Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Biochem Biophys Res Commun ; 647: 72-79, 2023 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-36731336

RESUMO

Apelin (APL), an endogenous ligand for APJ, has been reported to be upregulated in a murine model of acute colitis induced by sodium dextran sulfate, as well as inflammatory bowel diseases (IBD) in humans. However, the mechanisms and functions of APL/APJ axis in the pathogenesis of IBD are unclear. We herein analyzed CD4+ T cells to determine the functions of APL in a murine model of chronic colitis induced in Rag deficient mice (Rag-/-). In colonic tissues of wild-type mice (WT), we found that APL was expressed especially in the lamina propria lymphocytes, where CD4+ T cells are dominant, rather than the epithelial cells. Unexpectedly, the APL expression was rather downregulated in the colonic tissue of the chronic colitis group compared to the control groups (Rag-/- before colitis induction and WT). The APL expression was downregulated when naïve T cells were differentiated into effecter T cells. A lack of APL resulted in decreased naïve T cells and increased effecter T cells in secondary lymphoid organs. A synthetic APL peptide, [Pyr1]-APL-13, increased IL-10 and decreased IFN-γ productions by effecter T cells. Administration of [Pyr1]-APL-13 improved survival rate in association with lessened colitis severity and decreased pro-inflammatory cytokine production. This is the first report showing immunological function of APL specifically on T cells, and these results indicate that APL/APJ axis may be a novel therapeutic target for IBD.


Assuntos
Colite , Doenças Inflamatórias Intestinais , Camundongos , Humanos , Animais , Linfócitos T/metabolismo , Apelina/metabolismo , Modelos Animais de Doenças , Colite/patologia , Doenças Inflamatórias Intestinais/metabolismo , Sulfato de Dextrana , Camundongos Endogâmicos C57BL , Linfócitos T CD4-Positivos
2.
Proc Natl Acad Sci U S A ; 117(6): 3150-3156, 2020 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-31992639

RESUMO

Heart failure and chronic kidney disease are major causes of morbidity and mortality internationally. Although these dysfunctions are common and frequently coexist, the factors involved in their relationship in cardiorenal regulation are still largely unknown, mainly due to a lack of detailed molecular targets. Here, we found the increased plasma histamine in a preclinical mouse model of severe cardiac dysfunction, that had been cotreated with angiotensin II (Ang II), nephrectomy, and salt (ANS). The ANS mice exhibited impaired renal function accompanied with heart failure, and histamine depletion, by the genetic inactivation of histidine decarboxylase in mice, exacerbated the ANS-induced cardiac and renal abnormalities, including the reduction of left ventricular fractional shortening and renal glomerular and tubular injuries. Interestingly, while the pharmacological inhibition of the histamine receptor H3 facilitated heart failure and kidney injury in ANS mice, administration of the H3 agonist immethridine (Imm) was protective against cardiorenal damages. Transcriptome analysis of the kidney and biochemical examinations using blood samples illustrated that the increased inflammation in ANS mice was alleviated by Imm. Our results extend the pharmacological use of H3 agonists beyond the initial purposes of its drug development for neurogenerative diseases and have implications for therapeutic potential of H3 agonists that invoke the anti-inflammatory gene expression programming against cardiorenal damages.


Assuntos
Anti-Inflamatórios/farmacologia , Insuficiência Cardíaca/metabolismo , Agonistas dos Receptores Histamínicos/farmacologia , Histamina/metabolismo , Nefropatias/metabolismo , Animais , Modelos Animais de Doenças , Coração/efeitos dos fármacos , Histamina/sangue , Rim/efeitos dos fármacos , Camundongos , Substâncias Protetoras/farmacologia , Receptores Histamínicos H3/metabolismo
3.
Circulation ; 141(7): 571-588, 2020 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-31665900

RESUMO

BACKGROUND: The maternal circulatory system and hormone balance both change dynamically during pregnancy, delivery, and the postpartum period. Although atrial natriuretic peptides and brain natriuretic peptides produced in the heart control circulatory homeostasis through their common receptor, NPR1, the physiologic and pathophysiologic roles of endogenous atrial natriuretic peptide/brain natriuretic peptide in the perinatal period are not fully understood. METHODS: To clarify the physiologic and pathophysiologic roles of the endogenous atrial natriuretic peptide/brain natriuretic peptide-NPR1 system during the perinatal period, the phenotype of female wild-type and conventional or tissue-specific Npr1-knockout mice during the perinatal period was examined, especially focusing on maternal heart weight, blood pressure, and cardiac function. RESULTS: In wild-type mice, lactation but not pregnancy induced reversible cardiac hypertrophy accompanied by increases in fetal cardiac gene mRNAs and ERK1/2 (extracellular signaling-regulated kinase) phosphorylation. Npr1-knockout mice exhibited significantly higher plasma aldosterone level than did wild-type mice, severe cardiac hypertrophy accompanied by fibrosis, and left ventricular dysfunction in the lactation period. Npr1-knockout mice showed a high mortality rate over consecutive pregnancy-lactation cycles. In the hearts of Npr1-knockout mice during or after the lactation period, an increase in interleukin-6 mRNA expression, phosphorylation of signal transducer and activator of transcription 3, and activation of the calcineurin-nuclear factor of the activated T cells pathway were observed. Pharmacologic inhibition of the mineralocorticoid receptor or neuron-specific deletion of the mineralocorticoid receptor gene significantly ameliorated cardiac hypertrophy in lactating Npr1-knockout mice. Anti-interleukin-6 receptor antibody administration tended to reduce cardiac hypertrophy in lactating Npr1-knockout mice. CONCLUSIONS: These results suggest that the characteristics of lactation-induced cardiac hypertrophy in wild-type mice are different from exercise-induced cardiac hypertrophy, and that the endogenous atrial natriuretic peptide/brain natriuretic peptide-NPR1 system plays an important role in protecting the maternal heart from interleukin-6-induced inflammation and remodeling in the lactation period, a condition mimicking peripartum cardiomyopathy.


Assuntos
Fator Natriurético Atrial/deficiência , Cardiomegalia/metabolismo , Lactação , Sistema de Sinalização das MAP Quinases , Período Periparto , Receptores do Fator Natriurético Atrial/deficiência , Animais , Cardiomegalia/genética , Cardiomegalia/patologia , Feminino , Camundongos , Camundongos Knockout
4.
J Neurochem ; 156(6): 834-847, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33460120

RESUMO

PRMT1, a major arginine methyltransferase, plays critical roles in transcription, DNA damage response, and cell proliferation. Although we have previously discovered the crucial roles of PRMT1 for oligodendrocyte lineage progression in the central nervous system of neural stem cell-specific PRMT1 conditional knockout (PRMT1-CKO) mice, the context of other glial cell states that may cause the hypomyelination phenotype in PRMT1-CKO mice has not been explored so far. Here, we performed RNA-seq of the neonatal cortices of PRMT1-CKO mice to reveal overall gene expression changes and show the up-regulation of inflammatory signaling which is generally mediated by astrocytes and microglia in advance of the myelination defects. In particular, qRT-PCR analyses revealed Interleukin-6 (Il-6), a major central nervous system cytokine, was dramatically increased in the PRMT1-CKO brains. The gene expression changes led to augmentation of glial fibrillary acidic protein and Vimentin protein levels in PRMT1-CKO mice, showing severe reactive astrogliosis after birth. We further show that IBA1-positive and CD68-positive activated microglia were increased in PRMT1-CKO mice, in spite of intact Prmt1 gene expression in purified microglia from the mutant mice. Our results indicate that PRMT1 loss in the neural stem cell lineage causes disruptive changes in all glial types perturbing postnatal brain development and myelination.


Assuntos
Astrócitos , Encéfalo/crescimento & desenvolvimento , Sistema Nervoso Central/metabolismo , Microglia , Proteína-Arginina N-Metiltransferases/genética , Animais , Animais Recém-Nascidos , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Encefalite/fisiopatologia , Feminino , Interleucina-6/metabolismo , Camundongos , Camundongos Knockout , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Mutação , Bainha de Mielina , Células-Tronco Neurais/metabolismo , Gravidez , RNA Interferente Pequeno/farmacologia , Transdução de Sinais
5.
Int J Mol Sci ; 21(18)2020 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-32937976

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a progressive fibrosing lung disease that is caused by the dysregulation of alveolar epithelial type II cells (AEC II). The mechanisms involved in the progression of IPF remain incompletely understood, although the immune response accompanied by p38 mitogen-activated protein kinase (MAPK) activation may contribute to some of them. This study aimed to examine the association of p38 activity in the lungs with bleomycin (BLM)-induced pulmonary fibrosis and its transcriptomic profiling. Accordingly, we evaluated BLM-induced pulmonary fibrosis during an active fibrosis phase in three genotypes of mice carrying stepwise variations in intrinsic p38 activity in the AEC II and performed RNA sequencing of their lungs. Stepwise elevation of p38 signaling in the lungs of the three genotypes was correlated with increased severity of BLM-induced pulmonary fibrosis exhibiting reduced static compliance and higher collagen content. Transcriptome analysis of these lung samples also showed that the enhanced p38 signaling in the lungs was associated with increased transcription of the genes driving the p38 MAPK pathway and differentially expressed genes elicited by BLM, including those related to fibrosis as well as the immune system. Our findings underscore the significance of p38 MAPK in the progression of pulmonary fibrosis.


Assuntos
Fibrose Pulmonar Idiopática/genética , Pulmão/metabolismo , Transcriptoma/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Células Epiteliais Alveolares/metabolismo , Animais , Bleomicina/farmacologia , Colágeno/metabolismo , Feminino , Fibrose Pulmonar Idiopática/induzido quimicamente , Fibrose Pulmonar Idiopática/metabolismo , Sistema de Sinalização das MAP Quinases/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
6.
Proc Natl Acad Sci U S A ; 113(38): 10702-7, 2016 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-27601664

RESUMO

GABA acts as the major inhibitory neurotransmitter in the mammalian brain, shaping neuronal and circuit activity. For sustained synaptic transmission, synaptic vesicles (SVs) are required to be recycled and refilled with neurotransmitters using an H(+) electrochemical gradient. However, neither the mechanism underlying vesicular GABA uptake nor the kinetics of GABA loading in living neurons have been fully elucidated. To characterize the process of GABA uptake into SVs in functional synapses, we monitored luminal pH of GABAergic SVs separately from that of excitatory glutamatergic SVs in cultured hippocampal neurons. By using a pH sensor optimal for the SV lumen, we found that GABAergic SVs exhibited an unexpectedly higher resting pH (∼6.4) than glutamatergic SVs (pH ∼5.8). Moreover, unlike glutamatergic SVs, GABAergic SVs displayed unique pH dynamics after endocytosis that involved initial overacidification and subsequent alkalization that restored their resting pH. GABAergic SVs that lacked the vesicular GABA transporter (VGAT) did not show the pH overshoot and acidified further to ∼6.0. Comparison of luminal pH dynamics in the presence or absence of VGAT showed that VGAT operates as a GABA/H(+) exchanger, which is continuously required to offset GABA leakage. Furthermore, the kinetics of GABA transport was slower (τ > 20 s at physiological temperature) than that of glutamate uptake and may exceed the time required for reuse of exocytosed SVs, allowing reuse of incompletely filled vesicles in the presence of high demand for inhibitory transmission.


Assuntos
Neurônios/metabolismo , Neurotransmissores/metabolismo , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Exocitose/genética , Ácido Glutâmico/metabolismo , Hipocampo/metabolismo , Hipocampo/fisiologia , Concentração de Íons de Hidrogênio , Cinética , Camundongos , Neurônios/fisiologia , Neurotransmissores/genética , Terminações Pré-Sinápticas/metabolismo , Terminações Pré-Sinápticas/fisiologia , Sinapses/genética , Sinapses/metabolismo , Transmissão Sináptica/fisiologia , Vesículas Sinápticas/metabolismo , Proteínas Vesiculares de Transporte de Aminoácidos Inibidores/genética
7.
J Biol Chem ; 291(5): 2237-45, 2016 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-26637354

RESUMO

Protein arginine methyltransferase 1 (PRMT1) is involved in cell proliferation, DNA damage response, and transcriptional regulation. Although PRMT1 is extensively expressed in the CNS at embryonic and perinatal stages, the physiological role of PRMT1 has been poorly understood. Here, to investigate the primary function of PRMT1 in the CNS, we generated CNS-specific PRMT1 knock-out mice by the Cre-loxP system. These mice exhibited postnatal growth retardation with tremors, and most of them died within 2 weeks after birth. Brain histological analyses revealed prominent cell reduction in the white matter tracts of the mutant mice. Furthermore, ultrastructural analysis demonstrated that myelin sheath was almost completely ablated in the CNS of these animals. In agreement with hypomyelination, we also observed that most major myelin proteins including myelin basic protein (MBP), 2',3'-cyclic-nucleotide 3'-phosphodiesterase (CNPase), and myelin-associated glycoprotein (MAG) were dramatically decreased, although neuronal and astrocytic markers were preserved in the brain of CNS-specific PRMT1 knock-out mice. These animals had a reduced number of OLIG2(+) oligodendrocyte lineage cells in the white matter. We found that expressions of transcription factors essential for oligodendrocyte specification and further maturation were significantly suppressed in the brain of the mutant mice. Our findings provide evidence that PRMT1 is required for CNS development, especially for oligodendrocyte maturation processes.


Assuntos
Sistema Nervoso Central/fisiopatologia , Bainha de Mielina/metabolismo , Bainha de Mielina/patologia , Proteína-Arginina N-Metiltransferases/metabolismo , Animais , Encéfalo/metabolismo , Linhagem da Célula , Proliferação de Células , Sistema Nervoso Central/metabolismo , Dano ao DNA , Deleção de Genes , Genótipo , Glicoproteínas/metabolismo , Camundongos , Camundongos Knockout , Mutação , Proteína Básica da Mielina/metabolismo , Oligodendroglia/citologia , Oligodendroglia/metabolismo , Processamento de Proteína Pós-Traducional , Proteína-Arginina N-Metiltransferases/deficiência , Proteína-Arginina N-Metiltransferases/genética
8.
J Biol Chem ; 291(21): 11241-51, 2016 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-27033703

RESUMO

The G protein-coupled receptor APJ and its ligand apelin are highly expressed in cardiovascular tissues and are associated with the regulation of blood pressure and cardiac function. Although accumulating evidence suggests that APJ plays a crucial role in the heart, it remains unclear whether up-regulation of APJ affects cardiac function. Here we generated cardiomyocyte-specific APJ-overexpressing (APJ-TG) mice and investigated the cardiac phenotype in APJ-TG mice. Male and non-pregnant APJ-TG mice showed cardiac hypertrophy, contractile dysfunction, and elevation of B-type natriuretic peptide gene expression in the heart but not cardiac fibrosis and symptoms of heart failure, including breathing abnormality and pleural effusion. We further examined the influence of APJ overexpression in response to physiological stress induced by pregnancy and lactation in the heart. Interestingly, repeating pregnancy and lactation (pregnancy-lactation cycle) exacerbated cardiac hypertrophy and systolic dysfunction and induced cardiac fibrosis, lung congestion, pleural effusion, and abnormal breathing in APJ-TG mice. These data indicate that female APJ-TG mice develop postpartum cardiomyopathy. We showed that lactation, but not parturition, was critical for the onset of postpartum cardiomyopathy in APJ-TG mice. Furthermore, we found that lactating APJ-TG mice showed impaired myocardial angiogenesis and imbalance of pro- and antiangiogenic gene expression in the heart. These results demonstrate that overexpression of APJ in cardiomyocytes has adverse effects on cardiac function in male and non-pregnant mice and that lactation contributes to the development of postpartum cardiomyopathy in the heart with APJ overexpression.


Assuntos
Insuficiência Cardíaca/metabolismo , Lactação , Miócitos Cardíacos/metabolismo , Período Pós-Parto/metabolismo , Receptores Acoplados a Proteínas G/biossíntese , Animais , Receptores de Apelina , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Feminino , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/patologia , Humanos , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/patologia , Derrame Pleural/genética , Derrame Pleural/patologia , Período Pós-Parto/genética , Gravidez , Receptores Acoplados a Proteínas G/genética
9.
Am J Physiol Heart Circ Physiol ; 308(8): H931-41, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25681427

RESUMO

Apelin peptide is an endogenous ligand of APJ (a putative receptor protein related to the angiotensin II type 1 receptor), which is a member of a G protein-coupled receptor superfamily with seven transmembrane domains. Recent findings have suggested that the apelin-APJ system plays a potential role in cardiac contraction and cardioprotection. In the present study, we show that the apelin-APJ system is disrupted in doxorubicin (Dox)-induced cardiotoxicity. We found downregulation of apelin and APJ mRNA expression in C57Bl/6J mouse hearts on days 1 and 5 after Dox administration (20 mg/kg ip). Plasma apelin levels and cardiac APJ protein expression were significantly decreased on day 5 after Dox injection. Cardiac apelin contents were reduced on day 1 but increased to basal levels on day 5 after Dox injection. We also examined the effects of APJ gene deletion on Dox-induced cardiotoxicity. Compared with wild-type mice, APJ knockout mice showed a significant depression in cardiac contractility on day 5 after Dox (15 mg/kg ip) treatment followed by a decrease in 14-day survival rates. Moreover, Dox-induced myocardial damage, cardiac protein carbonylation, and autophagic dysfunction were accelerated in APJ knockout mice. Rat cardiac H9c2 cells showed Dox-induced decreases in viability, which were prevented by APJ overexpression and the combination with apelin treatment. These results suggest that the suppression of APJ expression after Dox administration can exacerbate Dox-induced cardiotoxicity, which may be responsible for depressed protective function of the endogenous apelin-APJ system. Modulation of the apelin-APJ system may hold promise for the treatment of Dox-induced cardiotoxicity.


Assuntos
Adipocinas/metabolismo , Regulação para Baixo , Doxorrubicina/toxicidade , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Receptores Acoplados a Proteínas G/metabolismo , Adipocinas/genética , Animais , Apelina , Receptores de Apelina , Autofagia , Linhagem Celular , Doxorrubicina/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Contração Miocárdica , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/fisiologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Receptores Acoplados a Proteínas G/genética
10.
Lab Invest ; 94(3): 321-30, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24336072

RESUMO

Agenesis of the corpus callosum (ACC) is a congenital abnormality of the brain structure. More than 60 genes are known to be involved in corpus callosum development. However, the molecular mechanisms underlying ACC are not fully understood. Previously, we produced a novel transgenic mouse strain, TAS, carrying genes of the tetracycline-inducible expression system that are not involved in brain development, and inherited ACC was observed in the brains of all homozygous TAS mice. Although ACC was probably induced by transgene insertion mutation, the causative gene and the molecular mechanism of its pathogenesis remain unclear. Here, we first performed interphase three-color fluorescence in situ hybridization (FISH) analysis to determine the genomic insertion site. Transgenes were inserted into chromosome 18 ∼12.0 Mb from the centromere. Gene expression analysis and genomic PCR walking showed that the genomic region containing exon 4 of Cables1 was deleted by transgene insertion and the other exons of Cables1 were intact. The mutant allele was designated as Cables1(TAS). Interestingly, Cables1(TAS) mRNA consisted of exons 1-3 of Cables1 and part of the transgene that encoded a novel truncated Cables1 protein. Homozygous TAS mice exhibited mRNA expression of Cables1(TAS) in the fetal cerebrum, but not that of wild-type Cables1. To investigate whether a dominant negative effect of Cables1(TAS) or complete loss of function of Cables1 gives rise to ACC, we produced Cables1-null mutant mice. ACC was not observed in Cables1-null mutant mice, suggesting that a dominant negative effect of Cables1(TAS) impairs callosal formation. Moreover, ACC frequency in Cables1(+/TAS) mice was significantly lower than that in Cables1(-/TAS) mice, indicating that wild-type Cables1 interfered with the dominant negative effect of Cables1(TAS). This study indicated that truncated Cables1 causes ACC and wild-type Cables1 contributes to callosal formation.


Assuntos
Agenesia do Corpo Caloso/genética , Proteínas de Transporte/genética , Ciclinas/deficiência , Ciclinas/genética , Fosfoproteínas/deficiência , Fosfoproteínas/genética , Agenesia do Corpo Caloso/metabolismo , Agenesia do Corpo Caloso/patologia , Animais , Éxons , Estudos de Associação Genética , Homozigoto , Hibridização in Situ Fluorescente , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Knockout , Camundongos Transgênicos , Mutagênese Insercional , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
11.
J Recept Signal Transduct Res ; 34(4): 307-12, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24512471

RESUMO

G-protein-coupled receptor 39 (GPR39), a member of the ghrelin receptor family, has a full-length isoform GPR39-1a and a truncated isoform GPR39-1b. While GPR39-1a was clarified as a receptor of Zn(2+), the characteristic property of GPR39-1b remains unknown. Therefore, in this study, the molecular functions of GPR39-1b were explored in cell culture. In contrast to GPR39-1a, GPR39-1b showed no response to Zn(2+) stimulation in calcium mobilization assays, suggesting that GPR39-1b is not a functional receptor of Zn(2+). To understand the signaling interaction of GPR39-1b, we investigated the dimerization between the isoforms, and conducted bioluminescence resonance energy transfer (BRET(2)) assays. The results indicated that GPR39-1b homodimerized, but did not heterodimerize with GPR39-1a. We subsequently attempted to search the heterodimeric counterparts of GPR39-1b. Neurotensin receptor 1 (NTSR1) was also targeted as a GPR39-1b interacting partner because of its highly conserved amino acid sequence and mRNA localization, which was similar to GPR39-1b. BRET(2) assays demonstrated that GPR39-1b heterodimerized with NTSR1. To examine the effect of GPR39-1b on NTRS1-mediated cAMP/PKA signaling, we used the cAMP responsive element-luciferase assays and observed that GPR39-1b attenuated neurotensin-induced NTSR1 signaling. Taken together, our results provided a novel regulatory mechanism for GPR39-1b in NTRS1 signaling.


Assuntos
Cálcio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neurotensina/metabolismo , Transdução de Sinais/genética , Técnicas de Transferência de Energia por Ressonância de Bioluminescência , Cálcio/química , Linhagem Celular , AMP Cíclico/metabolismo , Dimerização , Células HEK293 , Humanos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/genética , Receptores Acoplados a Proteínas G/química , Receptores de Neurotensina/química , Transdução de Sinais/efeitos dos fármacos , Zinco/metabolismo
12.
J Recept Signal Transduct Res ; 34(5): 401-9, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24734888

RESUMO

Renin is predominantly expressed in juxtaglomerular cells in the kidney and regulates blood pressure homeostasis. To examine possible in vivo functions of a mouse distal enhancer (mdE), we generated transgenic mice (TgM) carrying either wild-type or mdE-deficient renin BACs (bacterial artificial chromosome), integrated at the identical chromosomal site. In the kidneys of the TgM, the mdE contributed 80% to basal renin promoter activity. To test for possible physiological roles for the mdE, renin BAC transgenes were used to rescue the hypotensive renin-null mice. Interestingly, renal renin expression in the Tg(BAC):renin-null compound mice was indistinguishable between the wild-type and mutant BAC carriers. Surprisingly, however, the plasma renin activity and angiotensin I concentration in the mdE compound mutant mice were significantly lower than the same parameters in the control mice, and the mutants were consistently hypotensive, demonstrating that blood pressure homeostasis is regulated through transcriptional cis elements controlling renin activity.


Assuntos
Pressão Sanguínea/fisiologia , Elementos Facilitadores Genéticos/genética , Homeostase/fisiologia , Renina/genética , Renina/metabolismo , Ativação Transcricional/genética , Animais , Cromossomos Artificiais Bacterianos/genética , Camundongos , Camundongos Knockout , Camundongos Mutantes , Camundongos Transgênicos , Regiões Promotoras Genéticas/genética
13.
J Recept Signal Transduct Res ; 34(4): 299-306, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24593255

RESUMO

CONTEXT: There are few short-term mouse models of chronic obstructive pulmonary disease (COPD) mimicking the human disease. In addition, p38 is recently recognized as a target for the treatment of COPD. However, the precise mechanism how p38 contributes to the pathogenesis of COPD is still unknown. OBJECTIVE: We attempted to create a new mouse model for COPD by intra-tracheal administration of a mixture of lipopolysaccharide (LPS) and cigarette smoke solution (CSS), and investigated the importance of the p38 mitogen-activated protein kinase (p38) pathway in the pathogenesis of COPD. METHODS: Mice were administered LPS + CSS once a day on days 0-4 and 7-11. Thereafter, CSS alone was administered to mice once a day on days 14-18. On day 28, histopathological changes of the lung were evaluated, and bronchoalveolar lavage fluid (BALF) was subjected to western blot array for cytokines. Transgenic (TG) mice expressing a constitutive-active form of MKK6, a p38-specific activator in the lung, were subjected to our experimental protocol of COPD model. RESULTS: LPS + CSS administration induced enlargement of alveolar air spaces and destruction of lung parenchyma. BALF analyses of the LPS + CSS group revealed an increase in expression levels of several cytokines involved in the pathogenesis of human COPD. These results suggest that our experimental protocol can induce COPD in mice. Likewise, histopathological findings of the lung and induction of cytokines in BALF from MKK6 c.a.-TG mice were more marked than those in WT mice. CONCLUSION: In a new experimental COPD mouse model, p38 accelerates the development of emphysema.


Assuntos
Enfisema/genética , MAP Quinase Quinase 6/metabolismo , Doença Pulmonar Obstrutiva Crônica/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Animais , Modelos Animais de Doenças , Enfisema/etiologia , Enfisema/patologia , Humanos , Lipopolissacarídeos/toxicidade , MAP Quinase Quinase 6/genética , Camundongos , Camundongos Transgênicos , Doença Pulmonar Obstrutiva Crônica/induzido quimicamente , Doença Pulmonar Obstrutiva Crônica/complicações , Doença Pulmonar Obstrutiva Crônica/patologia , Fumar/efeitos adversos , Produtos do Tabaco/toxicidade , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese
14.
Exp Anim ; 73(1): 11-19, 2024 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-37460310

RESUMO

The interconnection of heart performance and kidney function plays an important role for maintaining homeostasis through a variety of physiological crosstalk between these organs. It has been suggested that acute or chronic dysfunction in one organ causes dysregulation in another one, like patients with cardiorenal syndrome. Despite its growing recognition as global health issues, still little is known on pathophysiological evaluation between the two organs. Previously, we established a preclinical murine model with cardiac hypertrophy and fibrosis, and impaired kidney function with renal enlargement and increased urinary albumin levels induced by co-treatment with vasopressor angiotensin II (A), unilateral nephrectomy (N), and salt loading (S) (defined as ANS treatment) for 4 weeks. However, how both tissues, heart and kidney, are initially affected by ANS treatment during the progression of tissue damages remains to be determined. Here, at one week after ANS treatment, we found that cardiac function in ANS-treated mice (ANS mice) are sustained despite hypertrophy. On the other hand, kidney dysfunction is evident in ANS mice, associated with high blood pressure, enlarged glomeruli, increased levels of urinary albumin and urinary neutrophil gelatinase-associated lipocalin, and reduced creatinine clearance. Our results suggest that cardiorenal tissues become damaged at one week after ANS treatment and that ANS mice are useful as a model causing transition from early to late-stage damages of cardiorenal tissues.


Assuntos
Angiotensina II , Síndrome Cardiorrenal , Humanos , Camundongos , Animais , Cloreto de Sódio na Dieta/efeitos adversos , Nefrectomia/efeitos adversos , Rim , Síndrome Cardiorrenal/tratamento farmacológico , Albuminas
15.
J Biol Chem ; 287(29): 24228-38, 2012 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-22637476

RESUMO

One of the mitogen-activated protein kinases, p38, has been found to play a crucial role in various inflammatory responses. In this study, we analyzed the roles of p38α in multiple sclerosis, using an animal model, experimental autoimmune encephalomyelitis (EAE). p38α(+/-) mice (p38α(-/-) showed embryonic lethality) showed less severe neurological signs than WT mice. Adoptive transfer of lymph node cells (LNC) from sensitized WT mice with MOG(35-55) to naive WT-induced EAE was much more severe compared with the case using LNC from sensitized p38α(+/-) mice. Comprehensive analysis of cytokines from MOG(35-55)-challenged LNC by Western blot array revealed that production of IL-17 was significantly reduced by a single copy disruption of the p38α gene or a p38 inhibitor. Likewise, by a luciferase reporter assay, an electrophoresis mobility shift assay, and characterization of the relationship between p38 activity and IL-17 mRNA expression, we confirmed that p38 positively regulates transcription of the Il17 gene. Furthermore, oral administration of a highly specific p38α inhibitor (UR-5269) to WT mice at the onset of EAE markedly suppressed the progression of EAE compared with a vehicle group. These results suggest that p38α participates in the pathogenesis of EAE through IL-17 induction.


Assuntos
Encefalomielite Autoimune Experimental/metabolismo , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Animais , Ensaio de Desvio de Mobilidade Eletroforética , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/genética , Inibidores Enzimáticos/uso terapêutico , Feminino , Interleucina-17/genética , Interleucina-17/metabolismo , Masculino , Camundongos , Proteína Quinase 14 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 14 Ativada por Mitógeno/genética , Regiões Promotoras Genéticas , Estabilidade de RNA/genética
16.
Hypertension ; 79(7): 1409-1422, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35534926

RESUMO

BACKGROUND: ANP (atrial natriuretic peptide), acting through NPR1 (natriuretic peptide receptor 1), provokes hypotension. Such hypotension is thought to be due to ANP inducing vasodilation via NPR1 in the vasculature; however, the underlying mechanism remains unclear. Here, we investigated the mechanisms of acute and chronic blood pressure regulation by ANP. METHODS AND RESULTS: Immunohistochemical analysis of rat tissues revealed that NPR1 was abundantly expressed in endothelial cells and smooth muscle cells of small arteries and arterioles. Intravenous infusion of ANP significantly lowered systolic blood pressure in wild-type mice. ANP also significantly lowered systolic blood pressure in smooth muscle cell-specific Npr1-knockout mice but not in endothelial cell-specific Npr1-knockout mice. Moreover, ANP significantly lowered systolic blood pressure in Nos3-knockout mice. In human umbilical vein endothelial cells, treatment with ANP did not influence nitric oxide production or intracellular Ca2+ concentration, but it did hyperpolarize the cells. ANP-induced hyperpolarization of human umbilical vein endothelial cells was inhibited by several potassium channel blockers and was also abolished under knockdown of RGS2 (regulator of G-protein signaling 2), an GTPase activating protein in G-protein α-subunit. ANP increased Rgs2 mRNA expression in human umbilical vein endothelial cells but failed to lower systolic blood pressure in Rgs2-knockout mice. Endothelial cell-specific Npr1-overexpressing mice exhibited lower blood pressure than did wild-type mice independent of RGS2, and showed dilation of arterial vessels on synchrotron radiation microangiography. CONCLUSIONS: Together, these results indicate that vascular endothelial NPR1 plays a crucial role in ANP-mediated blood pressure regulation, presumably by a mechanism that is RGS2-dependent in the acute phase and RGS2-independent in the chronic phase.


Assuntos
Fator Natriurético Atrial , Pressão Sanguínea , Receptores do Fator Natriurético Atrial , Animais , Fator Natriurético Atrial/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Camundongos , Camundongos Knockout , Ratos , Receptores do Fator Natriurético Atrial/metabolismo
17.
Elife ; 102021 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-33949947

RESUMO

In vivo function of CDK5 and Abl enzyme substrate 2 (Cables2), belonging to the Cables protein family, is unknown. Here, we found that targeted disruption of the entire Cables2 locus (Cables2d) caused growth retardation and enhanced apoptosis at the gastrulation stage and then induced embryonic lethality in mice. Comparative transcriptome analysis revealed disruption of Cables2, 50% down-regulation of Rps21 abutting on the Cables2 locus, and up-regulation of p53-target genes in Cables2d gastrulas. We further revealed the lethality phenotype in Rps21-deleted mice and unexpectedly, the exon 1-deleted Cables2 mice survived. Interestingly, chimeric mice derived from Cables2d ESCs carrying exogenous Cables2 and tetraploid wild-type embryo overcame gastrulation. These results suggest that the diminished expression of Rps21 and the completed lack of Cables2 expression are intricately involved in the embryonic lethality via the p53 pathway. This study sheds light on the importance of Cables2 locus in mouse embryonic development.


Assuntos
Proteínas de Ciclo Celular/genética , Gastrulação/genética , Expressão Gênica , Proteínas Ribossômicas/genética , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos ICR , Fenótipo , Ativação Transcricional , Proteína Supressora de Tumor p53/genética , Regulação para Cima
18.
J Biochem ; 166(5): 383-392, 2019 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-31504625

RESUMO

The apelin receptor (APJ), a receptor for apelin and elabela/apela, induces vasodilation and vasoconstriction in blood vessels. However, the prolonged effects of increased APJ-mediated signalling, involving vasoconstriction, in smooth muscle cells have not been fully characterized. Here, we investigated the vasoactive effects of APJ gain of function under the control of the smooth muscle actin (SMA) gene promoter in mice. Transgenic overexpression of APJ (SMA-APJ) conferred sensitivity to blood pressure and vascular contraction induced by apelin administration in vivo. Interestingly, ex vivo experiments showed that apelin markedly increased the vasoconstriction of isolated aorta induced by noradrenaline (NA), an agonist for α- and ß-adrenergic receptors, or phenylephrine, a specific agonist for α1-adrenergic receptor (α1-AR). In addition, intracellular calcium influx was augmented by apelin with NA in HEK293T cells expressing APJ and α1A-AR. To examine the cooperative action of APJ and α1A-AR in the regulation of vasoconstriction, we developed α1A-AR deficient mice using a genome-editing technique, and then established SMA-APJ/α1A-AR-KO mice. In the latter mouse line, aortic vasoconstriction induced by a specific agonist for α1A-AR, A-61603, were significantly less than in SMA-APJ mice. These results suggest that the APJ-enhanced response requires α1A-AR to contract vessels coordinately.


Assuntos
Receptores de Apelina/metabolismo , Músculo Liso Vascular/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Vasoconstrição , Animais , Humanos , Camundongos , Camundongos Endogâmicos ICR , Camundongos Transgênicos , Músculo Liso Vascular/química
19.
Lab Invest ; 88(4): 416-29, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18268474

RESUMO

Preeclampsia is a serious disorder that may result in severe morbidity and mortality for mother and fetus, and it is thought that the placental dysfunction is important in the pathogenesis of preeclampsia. As the model of preeclampsia, we previously generated a transgenic mouse model that developed pregnancy-associated hypertension (PAH) by mating females expressing human angiotensinogen with males expressing human renin. In PAH mice, maternal blood pressure started to rise from days 12 to 13 of gestation (E12-13) to term (E19-20), which is accompanied by the fetal intrauterine growth retardation and systemic maternal disorders including proteinuria and convulsion. To understand the pathology of the complications in PAH mice that overlap with those in human preeclampsia, we analyzed the PAH placenta sequentially from the onset of hypertension to the term of delivery. In PAH placenta, histological analysis revealed that the microvessel densities of fetal vasculature at term were significantly lower than those of normal placenta, and the majority of terminal vessels of PAH placenta were lacking for pericytes and basement membrane. The interaction between fetal vasculature and maternal blood canal at labyrinth of PAH placenta was morphologically distorted, and the expression patterns of key molecules in neovascularization of PAH placenta were distinct from those of normal placenta during pregnancy. In addition, maternal plasma level of soluble form of vascular endothelial growth factor receptor-1 (sVEGFR-1) was significantly increased in PAH at E19. Furthermore, in uteroplacental site, in situ proteolytic activity of PAH mice was suppressed from E16 to term compared to that of normal pregnancy, and the expression of matrix metalloproteinase-2 mRNA was strikingly downregulated at E16 in PAH mice. Collective data suggest that the impairments of fetoplacental neovascularization and uteroplacental remodeling contribute to the development of complications in PAH.


Assuntos
Membrana Basal/patologia , Hipertensão Induzida pela Gravidez/patologia , Neovascularização Fisiológica/fisiologia , Placenta/patologia , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/sangue , Animais , Feminino , Feto/patologia , Expressão Gênica , Hipertensão Induzida pela Gravidez/sangue , Hipertensão Induzida pela Gravidez/fisiopatologia , Infarto/patologia , Masculino , Metaloproteinases da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Peptídeo Hidrolases/metabolismo , Pericitos/citologia , Placenta/irrigação sanguínea , Placenta/enzimologia , Placenta/fisiopatologia , Gravidez , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/citologia
20.
Lab Invest ; 88(7): 731-9, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18490898

RESUMO

The renin-angiotensin system (RAS) modulates end-organ damages, resulting in cardiovascular and kidney diseases. Experiments both in vitro and in vivo demonstrate that the angiotensin II (Ang II) type 1 (AT1) receptor pathway also exerts pro-inflammatory and pro-atherogenic effects on bone marrow-derived cells (BMDCs). Here, we investigated how AT1 receptor expression by BMDCs contributes to atherosclerosis and kidney injury in vivo by transplanting BM into RAS-activated transgenic mice. There was no difference in the extent of kidney damage between mice receiving BM transplants from mutant mice lacking the angiotensin II type 1a receptor (AT1a) gene and mice receiving transplants from wild-type (WT) mice. However, mice receiving transplants from AT1a 'knockout' (KO) mice displayed accelerated lethality and atherosclerotic lesions. These results indicated that the effects of AT1a receptor on BMDCs are organ dependent. Microarray expression profiling of macrophages from AT1a-KO mice revealed significant changes in the mRNA levels for a number of genes implicated in atherosclerosis. In accordance with the in vivo atherosclerosis results, AT1a-KO macrophages exhibited greater uptake of modified lipoproteins relative to macrophages from WT mice. We propose that the expression of AT1a receptor by BMDCs limits atherosclerosis in vivo.


Assuntos
Aterosclerose/patologia , Células da Medula Óssea/patologia , Receptor Tipo 1 de Angiotensina/fisiologia , Animais , Aorta/patologia , Aterosclerose/metabolismo , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea , Células Cultivadas , Feminino , Perfilação da Expressão Gênica , Rim/metabolismo , Rim/patologia , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/patologia , Camundongos , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Receptor Tipo 1 de Angiotensina/genética , Baço/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa