistochemically, Notch signaling receptor Notch2 and the ligand Jagged1 were localized within E-cadherin-positive cells in the marginal cell layer and in the main part of the anterior lobe, whereas Notch1 was localized in E-cadherin-positive and -negative cells. Activation of Notch signaling within E-cadherin-positive cells was confirmed by immunostaining of the Notch target HES1. Notch2 and Jagged1 were always co-localized within the same cells suggesting that homologous cells have reciprocal effects in activating Notch signaling. When the E-cadherin function was inhibited by exposure to a monoclonal antibody (DECMA-1) in primary monolayer cell culture, the percentage of HES1-positive cells among Notch2-positive cells was less than half that of the control. The present results suggest that E-cadherin-mediated cell attachment is necessary for the activation of Notch signaling in the anterior pituitary gland but not for the expression of the Notch2 molecule.


Asunto(s)
Cadherinas/metabolismo , Comunicación Celular , Adenohipófisis/citología , Adenohipófisis/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Animales , Adhesión Celular , Proteína Jagged-1/metabolismo , Ligandos , Masculino , Ratas Wistar , Factor de Transcripción HES-1/metabolismo
9.
Endocr J ; 63(6): 555-61, 2016 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-27052215

RESUMEN

Retinoic acid (RA) is an important signaling molecule in embryonic development and adult tissue. The actions of RA are mediated by the nuclear receptors retinoic acid receptor (RAR) and retinoid X receptor (RXR), which regulate gene expression. RAR and RXR are widely expressed in the anterior pituitary gland. RA was reported to stimulate growth hormone (GH) gene expression in the anterior pituitary cells. However, current evidence is unclear on the role of RA in gene expression of growth hormone-releasing hormone receptor (Ghrh-r), growth hormone secretagogue receptor (Ghs-r) and somatostatin receptors (Sst-rs). Using isolated anterior pituitary cells of rats, we examined the effects of RA on gene expression of these receptors and GH release. Quantitative real-time PCR revealed that treatment with all-trans retinoic acid (ATRA; 10(-6) M) for 24 h increased gene expression levels of Ghrh-r and Ghs-r; however, expressions of Sst-r2 and Sst-r5 were unchanged. Combination treatment with the RAR-agonist Am80 and RXR-agonist PA024 mimicked the effects of ATRA on Ghrh-r and Ghs-r gene expressions. Exposure of isolated pituitary cells to ATRA had no effect on basal GH release. In contrast, ATRA increased growth hormone-releasing hormone (GHRH)- and ghrelin-stimulated GH release from cultured anterior pituitary cells. Our results suggest that expressions of Ghrh-r and Ghs-r are regulated by RA through the RAR-RXR receptor complex and that RA enhances the effects of GHRH and ghrelin on GH release from the anterior pituitary gland.


Asunto(s)
Hormona del Crecimiento/metabolismo , Adenohipófisis/efectos de los fármacos , Adenohipófisis/metabolismo , Receptores de Ghrelina/genética , Receptores de Neuropéptido/genética , Receptores de Hormona Reguladora de Hormona Hipofisaria/genética , Tretinoina/farmacología , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Ratas , Ratas Wistar , Factores de Tiempo
10.
J Endocrinol ; 229(2): 159-70, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26957638

RESUMEN

The anterior pituitary gland comprises five types of endocrine cells plus non-endocrine cells including folliculostellate cells, endothelial cells, and capillary mural cells (pericytes). In addition to being controlled by the hypothalamic-pituitary-target organ axis, the functions of these cells are likely regulated by local cell and extracellular matrix (ECM) interactions. However, these complex interactions are not fully understood. We investigated folliculostellate cell-mediated cell-to-cell interaction. Using S100ß-GFP transgenic rats, which express GFP in folliculostellate cells, we designed a three-dimensional cell culture to examine the effects of folliculostellate cells. Interestingly, removal of folliculostellate cells reduced collagen synthesis (Col1a1 and Col3a1). Because pericytes are important collagen-producing cells in the gland, we stained for desmin (a pericyte marker). Removal of folliculostellate cells resulted in fewer desmin-positive pericytes and less desmin mRNA. We then attempted to identify the factor mediating folliculostellate cell-pericyte interaction. RT-PCR and in situ hybridization revealed that the important profibrotic factor transforming growth factor beta-2 (TGFß2) was specifically expressed in folliculostellate cells and that TGFß receptor II was expressed in pericytes, endothelial cells, and parenchymal cells. Immunocytochemistry showed that TGFß2 induced SMAD2 nuclear translocation in pericytes. TGFß2 increased collagen synthesis in a dose-dependent manner. This action was completely blocked by TGFß receptor I inhibitor (SB431542). Diminished collagen synthesis in folliculostellate cell-deficient cell aggregates was partially recovered by TGFß2. TGFß2-mediated folliculostellate cell-pericyte interaction appears to be essential for collagen synthesis in rat anterior pituitary. This finding sheds new light on local cell-ECM interactions in the gland.


Asunto(s)
Pericitos/metabolismo , Adenohipófisis/citología , Adenohipófisis/metabolismo , Factor de Crecimiento Transformador beta2/metabolismo , Transporte Activo de Núcleo Celular , Animales , Benzamidas/farmacología , Agregación Celular , Comunicación Celular , Células Cultivadas , Colágeno/biosíntesis , Dioxoles/farmacología , Masculino , Pericitos/citología , Adenohipófisis/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Ratas , Ratas Transgénicas , Ratas Wistar , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta2/genética
11.
Biochem Biophys Res Commun ; 457(4): 493-9, 2015 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-25576873

RESUMEN

Mammalian ovarian G-protein-coupled receptor 1 (OGR1) and GPR4 are identified as a proton-sensing G-protein-coupled receptor coupling to multiple intracellular signaling pathways. In the present study, we examined whether zebra fish OGR1 and GPR4 homologs (zOGR1 and zGPR4) could sense protons and activate the multiple intracellular signaling pathways and, if so, whether the similar positions of histidine residue, which is critical for sensing protons in mammalian OGR and GPR4, also play a role to sense protons and activate the multiple signaling pathways in the zebra fish receptors. We found that extracellular acidic pH stimulated CRE-, SRE-, and NFAT-promoter activities in zOGR1 overexpressed cells and stimulated CRE- and SRE- but not NFAT-promoter activities in zGPR4 overexpressed cells. The substitution of histidine residues at the 12th, 15th, 162th, and 264th positions from the N-terminal of zOGR1 with phenylalanine attenuated the proton-induced SRE-promoter activities. The mutation of the histidine residue at the 78th but not the 84th position from the N-terminal of zGPR4 to phenylalanine attenuated the proton-induced SRE-promoter activities. These results suggest that zOGR1 and zGPR4 are also proton-sensing G-protein-coupled receptors, and the receptor activation mechanisms may be similar to those of the mammalian receptors.


Asunto(s)
Protones , Receptores Acoplados a Proteínas G/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Secuencia de Aminoácidos , Animales , Regulación de la Expresión Génica , Células HEK293 , Humanos , Concentración de Iones de Hidrógeno , Datos de Secuencia Molecular , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Alineación de Secuencia , Transducción de Señal , Pez Cebra/genética , Proteínas de Pez Cebra/química , Proteínas de Pez Cebra/genética
12.
Cell Tissue Res ; 357(1): 337-44, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24816986

RESUMEN

Pituitary gland development is controlled by numerous signaling molecules, which are produced in the oral ectoderm and diencephalon. A newly described family of heparin-binding growth factors, namely midkine (MK)/pleiotrophin (PTN), is involved in regulating the growth and differentiation of many tissues and organs. Using in situ hybridization with digoxigenin-labeled cRNA probes, we detected cells expressing MK and PTN in the developing rat pituitary gland. At embryonic day 12.5 (E12.5), MK expression was localized in Rathke's pouch (derived from the oral ectoderm) and in the neurohypophyseal bud (derived from the diencephalon). From E12.5 to E19.5, MK mRNA was expressed in the developing neurohypophysis, and expression gradually decreased in the developing adenohypophysis. To characterize MK-expressing cells, we performed double-staining of MK mRNA and anterior pituitary hormones. At E19.5, no MK-expressing cells were stained with any hormone. In contrast, PTN was expressed only in the neurohypophysis primordium during all embryonic stages. In situ hybridization clearly showed that MK was expressed in primitive (immature/undifferentiated) adenohypophyseal cells and neurohypophyseal cells, whereas PTN was expressed only in neurohypophyseal cells. Thus, MK and PTN might play roles as signaling molecules during pituitary development.


Asunto(s)
Proteínas Portadoras/biosíntesis , Citocinas/biosíntesis , Hipófisis/metabolismo , Animales , Proteínas Portadoras/genética , Citocinas/genética , Femenino , Hibridación in Situ , Midkina , Hipófisis/embriología , Embarazo , Ratas , Ratas Wistar
13.
Obes Res Clin Pract ; 7(1): e1-7, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24331677

RESUMEN

Food intake is a fundamental for animals to surviving and keeping offspring. The hypothalamic region of the brain and the brain stem in vertebrates are a center that plays an important role in the control of feeding and its related behaviors including locomotor and psychomotor activities. Pituitary adenylate cyclase-activating polypeptide (PACAP) has firstly been identified as a hypophysiotropic hormone involved in adenohypophyseal hormone release, and subsequently has been considered as a neuropeptide exerting multifunctional roles in the central and peripheral nervous systems and several tissues in vertebrates. For example, PACAP is involved in the neuroendocrine control of food intake and acts as an anorexigenic peptide to regulate satiety. Recent works on animal models such as rodents and goldfish which are both excellent animal models for investigating the neuroendocrinological roles of PACAP have been extensively examined and considerable information has been accumulated. In addition, psychophysiological effects of PACAP on emotional behavior have recently been found. Therefore, this review article provides an overview of the neuroendocrine regulation of feeding behavior and psyphysiological activity by PACAP in vertebrates.


Asunto(s)
Regulación del Apetito , Encéfalo/metabolismo , Ingestión de Alimentos , Conducta Alimentaria/fisiología , Actividad Motora/fisiología , Sistemas Neurosecretores , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Animales , Emociones/fisiología , Humanos , Saciedad/fisiología , Vertebrados
14.
Peptides ; 43: 40-7, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23422837

RESUMEN

Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide that stimulates the release of adenohypophyseal hormone from the pituitary in fish. In the goldfish, PACAP induces the release of somatolactin (SL), in particular, from cultured pituitary cells. SL belongs to the growth hormone and prolactin family, and comprises two molecular variants termed SL-α and SL-ß in goldfish. However, there is no information about the involvement of PACAP in the regulation of SL-α and SL-ß release and the expression of their mRNAs. Therefore, we examined the effect of PACAP on SL-α and SL-ß release from cultured goldfish pituitary cells. Treatment with PACAP (10(-10)-10(-7)M) increased the release of both SL-α and SL-ß. The stimulatory action of PACAP (10(-9)M) on SL-α and SL-ß release was blocked by treatment with a PACAP-selective receptor (PAC1R) antagonist, PACAP(6-38) (10(-6)M). We also examined whether PACAP affects the expression of SL-α and SL-ß mRNAs in cultured pituitary cells. Treatment with PACAP (10(-9) and 10(-8)M) for 6h decreased the expression level of SL-α mRNA but increased that of SL-ß mRNA. The action of PACAP (10(-8)M) on SL-ß mRNA expression was blocked by treatment with PACAP(6-38) (10(-6)M), whereas PACAP(6-38) elicited no change in the expression of SL-α mRNA. These results indicate that in cultured goldfish pituitary cells, PACAP stimulates the release of SL-α and SL-ß, and expression of SL-ß mRNA, via the PAC1R-signaling pathway. However, the mechanism whereby PACAP inhibits the expression of SL-α mRNA does not seem to be mediated by PAC1R signaling.


Asunto(s)
Proteínas de Peces/metabolismo , Glicoproteínas/metabolismo , Carpa Dorada , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Hipófisis/citología , Hormonas Hipofisarias/metabolismo , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/metabolismo , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Animales , Células Cultivadas , Proteínas de Peces/genética , Perfilación de la Expresión Génica , Glicoproteínas/genética , Hipófisis/efectos de los fármacos , Hipófisis/metabolismo , Hormonas Hipofisarias/genética , ARN Mensajero/genética
15.
Neuropeptides ; 46(6): 275-83, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23122775

RESUMEN

Neuropeptide Y (NPY) is a neuropeptide distributed widely among vertebrates. In mammals, NPY and its related peptides such as pancreatic polypeptide and peptide YY (PYY) are distributed throughout the brain and gastrointestinal tissues, and are centrally involved in many physiological functions such as the regulation of food intake, locomotion and psychomotor activities through their receptors. With regard to non-mammalian vertebrates, there has also been intensive study aimed at the identification and functional characterization of NPY, PYY and their receptors, and recent investigations of the role of NPY have revealed that it exerts several behavioral effects in goldfish and zebrafish. Both of these species are excellent teleost fish models, in which it has been demonstrated that NPY increases food consumption as an orexigenic factor and reduces locomotor activity, as is the case in mammals. This paper reviews current knowledge of NPY derived from studies of teleost fish, as representative non-mammals, focusing particularly on the role of the NPY system, and examines its significance from a comparative viewpoint.


Asunto(s)
Conducta Alimentaria/fisiología , Peces/fisiología , Actividad Motora/fisiología , Neuropéptido Y/fisiología , Sistemas Neurosecretores/fisiología , Animales , Polipéptido Pancreático/fisiología , Receptores de Neuropéptido Y/fisiología
16.
Cell Tissue Res ; 350(1): 167-76, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22729486

RESUMEN

Somatolactin (SL) is a pituitary hormone belonging to the growth hormone/prolactin family of adenohypophyseal hormones. In teleost fish, SL is encoded by one or two paralogous genes, namely SL-α and -ß. Our previous studies have revealed that pituitary adenylate-cyclase-activating polypeptide stimulates SL release from cultured goldfish pituitary cells, whereas melanin-concentrating hormone suppresses this release. As in other fish, the goldfish possesses SL-α and -ß. So far, however, no useful means of detecting the respective SLs immunologically in this species has been possible. In order to achieve this aim, we raised rabbit antisera against synthetic peptide fragments deduced from the goldfish SL-α and -ß cDNA sequences. Using these antisera, we observed adenohypophyseal cells showing SL-α- and -ß-like immunoreactivities in the goldfish pituitary, especially the pars intermedia (PI). Several cells in the PI showed the colocalization of SL-α- and -ß-like immunoreactivities. Then, using single-cell polymerase chain reaction with laser microdissection, we examined SL-α and -ß gene expression in adenohypophyseal cells showing SL-α- or -ß-like immunoreactivity. Among cultured pituitary cells, we observed three types of cell: those that possess transcripts of SL-α, -ß, or both. These results suggest a polymorphism of SL-producing cells in the goldfish pituitary.


Asunto(s)
Proteínas de Peces/metabolismo , Glicoproteínas/metabolismo , Carpa Dorada/metabolismo , Hipófisis/citología , Hipófisis/metabolismo , Hormonas Hipofisarias/metabolismo , Secuencia de Aminoácidos , Animales , Especificidad de Anticuerpos/inmunología , Proteínas de Peces/química , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Regulación de la Expresión Génica , Glicoproteínas/química , Glicoproteínas/genética , Glicoproteínas/inmunología , Hormona del Crecimiento , Sueros Inmunes/inmunología , Inmunohistoquímica , Rayos Láser , Microdisección , Datos de Secuencia Molecular , Péptidos/química , Péptidos/inmunología , Adenohipófisis/citología , Adenohipófisis/metabolismo , Hormonas Hipofisarias/química , Hormonas Hipofisarias/genética , Hormonas Hipofisarias/inmunología , Prolactina , ARN Mensajero/genética , ARN Mensajero/metabolismo , Alineación de Secuencia
17.
Peptides ; 32(1): 31-5, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20955748

RESUMEN

Intracerebroventricular (ICV) administration of gonadotropin-releasing hormone II (GnRH II), which plays a crucial role in the regulation of reproduction in vertebrates, markedly reduces food intake in goldfish. However, the neurochemical pathways involved in the anorexigenic action of GnRH II and its interaction with other neuropeptides have not yet been identified. Alpha-melanocyte-stimulating hormone (α-MSH), corticotropin-releasing hormone (CRH) and CRH-related peptides play a major role in feeding control as potent anorexigenic neuropeptides in goldfish. However, our previous study has indicated that the GnRH II-induced anorexigenic action is not blocked by treatment with melanocortin 4 receptor (MC4R) and CRH receptor antagonists. Therefore, in the present study, we further examined whether the anorexigenic effects of α-MSH and CRH in goldfish could be mediated through the GnRH receptor neuronal pathway. ICV injection of the MC4R agonist, melanotan II (80 pmol/g body weight; BW), significantly reduced food intake, and its anorexigenic effect was suppressed by ICV pre-administration of the GnRH type I receptor antagonist, antide (100 pmol/gBW). The CRH-induced (50 pmol/gBW) anorexigenic action was also blocked by treatment with antide. ICV injection of CRH (50 pmol/gBW) induced a significant increase of the GnRH II mRNA level in the hypothalamus, while ICV injection of melanotan II (80 pmol/gBW) had no effect on the level of GnRH II mRNA. These results indicate that, in goldfish, the anorexigenic actions of α-MSH and CRH are mediated through the GnRH type I receptor-signaling pathway, and that the GnRH II system regulates feeding behavior.


Asunto(s)
Depresores del Apetito/farmacología , Hormona Liberadora de Corticotropina/farmacología , Hormona Liberadora de Gonadotropina/metabolismo , Péptidos Cíclicos/farmacología , alfa-MSH/análogos & derivados , Animales , Carpa Dorada/metabolismo , Hormona Liberadora de Gonadotropina/genética , Hipotálamo/metabolismo , ARN Mensajero/metabolismo , Transducción de Señal , alfa-MSH/farmacología
18.
J Endocrinol ; 203(3): 389-98, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19752150

RESUMEN

Melanin-concentrating hormone (MCH)-containing neurons directly innervate the adenohypophysis in the teleost pituitary. We examined immunohistochemically the relationship between MCH-containing nerve fibres or endings and somatolactin (SL)-producing cells in the goldfish pituitary. Nerve fibres or endings with MCH-like immunoreactivity were identified in the neurohypophysis in close proximity to the adenohypophysial cells showing SL-like immunoreactivity. We also examined the effect of MCH on SL release from cultured goldfish pituitary cells and SL synthesis using a cell immunoblot and a real-time PCR method. Treatment of individually dispersed pituitary cells with MCH 10(-7) M for 3 h decreased the area of SL-like immunoreactivity on immunoblots, and MCH-induced reductions in SL release were blocked by treatment with the mammalian MCH receptor (MCHR) antagonist, compound-30, at a concentration of 10(-5) M. Treatment with 10(-7) M MCH for 3 h did not affect sl-alpha and -beta (smtla and -b as given in the Zfin Database) mRNA expression levels. These led us to explore the signal transduction mechanism leading to the inhibition of SL release, for which we examined whether MCH-induced reductions in SL release are mediated by the G(i) or G(q) protein-coupled signalling pathway. The MCH-induced reductions in SL release were abolished by treatment with the G(i/o) protein inhibitors, NF023 (10(-5) M) or pertussis toxin (260 ng/ml), but not by the phospholipase C inhibitor, U-73122 (3x10(-6) M). These results indicate that MCH can potentially function as a hypothalamic factor suppressing SL release via the MCHR, and subsequently through the G(i) protein to inhibit the adenylate cyclase/cAMP/protein kinase A-signalling pathway in goldfish pituitary cells.


Asunto(s)
Proteínas de Peces/metabolismo , Glicoproteínas/metabolismo , Carpa Dorada/metabolismo , Hormonas Hipotalámicas/metabolismo , Melaninas/metabolismo , Fibras Nerviosas/fisiología , Hipófisis/metabolismo , Hormonas Hipofisarias/metabolismo , Animales , Células Cultivadas , Estrenos , Femenino , Immunoblotting , Masculino , Toxina del Pertussis , Hipófisis/citología , Pirrolidinonas , ARN Mensajero/metabolismo , Receptores de la Hormona Hipofisaria/antagonistas & inhibidores , Suramina/análogos & derivados , Fosfolipasas de Tipo C/antagonistas & inhibidores
19.
Peptides ; 30(7): 1260-6, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19540424

RESUMEN

In the goldfish pituitary, nerve fibers containing pituitary adenylate cyclase-activating polypeptide (PACAP) are located in close proximity to somatolactin (SL)-producing cells, and PACAP enhances SL release from cultured pituitary cells. However, there is little information about the mechanism of PACAP-induced SL release. In order to elucidate this issue, we used the cell immunoblot method. Treatment with PACAP at 10(-8) and 10(-7)M, but not with vasoactive intestinal polypeptide (VIP) at the same concentrations, increased the immunoblot area for SL-like immunoreactivity from dispersed pituitary cells, and PACAP-induced SL release was blocked by treatment with the PACAP selective receptor (PAC(1)R) antagonist, PACAP(6-38), at 10(-6)M, but not with the PACAP/VIP receptor antagonist, VIP(6-28). PACAP-induced SL release was also attenuated by treatment with the calmodulin inhibitor, calmidazolium at 10(-6)M. This led us to explore the signal transduction mechanism up to SL release, and we examined whether PACAP-induced SL release is mediated by the adenylate cyclase (AC)/cAMP/protein kinase A (PKA)- or the phospholipase C (PLC)/inositol 1,4,5-trisphosphate (IP(3))/protein kinase C (PKC)-signaling pathway. PACAP-induced SL release was attenuated by treatment with the AC inhibitor, MDL-12330A, at 10(-5)M or with the PKA inhibitor, H-89, at 10(-5)M. PACAP-induced SL release was suppressed by treatment with the PLC inhibitor, U-73122, at 3 x 10(-6)M or with the PKC inhibitor, GF109203X, at 10(-6)M. These results suggest that PACAP can potentially function as a hypophysiotropic factor mediating SL release via the PAC(1)R and subsequently through perhaps the AC/cAMP/PKA- and the PLC/IP(3)/PKC-signaling pathways in goldfish pituitary cells.


Asunto(s)
Proteínas de Peces/metabolismo , Glicoproteínas/metabolismo , Sustancias de Crecimiento/farmacología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Hipófisis/citología , Hipófisis/metabolismo , Hormonas Hipofisarias/metabolismo , Inhibidores de Adenilato Ciclasa , Adenilil Ciclasas/metabolismo , Animales , Calmodulina/antagonistas & inhibidores , Calmodulina/metabolismo , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Estrenos/farmacología , Femenino , Carpa Dorada , Imidazoles/farmacología , Iminas/farmacología , Immunoblotting , Indoles/farmacología , Péptidos y Proteínas de Señalización Intracelular/farmacología , Isoquinolinas/farmacología , Masculino , Maleimidas/farmacología , Fosfoinositido Fosfolipasa C/antagonistas & inhibidores , Fosfoinositido Fosfolipasa C/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Pirrolidinonas/farmacología , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Sulfonamidas/farmacología , Péptido Intestinal Vasoactivo/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA