Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 107
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Biochem Biophys Res Commun ; 526(4): 920-926, 2020 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-32279993

RESUMO

Ovarian cancer G protein-coupled receptor 1 (OGR1), also known as GPR68, is a proton-sensing G protein-coupled receptor (GPCR) coupling to Gq/11/phospholipase C/Ca2+ signaling pathways. The specific histidine residues at the extracellular surface of OGR1 are suggested to be involved in the proton sensing. Later, some metal ions, including nickel ion (Ni2+), are also indicated to be OGR1 ligands. OGR1 polymorphic variants have recently been found in three families with amelogenesis imperfecta, which suggested that OGR1 is required for the process of dental enamel formation. One of these families possesses a missense mutation from leucine to proline at 74 (L74P) of OGR1. In the present study, we characterized HEK293 cells with L74P OGR1 (L74P-OGR1) and hemagglutinin (HA)-tag, as compared with cells with wild-type OGR1 (WT-OGR1) and HA-tag. We found that either acidic pH or NiCl2 induced intracellular Ca2+ mobilization and morphological change in WT-OGR1-transfected cells; however, the extracellular stimulus-induced actions were severely damaged in L74P-OGR1-transfected cells. We further confirmed that either WT-OGR1 or L74P-OGR1 is localized mainly in the surface of the cells, but only WT-OGR1 is internalized in response to acidification or NiCl2. Thus, the L74P-OGR1 protein may be distributed in the plasma membranes but severely damaged in the receptor functions. We speculate that L74P in the second transmembrane domain in OGR1 may result in conformational changes in the receptor, thereby disturbing the sensing extracellular signals, i.e., protons or metal ions, and/or transducing them to the intracellular signaling machinery through G proteins.


Assuntos
Amelogênese Imperfeita/genética , Amelogênese Imperfeita/metabolismo , Mutação de Sentido Incorreto/genética , Receptores Acoplados a Proteínas G/genética , Sinalização do Cálcio , Forma Celular/efeitos dos fármacos , Endocitose/efeitos dos fármacos , Células HEK293 , Humanos , Concentração de Íons de Hidrogênio , Lisofosfolipídeos/farmacologia , Níquel/toxicidade , Estrutura Secundária de Proteína , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo
2.
Zoolog Sci ; 37(2): 103-108, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32282140

RESUMO

Ogerin is a positive allosteric modulator of human and mouse ovarian cancer G protein-coupled receptors (OGR1s). In the present study, we found that ogerin differentially enhances the activation of OGR1 in various animal species. Amino acid residues of OGR1 that are associated with ogerin are conserved among the species. This suggests that other amino acid residues may be involved in the action of ogerin. Chimeric receptors between human and zebrafish OGR1s showed that the amino acid residues that determine the species specificity of ogerin-induced enhancement reside in the transmembrane and/or intracellular regions of OGR1. This result highlights the importance of first verifying the effectiveness of ogerin to the OGR1 of the species of interest at the cellular level prior to analyzing the physiological and pathophysiological roles of OGR1 in the species.


Assuntos
Álcoois Benzílicos/farmacologia , Prótons , Receptores Acoplados a Proteínas G/genética , Triazinas/farmacologia , Animais , Galinhas , Feminino , Células HEK293 , Humanos , Concentração de Íons de Hidrogênio , Manganês/administração & dosagem , Camundongos , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Síndrome Respiratória e Reprodutiva Suína , Ratos , Receptores Acoplados a Proteínas G/metabolismo , Análise de Sequência de Proteína , Suínos , Xenopus , Peixe-Zebra
3.
J Reprod Dev ; 66(2): 175-180, 2020 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-31956173

RESUMO

Hormone-secreting pituitary adenomas show unregulated hormonal hypersecretion and cause hyperpituitarism. However, the mechanism of the unregulated hormone production and secretion has not yet been fully elucidated. Solid tumors show reduced extracellular pH, partly due to lactate secretion from anaerobic glycolysis. It is known that extracellular acidification affects hormone secretion. However, whether and how the extracellular acidification influences the unregulated hormone production and secretion remain unknown. In the present study, we found that GPR4, a proton-sensing G protein-coupled receptor, was highly expressed in MtT/S cells, a growth hormone-producing and prolactin-producing pituitary tumor cell line. When we reduced the extracellular pH, growth hormone and prolactin mRNA expressions increased in the cells. Both increased expressions were partially suppressed by a GPR4 antagonist. We also found that extracellular acidification enhanced growth hormone-releasing factor-induced growth hormone secretion from MtT/S cells. These results suggest that GPR4 may play a role in hypersecretion of the hormone from hormone-producing pituitary tumors. A GPR4 antagonist will be a useful tool for preventing the hypersecretion.


Assuntos
Hormônio do Crescimento/metabolismo , Hipófise/metabolismo , Prolactina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Linhagem Celular Tumoral , Hormônio do Crescimento/genética , Concentração de Íons de Hidrogênio , Camundongos , Prolactina/genética , Ratos , Receptores Acoplados a Proteínas G/genética
4.
Am J Hum Genet ; 99(4): 984-990, 2016 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-27693231

RESUMO

Amelogenesis is the process of dental enamel formation, leading to the deposition of the hardest tissue in the human body. This process requires the intricate regulation of ion transport and controlled changes to the pH of the developing enamel matrix. The means by which the enamel organ regulates pH during amelogenesis is largely unknown. We identified rare homozygous variants in GPR68 in three families with amelogenesis imperfecta, a genetically and phenotypically heterogeneous group of inherited conditions associated with abnormal enamel formation. Each of these homozygous variants (a large in-frame deletion, a frameshift deletion, and a missense variant) were predicted to result in loss of function. GPR68 encodes a proton-sensing G-protein-coupled receptor with sensitivity in the pH range that occurs in the developing enamel matrix during amelogenesis. Immunohistochemistry of rat mandibles confirmed localization of GPR68 in the enamel organ at all stages of amelogenesis. Our data identify a role for GPR68 as a proton sensor that is required for proper enamel formation.


Assuntos
Amelogênese Imperfeita/genética , Mutação , Receptores Acoplados a Proteínas G/genética , Amelogênese/genética , Animais , Sequência de Bases , Esmalte Dentário/crescimento & desenvolvimento , Esmalte Dentário/patologia , Feminino , Homozigoto , Humanos , Concentração de Íons de Hidrogênio , Masculino , Linhagem , Ratos , Receptores Acoplados a Proteínas G/análise
5.
Biochem Biophys Res Commun ; 517(4): 636-641, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31400852

RESUMO

Extracellular acidification regulates endocrine cell functions. Ovarian cancer G protein-coupled receptor 1 (OGR1), also known as GPR68, is a proton-sensing G protein-coupled receptor and is activated by extracellular acidification, resulting in the activation of multiple intracellular signaling pathways. In the present study, we found that OGR1 was expressed in some gonadotropic cells in rat anterior pituitary and in LßΤ2 cells, which are used as a model of gonadotropic cells. When we reduced extracellular pH, a transient intracellular Ca2+ increase was detected in LßT2 cells. The Ca2+ increase was inhibited by a Gq/11 inhibitor and Cu2+, which is known as an OGR1 antagonist. We also found that extracellular acidification enhanced GnRH-induced Gaussia luciferase secretion from LßT2 cells. These results suggest that OGR1 may play a role in the regulation of gonadotropic cell function such as its hormone secretion.


Assuntos
Ácidos/metabolismo , Cálcio/metabolismo , Espaço Extracelular/metabolismo , Espaço Intracelular/metabolismo , Animais , Células Cultivadas , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Humanos , Luciferases/metabolismo , Hormônio Luteinizante/metabolismo , Adeno-Hipófise/citologia , Ratos , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/metabolismo , Fatores de Tempo
6.
Zoolog Sci ; 36(2): 105-111, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-31120644

RESUMO

Mammalian T cell death-associated gene 8 (TDAG8)s are activated by extracellular protons. In the present study, we examined whether the TDAG8 homologs of other species are activated by protons as they are in mammals. We found that Xenopus TDAG8 also stimulated cAMP response element (CRE)-driven promoter activities reflecting the activation of Gs/cAMP signaling pathways when they are stimulated by protons. On the other hand, the activities of chicken and zebrafish TDAG8s are hardly affected by protons. Results using chimeric receptors of human and zebrafish TDAG8s indicate that the specificity of the proton-induced activation lies in the extracellular region. These results suggest that protons are not an evolutionarily conserved agonist of TDAG8.


Assuntos
Prótons , Receptores Acoplados a Proteínas G/genética , Animais , Galinhas , AMP Cíclico/metabolismo , Células HEK293 , Humanos , Concentração de Íons de Hidrogênio , Receptores Acoplados a Proteínas G/metabolismo , Xenopus , Peixe-Zebra
7.
Zoolog Sci ; 35(2): 109-114, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29623784

RESUMO

Human, mouse, and zebrafish ovarian cancer G protein-coupled receptors (OGR1s) are activated by both metals and extracellular protons. In the present study, we examined whether pig, rat, chicken, and Xenopus OGR1 homologs could sense and be activated by protons and metals. We found that all homologs stimulated serum response element (SRE)-driven promoter activities when they are stimulated by protons. On the other hand, metals differentially activated the homologs. The results using chimeric receptors of human and zebrafish OGR1s indicate that the specificity of the metal-induced activation lies in the extracellular region. These results suggest that protons are an evolutionally conserved agonist of OGR1. However, the types of metals that activated the receptor differed among the homologs.


Assuntos
Galinhas/genética , Metais/administração & dosagem , Prótons , Ratos/genética , Receptores Acoplados a Proteínas G/genética , Sus scrofa/genética , Xenopus/genética , Animais , Galinhas/metabolismo , Feminino , Células HEK293 , Humanos , Neoplasias Ovarianas/genética , Ratos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Elemento de Resposta Sérica/efeitos dos fármacos , Sus scrofa/metabolismo , Xenopus/metabolismo
8.
Am J Physiol Lung Cell Mol Physiol ; 312(6): L835-L844, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28314803

RESUMO

A G protein-coupled receptor (GPCR) named free fatty acid receptor 4 (FFA4, also known as GPR120) was found to act as a GPCR for ω-3 polyunsaturated fatty acids. Its expression has been reported in lung epithelial club cells. We investigated whether supplementation of the ω-3 fatty acids benefits lung health. Omacor (7.75 mg/kg), clinically prescribed preparation of ω-3 fatty acids, and FFA4-knockout mice were utilized in a naphthalene-induced mouse model of acute airway injury (1 injection of 30 mg/kg ip). Naphthalene injection induced complete destruction of bronchiolar epithelial cells within a day. Appearance of bronchiolar epithelial cells was observed after 21 days in control mice. It was found, however, that supplementation of Omacor accelerated the recovery. The appearance of bronchiolar epithelial cells was observed between 7 and 14 days after naphthalene injury in Omacor-treated mice. In isolated club cells, ω-3 fatty acids were found to stimulate cell proliferation and migration but to inhibit cell differentiation. With the use of pharmacological tools and FFA4-knockout mice, FFA4 was found to be responsible for ω-3 fatty acids-induced proliferation in vitro in club cells. Furthermore, accelerated recovery from naphthalene-induced airway injury in Omacor-treated mice was not observed in FFA4-knockout mice in vivo. Present findings indicate that ω-3 fatty acids-induced proliferation of bronchiole epithelial cells through FFA4 is responsible for Omacor-induced accelerated recovery from airway injury. Therefore, intermittent administration of Omacor needs to be tested for acute airway injury because ω-3 fatty acids stimulate proliferation but inhibit differentiation of club cells.


Assuntos
Ácidos Graxos Ômega-3/farmacologia , Pulmão/patologia , Receptores Acoplados a Proteínas G/metabolismo , Cicatrização/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Separação Celular , Ácidos Docosa-Hexaenoicos/farmacologia , Combinação de Medicamentos , Ácido Eicosapentaenoico/farmacologia , Lesão Pulmonar/metabolismo , Lesão Pulmonar/patologia , Masculino , Camundongos Endogâmicos BALB C , Camundongos Knockout , Naftalenos
9.
J Recept Signal Transduct Res ; 37(4): 401-408, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28270026

RESUMO

Mammalian ovarian G-protein-coupled receptor 1 (OGR1) is activated by some metals in addition to extracellular protons and coupling to multiple intracellular signaling pathways. In the present study, we examined whether zebrafish OGR1, zebrafish GPR4, and human GPR4 (zOGR1, zGPR4, and hGPR4, respectively) could sense the metals and activate the intracellular signaling pathways. On one hand, we found that only manganese and cobalt of the tested metals stimulated SRE-promoter activities in zOGR1-overexpressed HEK293T cells. On the other hand, none of the metals tested stimulated the promoter activities in zGPR4- and hGPR4-overexpressed cells. The OGR1 mutant (H4F), which is lost to activation by extracellular protons, did not stimulate metal-induced SRE-promoter activities. These results suggest that zOGR1, but not GPR4, is also a metal-sensing G-protein-coupled receptor in addition to a proton-sensing G-protein-coupled receptor, although not all metals that activate hOGR1 activated zOGR1.


Assuntos
Receptores Acoplados a Proteínas G/genética , Proteínas de Peixe-Zebra/genética , Animais , Cobalto/farmacologia , AMP Cíclico , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Manganês/farmacologia , Regiões Promotoras Genéticas/genética , Prótons , Transdução de Sinais/efeitos dos fármacos , Peixe-Zebra/genética
10.
Biochem Biophys Res Commun ; 469(1): 81-86, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26614909

RESUMO

Human G2A is activated by various stimuli such as lysophosphatidylcholine (LPC), 9-hydroxyoctadecadienoic acid (9-HODE), and protons. The receptor is coupled to multiple intracellular signaling pathways, including the Gs-protein/cAMP/CRE, G12/13-protein/Rho/SRE, and Gq-protein/phospholipase C/NFAT pathways. In the present study, we examined whether zebrafish G2A homologs (zG2A-a and zG2A-b) could respond to these stimuli and activate multiple intracellular signaling pathways. We also examined whether histidine residue and basic amino acid residue in the N-terminus of the homologs also play roles similar to those played by human G2A residues if the homologs sense protons. We found that the zG2A-a showed the high CRE, SRE, and NFAT activities, however, zG2A-b showed only the high SRE activity under a pH of 8.0. Extracellular acidification from pH 7.4 to 6.3 ameliorated these activities in zG2A-a-expressing cells. On the other hand, acidification ameliorated the SRE activity but not the CRE and NFAT activities in zG2A-b-expressing cells. LPC or 9-HODE did not modify any activity of either homolog. The substitution of histidine residue at the 174(th) position from the N-terminus of zG2A-a to asparagine residue attenuated proton-induced CRE and NFAT activities but not SRE activity. The substitution of arginine residue at the 32nd position from the N-terminus of zG2A-a to the alanine residue also attenuated its high and the proton-induced CRE and NFAT activities. On the contrary, the substitution did not attenuate SRE activity. The substitution of the arginine residue at the 10th position from the N-terminus of zG2A-b to the alanine residue also did not attenuate its high or the proton-induced SRE activity. These results indicate that zebrafish G2A homologs were activated by protons but not by LPC and 9-HODE, and the activation mechanisms of the homologs were similar to those of human G2A.


Assuntos
Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Concentração de Íons de Hidrogênio , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/fisiologia , Peixe-Zebra/metabolismo , Sequência de Aminoácidos , Animais , Células HEK293 , Humanos , Líquido Intracelular/química , Líquido Intracelular/metabolismo , Dados de Sequência Molecular , Ligação Proteica , Relação Estrutura-Atividade
11.
Biochem Biophys Res Commun ; 457(4): 493-9, 2015 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-25576873

RESUMO

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.


Assuntos
Prótons , Receptores Acoplados a Proteínas G/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Sequência de Aminoácidos , Animais , Regulação da Expressão Gênica , Células HEK293 , Humanos , Concentração de Íons de Hidrogênio , Dados de Sequência Molecular , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Alinhamento de Sequência , Transdução de Sinais , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/química , Proteínas de Peixe-Zebra/genética
12.
Biochem Biophys Res Commun ; 464(3): 949-55, 2015 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-26196745

RESUMO

Although blood pH is maintained in a narrow range of around pH 7.4 in living organisms, inflammatory loci are characterized by acidic conditions. Mast cells tend to reside close to the surface of the body in areas such as the mucosa and skin where they may be exposed to exogenous acids, and they play an important role in immune responses. However, little is known about the effects of extracellular acidification on the functions of mast cell. Here, we found that extracellular acidification increased the dinitrophenyl-conjugated human serum albumin (DNP-HSA)-induced production of interleukin (IL)-6 and IL-13 in MC/9 cells or bone marrow-derived mouse mast cells sensitized with anti-DNP IgE. Extracellular acidification also inhibited migration of MC/9 cells toward DNP-HSA. In addition, acidic pH stimulated antigen-induced activation of p38 mitogen-activated protein kinase (MAPK) and protein kinase B (Akt). These findings suggest that extracellular acidification augmented antigen/IgE-induced and FcεRI-mediated production of IL-6 and IL-13 in mast cells, and that this was associated with the enhancement of p38 MAPK and Akt activation.


Assuntos
Interleucina-13/metabolismo , Interleucina-6/metabolismo , Mastócitos/metabolismo , Receptores de IgE/metabolismo , Animais , Movimento Celular , Células Cultivadas , Dinitrofenóis/farmacologia , Concentração de Íons de Hidrogênio , Imunoglobulina E/imunologia , Imunoglobulina E/metabolismo , Mastócitos/química , Mastócitos/efeitos dos fármacos , Mastócitos/imunologia , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Albumina Sérica/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
13.
Acta Pharmacol Sin ; 36(7): 813-20, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26005911

RESUMO

AIM: Free fatty acid receptor 4 (FFA4; formerly known as GPR120) is the G protein-coupled receptor (GPCR) for omega-3 polyunsaturated fatty acids. FFA4 has been found to express in the small intestines and colons of mice and humans. In this study we investigate the effects of omega-3 polyunsaturated fatty acids on FFA4 in human colon epithelial cells in vitro. METHODS: HCT116 and HT-29 human colon epithelial cell lines endogenously expressing FFA4 were used. Intracellular Ca(2+) concentration ([Ca(2+)]i) was measured in fura 2-AM-loaded cells with fluorescence spectrophotometry. RT-PCR and immunohistochemistry were used to detect FFA4. RESULTS: Ten to 100 µmol/L of omega-3 polyunsaturated fatty acids α-linolenic acid (αLA) or eicosapentaenoic acid (EPA) induced dose-dependent [Ca(2+)]i increase in HCT116 and HT-29 cells, whereas docosahexaenoic acid (DHA) had no effect. In addition, the omega-6 fatty acids linoleic acid and γ-linoleic acid also dose-dependently increase [Ca(2+)]i, but the mono-unsaturated fatty acid oleic acid and saturated fatty acids such as stearic acid and palmitic acid had no effect. In HCT116 and HT-29 cells, the αLA-induced [Ca(2+)]i increase was partially inhibited by pretreatment with EGTA, phospholipase C inhibitor edelfosine, cADPR inhibitors 8-bro-cADPR or DAB, and abolished by pretreatment with Ca(2+)ATPase inhibitor thapsigargin, but was not affected by Gi/o protein inhibitor PTX or IP3R inhibitor 2-APB. CONCLUSION: Omega-3 and omega-6 long-chain polyunsaturated fatty acids (C18-20) induce Ca(2+) mobilization responses in human colonic epithelial cells in vitro through activation of FFA4 and PTX-insensitive Gi/o protein, followed by Ca(2+) release from thapsigargin-sensitive Ca(2+) stores and Ca(2+) influx across the plasma membrane.


Assuntos
Cálcio/metabolismo , Colo/metabolismo , Células Epiteliais/metabolismo , Ácidos Graxos Ômega-3/farmacologia , Receptores Acoplados a Proteínas G/biossíntese , Colo/citologia , Colo/efeitos dos fármacos , Relação Dose-Resposta a Droga , Células Epiteliais/efeitos dos fármacos , Regulação da Expressão Gênica , Células HCT116 , Células HT29 , Humanos
14.
Int J Mol Sci ; 16(12): 28931-42, 2015 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-26690120

RESUMO

Acute lung injury is characterized by the infiltration of neutrophils into lungs and the subsequent impairment of lung function. Here we explored the role of TDAG8 in lung injury induced by lipopolysaccharide (LPS) administrated intratracheally. In this model, cytokines and chemokines released from resident macrophages are shown to cause neutrophilic inflammation in the lungs. We found that LPS treatment increased TDAG8 expression in the lungs and confirmed its expression in resident macrophages in bronchoalveolar lavage (BAL) fluids. LPS administration remarkably increased neutrophil accumulation without appreciable change in the resident macrophages, which was associated with increased penetration of blood proteins into BAL fluids, interstitial accumulation of inflammatory cells, and damage of the alveolar architecture. The LPS-induced neutrophil accumulation and the associated lung damage were enhanced in TDAG8-deficient mice as compared with those in wild-type mice. LPS also increased several mRNA and protein expressions of inflammatory cytokines and chemokines in the lungs or BAL fluids. Among these inflammatory mediators, mRNA and protein expression of KC (also known as CXCL1), a chemokine of neutrophils, were significantly enhanced by TDAG8 deficiency. We conclude that TDAG8 is a negative regulator for lung neutrophilic inflammation and injury, in part, through the inhibition of chemokine production.


Assuntos
Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/patologia , Pulmão/patologia , Lesão Pulmonar Aguda/genética , Animais , Quimiocinas/imunologia , Feminino , Deleção de Genes , Lipopolissacarídeos/imunologia , Pulmão/imunologia , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/imunologia , Neutrófilos/patologia , Prótons , RNA Mensageiro/genética , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/imunologia , Regulação para Cima
15.
J Neurochem ; 129(4): 683-95, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24447140

RESUMO

Interleukin-1ß (IL-1ß) is released from activated microglia and involved in the neurodegeneration of acute and chronic brain disorders, such as stroke and Alzheimer's disease, in which extracellular acidification has been shown to occur. Here, we examined the extracellular acidic pH regulation of IL-1ß production, especially focusing on TDAG8, a major proton-sensing G-protein-coupled receptor, in mouse microglia. Extracellular acidification inhibited lipopolysaccharide -induced IL-1ß production, which was associated with the inhibition of IL-1ß cytoplasmic precursor and mRNA expression. The IL-1ß mRNA and protein responses were significantly, though not completely, attenuated in microglia derived from TDAG8-deficient mice compared with those from wild-type mice. The acidic pH also stimulated cellular cAMP accumulation, which was completely inhibited by TDAG8 deficiency. Forskolin and a cAMP derivative, which specifically stimulates protein kinase A (PKA), mimicked the proton actions, and PKA inhibitors reversed the acidic pH-induced IL-1ß mRNA expression. The acidic pH-induced inhibitory IL-1ß responses were accompanied by the inhibition of extracellular signal-related kinase and c-Jun N-terminal kinase activities. The inhibitory enzyme activities in response to acidic pH were reversed by the PKA inhibitor and TDAG8 deficiency. We conclude that extracellular acidic pH inhibits lipopolysaccharide-induced IL-1ß production, at least partly, through the TDAG8/cAMP/PKA pathway, by inhibiting extracellular signal-related kinase and c-Jun N-terminal kinase activities, in mouse microglia.


Assuntos
AMP Cíclico/fisiologia , Líquido Extracelular/química , Regulação da Expressão Gênica/efeitos dos fármacos , Interleucina-1beta/biossíntese , Microglia/metabolismo , Sistemas do Segundo Mensageiro/fisiologia , 1-Metil-3-Isobutilxantina/farmacologia , Animais , Células Cultivadas , Colforsina/farmacologia , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Concentração de Íons de Hidrogênio , Proteínas I-kappa B/metabolismo , Interleucina-1beta/genética , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases JNK Ativadas por Mitógeno/fisiologia , Lipopolissacarídeos/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Inibidor de NF-kappaB alfa , NF-kappa B/metabolismo , Poli I-C/farmacologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/deficiência , Receptores Acoplados a Proteínas G/fisiologia , Sistemas do Segundo Mensageiro/efeitos dos fármacos
16.
Endocr J ; 61(2): 101-10, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24088601

RESUMO

Insulin secretion with respect to pH environments has been investigated for a long time but its mechanism remains largely unknown. Extracellular pH is usually maintained at around 7.4 and, its change has been thought to occur in non-physiological situations. Acidification takes place under ischemic and inflammatory microenvironments, where stimulation of anaerobic glycolysis results in the production of lactic acid. In addition to ionotropic ion channels, such as transient receptor potential V1 (TRPV1) and acid-sensing ion channels (ASICs), metabotropic proton-sensing G protein-coupled receptors (GPCRs) have also been identified recently as proton-sensing machineries. While ionotropic ion channels usually sense strong acidic pH, proton-sensing GPCRs sense pH of 7.6 to 6.0 and have been shown to mediate a variety of biological actions in neutral and mildly acidic pH environments. Studies with receptor knockout mice have revealed that proton-sensing receptors, including ovarian cancer G protein-coupled receptor 1 (OGR1), a proton-sensing GPCRs, play a role in the regulation of insulin secretion and glucose metabolism under physiological conditions. Small molecule 3,5-disubstituted isoxazoles have recently been identified as OGR1 agonists working at neutral pH and have been shown to stimulate pancreatic ß-cell differentiation and insulin synthesis. Thus, proton-sensing OGR1 may be an important player for insulin secretion and a potential target for improving ß-cell function.


Assuntos
Concentração de Íons de Hidrogênio , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Receptores Acoplados a Proteínas G/fisiologia , Animais , Diferenciação Celular/efeitos dos fármacos , Feminino , Glucose/metabolismo , Insulina/biossíntese , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Isoxazóis/farmacologia , Camundongos , Ratos , Receptores Acoplados a Proteínas G/agonistas , Canais de Cátion TRPV/fisiologia
17.
Mediators Inflamm ; 2014: 712962, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25197168

RESUMO

An acidic microenvironment has been shown to evoke a variety of airway responses, including cough, bronchoconstriction, airway hyperresponsiveness (AHR), infiltration of inflammatory cells in the lung, and stimulation of mucus hyperproduction. Except for the participation of transient receptor potential vanilloid-1 (TRPV1) and acid-sensing ion channels (ASICs) in severe acidic pH (of less than 6.0)-induced cough and bronchoconstriction through sensory neurons, the molecular mechanisms underlying extracellular acidic pH-induced actions in the airways have not been fully understood. Recent studies have revealed that ovarian cancer G protein-coupled receptor 1 (OGR1)-family G protein-coupled receptors, which sense pH of more than 6.0, are expressed in structural cells, such as airway smooth muscle cells and epithelial cells, and in inflammatory and immune cells, such as eosinophils and dendritic cells. They function in a variety of airway responses related to the pathophysiology of inflammatory diseases, including allergic asthma. In the present review, we discuss the roles of ionotropic TRPV1 and ASICs and metabotropic OGR1-family G protein-coupled receptors in the airway inflammation and AHR in asthma and respiratory diseases.


Assuntos
Asma/imunologia , Asma/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Canais Iônicos Sensíveis a Ácido/metabolismo , Feminino , Humanos , Masculino , Receptores Acoplados a Proteínas G/metabolismo , Canais de Cátion TRPV/metabolismo
18.
Biochem Biophys Res Commun ; 439(3): 357-62, 2013 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-23998930

RESUMO

Cysteinyl leukotrienes (cysLTs), which include leukotriene C4 (LTC4), are the predominant class of LTs synthesized by mast cells. CysLTs can induce many of the abnormalities seen in asthma. LTC4 is generated by the conjugation of LTA4 with reduced glutathione (GSH) by LTC4 synthase. During screening of the effects of prostanoids on high-affinity IgE receptor (FcεRI)-mediated LTC4 release from mast cells, we realized that some prostanoids, including ONO-AE1-259-01 and ONO-AE-248, inhibited LTC4 release, which was associated with a decrease in the amount of intracellular total GSH. We ascertained that l-buthionine-S,R-sulfoximine (BSO), a selective inhibitor of glutamate-cysteine ligase, inhibited LTC4 release. In addition, cell-permeable GSH, the glutathione reduced form ethyl ester (GSH-OEt), enhanced LTC4 release in accordance with the change in intracellular total GSH. Depletion of intracellular total GSH induced by ONO-AE-248 or BSO enhanced FcεRI-mediated LTB4 release in contrast to LTC4. Oxidative stress contributes to many pathological conditions including asthma. GSH is a major soluble antioxidant and a cofactor for several detoxifying enzymes including GSH peroxidase. Exposure of mast cells to hydrogen peroxide (H2O2) or diamide to mimic oxidative stress unexpectedly increased rather than decreased the intracellular reduced GSH content as well as total GSH in the late phase (i.e., 24 or 48 h after exposure), which was accompanied by an increase in LTC4 release. In conclusion, FcεRI-mediated LTC4 release from mast cells is mainly regulated by the amount of intracellular GSH. In some cases, oxidative stress may induce a late-phase increase in intracellular GSH, resulting in enhanced LTC4 release from mast cells.


Assuntos
Glutationa/metabolismo , Leucotrieno C4/metabolismo , Mastócitos/metabolismo , Estresse Oxidativo , Receptores de IgE/metabolismo , Animais , Basófilos/imunologia , Basófilos/metabolismo , Linhagem Celular , Células Cultivadas , Glutationa/imunologia , Humanos , Peróxido de Hidrogênio/imunologia , Peróxido de Hidrogênio/metabolismo , Leucotrieno C4/imunologia , Mastócitos/imunologia , Camundongos , Prostaglandinas/imunologia , Prostaglandinas/metabolismo , Receptores de IgE/imunologia
19.
Cancer Sci ; 103(6): 1099-104, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22348348

RESUMO

Pancreatic cancer is highly metastatic and has a poor prognosis. However, there is no established treatment for pancreatic cancer. Lysophosphatidic acid (LPA) has been shown to be present in effluents of cancers and involved in migration and proliferation in a variety of cancer cells, including pancreatic cancer cells, in vitro. In the current study, we examined whether an orally active LPA antagonist is effective for pancreatic cancer tumorigenesis and metastasis in vivo. Oral administration of Ki16198, which is effective for LPA(1) and LPA(3), into YAPC-PD pancreatic cancer cell-inoculated nude mice significantly inhibited tumor weight and remarkably attenuated invasion and metastasis to lung, liver, and brain, in association with inhibition of matrix metalloproteinase (MMP) accumulation in ascites in vivo. Ki16198 inhibited LPA-induced migration and invasion in several pancreatic cancer cells in vitro, which was associated with the inhibition of LPA-induced MMP production. In conclusion, Ki16198 is a promising orally active LPA antagonist for inhibiting the invasion and metastasis of pancreatic cancer cells. The inhibitory effects of the antagonist on invasion and metastasis in vivo may be partially explained by the inhibition of motility activity and MMP production in cancer cells.


Assuntos
Isoxazóis/farmacologia , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Propionatos/farmacologia , Receptores de Ácidos Lisofosfatídicos/antagonistas & inibidores , Animais , Ascite/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Humanos , Isoxazóis/administração & dosagem , Isoxazóis/uso terapêutico , Lisofosfolipídeos/farmacologia , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Inibidores de Metaloproteinases de Matriz , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica , Metástase Neoplásica/tratamento farmacológico , Neoplasias Pancreáticas/metabolismo , Neoplasias Peritoneais/patologia , Neoplasias Peritoneais/prevenção & controle , Neoplasias Peritoneais/secundário , Propionatos/administração & dosagem , Propionatos/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
20.
J Immunol ; 185(8): 4863-72, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20861350

RESUMO

Lysophosphatidic acid (LPA) is a phospholipid mediator that exerts a variety of biological responses through specific G-protein-coupled receptors (LPA(1)-LPA(5) and P2Y5). LPA is thought to be involved in airway inflammation by regulating the expression of anti-inflammatory and proinflammatory genes. Chemokines such as CCL5/RANTES are secreted from airway epithelium and play a key role in allergic airway inflammation. CCL5/RANTES is a chemoattractant for eosinophils, T lymphocytes, and monocytes and seems to exacerbate asthma. We stimulated CCL5/RANTES production in a human bronchial epithelial cell line, BEAS-2B, with IFN-γ and TNF-α. When LPA was added, CCL5/RANTES mRNA expression and protein secretion were inhibited, despite the presence of IFN-γ and TNF-α. The LPA effect was attenuated by Ki16425, a LPA(1)/LPA(3) antagonist, but not by dioctylglycerol pyrophosphate 8:0, an LPA(3) antagonist. Pertussis toxin, the inhibitors for PI3K and Akt also attenuated the inhibitory effect of LPA on CCL5/RANTES secretion. We also identify the transcription factor IFN regulatory factor-1 (IRF-1) as being essential for CCL5/RANTES production. Interestingly, LPA inhibited IFN-γ and TNF-α-induced IRF-1 activation by blocking the binding of IRF-1 to its DNA consensus sequence without changing IRF-1 induction and its nuclear translocation. Ki16425, pertussis toxin, and PI3K inhibitors attenuated the inhibitory effect of LPA on IRF-1 activation. Our results suggest that LPA inhibits IFN-γ- and TNF-α-induced CCL5/RANTES production in BEAS-2B cells by blocking the binding of IRF-1 to the CCL5/RANTES promoter. LPA(1) coupled to G(i) and activation of PI3K is required for this unique effect.


Assuntos
Quimiocina CCL5/metabolismo , Regulação da Expressão Gênica/imunologia , Fator Regulador 1 de Interferon/metabolismo , Lisofosfolipídeos/metabolismo , Mucosa Respiratória/metabolismo , Western Blotting , Brônquios/imunologia , Brônquios/metabolismo , Linhagem Celular , Quimiocina CCL5/imunologia , Ensaio de Desvio de Mobilidade Eletroforética , Ensaio de Imunoadsorção Enzimática , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Expressão Gênica , Humanos , Fator Regulador 1 de Interferon/imunologia , Lisofosfolipídeos/imunologia , Regiões Promotoras Genéticas , Mucosa Respiratória/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/imunologia , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA