Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 138
Filtrar
1.
Physiol Rev ; 92(4): 1813-64, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23073632

RESUMO

The neurohypophysial hormone arginine vasopressin (AVP) is essential for a wide range of physiological functions, including water reabsorption, cardiovascular homeostasis, hormone secretion, and social behavior. These and other actions of AVP are mediated by at least three distinct receptor subtypes: V1a, V1b, and V2. Although the antidiuretic action of AVP and V2 receptor in renal distal tubules and collecting ducts is relatively well understood, recent years have seen an increasing understanding of the physiological roles of V1a and V1b receptors. The V1a receptor is originally found in the vascular smooth muscle and the V1b receptor in the anterior pituitary. Deletion of V1a or V1b receptor genes in mice revealed that the contributions of these receptors extend far beyond cardiovascular or hormone-secreting functions. Together with extensively developed pharmacological tools, genetically altered rodent models have advanced the understanding of a variety of AVP systems. Our report reviews the findings in this important field by covering a wide range of research, from the molecular physiology of V1a and V1b receptors to studies on whole animals, including gene knockout/knockdown studies.


Assuntos
Receptores de Vasopressinas/metabolismo , Vasopressinas/metabolismo , Animais , Camundongos , Receptores de Vasopressinas/genética , Transdução de Sinais/fisiologia
2.
Biochem Biophys Res Commun ; 486(2): 506-513, 2017 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-28322798

RESUMO

In an embryonic developmental stage of the peripheral nervous system (PNS), Schwann cell precursors migrate along neuronal axons to their final destinations. After birth, they eventually wrap around individual axons to form myelin sheaths, which insulate axons to increase the nerve conduction velocity. Some growth factors and adhesion molecules are known to control these developmental stages from in the fish to in the mammal. Neuregulin-1 (NRG1), which is composed of many alternative splicing variants, is such a growth factor. Among these variants, the type III isoform of NRG1, interacting with ErbB2 and ErbB3 receptors on Schwann cells, plays an essential role in myelination in the fish and the mammal. NRG1 type III is also known to promote migration of fish Schwann cell precursors; however, it still remains to be clarified whether mammalian type III isoform does it. We have therefore generated type III isoform-specific knockout mice in inbred strain. The mice result in delayed migration of the precursors from the dorsal to ventral root via a peripheral ganglion, comparing littermate controls. Similar results are observed in an in vitro migration assay using reaggregated Schwann cell precursors. Furthermore, the knockout mice exhibit reduced myelin thickness, consistent with the established role of NRG1 type III in myelination. These results indicate that in mice, NRG1 type III plays a key role not only in myelination but also in migration.


Assuntos
Bainha de Mielina/genética , Neuregulina-1/metabolismo , Receptor ErbB-2/metabolismo , Receptor ErbB-3/metabolismo , Células de Schwann/metabolismo , Medula Espinal/metabolismo , Animais , Diferenciação Celular , Movimento Celular , Expressão Gênica , Camundongos , Camundongos Knockout , Bainha de Mielina/metabolismo , Bainha de Mielina/patologia , Neuregulina-1/genética , Ratos , Ratos Sprague-Dawley , Receptor ErbB-2/genética , Receptor ErbB-3/genética , Células de Schwann/patologia , Transdução de Sinais , Medula Espinal/patologia
3.
J Biol Chem ; 289(49): 33887-903, 2014 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-25326380

RESUMO

The mechanism of neurite growth is complicated, involving continuous cytoskeletal rearrangement and vesicular trafficking. Cytohesin-2 is a guanine nucleotide exchange factor for Arf6, an Arf family molecular switch protein, controlling cell morphological changes such as neuritogenesis. Here, we show that cytohesin-2 binds to a protein with a previously unknown function, CCDC120, which contains three coiled-coil domains, and is transported along neurites in differentiating N1E-115 cells. Transfection of the small interfering RNA (siRNA) specific for CCDC120 into cells inhibits neurite growth and Arf6 activation. When neurites start to extend, vesicles containing CCDC120 and cytohesin-2 are transported in an anterograde manner rather than a retrograde one. As neurites continue extension, anterograde vesicle transport decreases. CCDC120 knockdown inhibits cytohesin-2 localization into vesicles containing CCDC120 and diffuses cytohesin-2 in cytoplasmic regions, illustrating that CCDC120 determines cytohesin-2 localization in growing neurites. Reintroduction of the wild type CCDC120 construct into cells transfected with CCDC120 siRNA reverses blunted neurite growth and Arf6 activity, whereas the cytohesin-2-binding CC1 region-deficient CCDC120 construct does not. Thus, cytohesin-2 is transported along neurites by vesicles containing CCDC120, and it mediates neurite growth. These results suggest a mechanism by which guanine nucleotide exchange factor for Arf6 is transported to mediate neurite growth.


Assuntos
Proteínas Ativadoras de GTPase/genética , Proteínas do Tecido Nervoso/genética , Neurogênese/genética , Neurônios/metabolismo , Vesículas Transportadoras/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Linhagem Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Regulação da Expressão Gênica , Humanos , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais
4.
Biochem Biophys Res Commun ; 465(3): 450-7, 2015 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-26277388

RESUMO

During development of the peripheral nervous system (PNS), Schwann cells wrap neuronal axons, becoming the myelin sheaths that help axonal functions. While the intercellular signals controlling the myelination process between Schwann cells and peripheral neurons are well studied, the transduction of these signals in Schwann cells still remains elusive. Here, we show that Arf6, an Arf protein of the small GTPase family, is involved in promoting the myelination process. Knockdown of Arf6 with the small-interfering (si)RNA in primary Schwann cells markedly decreases dibutyl-cyclic AMP-induced myelin marker protein expression, indicating that Arf6 plays a role in differentiation-like phenotypic changes. To obtain in vivo evidence, we generated small-hairpin (sh)RNA transgenic mice targeting Arf6 for Schwann cells. Transgenic mice exhibited reduced myelin thickness compared to littermate controls, consistent with the defective myelin formation observed in the transgenic mouse-derived Schwann cell and neuronal culture system. Transgenic mice also exhibited decreased phosphorylation of myelination-related signaling molecules such as Akt kinase cascade proteins as well as downregulation of myelin marker proteins. These results suggest that signaling through Arf6 is required for Schwann cell myelination, adding Arf6 to the list of intracellular signaling molecules involved in the myelination process.


Assuntos
Fatores de Ribosilação do ADP/metabolismo , Diferenciação Celular/fisiologia , Bainha de Mielina/fisiologia , Neurogênese/fisiologia , Células de Schwann/citologia , Células de Schwann/metabolismo , Fator 6 de Ribosilação do ADP , Animais , Células Cultivadas , Feminino , Técnicas de Silenciamento de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia
5.
Biochem Biophys Res Commun ; 462(3): 275-81, 2015 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-25957474

RESUMO

Myelin-forming glial cells undergo dynamic morphological changes in order to produce mature myelin sheaths with multiple layers. In the central nervous system (CNS), oligodendrocytes differentiate to insulate neuronal axons with myelin sheaths. Myelin sheaths play a key role in homeostasis of the nervous system, but their related disorders lead not only to dismyelination and repeated demyelination but also to severe neuropathies. Hereditary hypomyelinating leukodystrophies (HLDs) are a group of such diseases affecting oligodendrocytes and are often caused by missense mutations of the respective responsible genes. Despite increasing identification of gene mutations through advanced nucleotide sequencing technology, studies on the relationships between gene mutations and their effects on cellular and subcellular aberrance have not followed at the same rapid pace. In this study, we report that an HLD4-associated (Asp-29-to-Gly) mutant of mitochondrial heat shock 60-kDa protein 1 (HSPD1) causes short-length morphologies and increases the numbers of mitochondria due to their aberrant fission and fusion cycles. In experiments using a fluorescent dye probe, this mutation decreases the mitochondrial membrane potential. Also, mitochondria accumulate in perinuclear regions. HLD4-associated HSPD1 mutant blunts mitochondrial dynamics, probably resulting in oligodendrocyte malfunction. This study constitutes a first finding concerning the relationship between disease-associated HSPD1 mutation and mitochondrial dynamics, which may be similar to the relationship between another disease-associated HSPD1 mutation (MitCHAP-60 disease) and aberrant mitochondrial dynamics.


Assuntos
Chaperonina 60/genética , Doenças Desmielinizantes Hereditárias do Sistema Nervoso Central/genética , Doenças Mitocondriais/genética , Dinâmica Mitocondrial/genética , Proteínas Mitocondriais/genética , Mutação de Sentido Incorreto , Substituição de Aminoácidos , Animais , Células COS , Chaperonina 60/metabolismo , Chlorocebus aethiops , Doenças Desmielinizantes Hereditárias do Sistema Nervoso Central/metabolismo , Doenças Desmielinizantes Hereditárias do Sistema Nervoso Central/patologia , Humanos , Mitocôndrias/genética , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Doenças Mitocondriais/metabolismo , Doenças Mitocondriais/patologia , Proteínas Mitocondriais/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
6.
Biochem Biophys Res Commun ; 460(3): 819-25, 2015 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-25824033

RESUMO

In postnatal development of the peripheral nervous system (PNS), Schwann cells differentiate to insulate neuronal axons with myelin sheaths, increasing the nerve conduction velocity. To produce the mature myelin sheath with its multiple layers, Schwann cells undergo dynamic morphological changes. While extracellular molecules such as growth factors and cell adhesion ligands are known to regulate the myelination process, the intracellular molecular mechanism underlying myelination remains unclear. In this study, we have produced Schwann cell-specific conditional knockout mice for cytohesin-2, a guanine-nucleotide exchange factor (GEF) specifically activating Arf6. Arf6, a member of the Ras-like protein family, participates in various cellular functions including cell morphological changes. Cytohesin-2 knockout mice exhibit decreased Arf6 activity and reduced myelin thickness in the sciatic nerves, with decreased expression levels of myelin protein zero (MPZ), the major myelin marker protein. These results are consistent with those of experiments in which Schwann cell-neuronal cultures were treated with pan-cytohesin inhibitor SecinH3. On the other hand, the numbers of Ki67-positive cells in knockout mice and controls are comparable, indicating that cytohesin-2 does not have a positive effect on cell numbers. Thus, signaling through cytohesin-2 is required for myelination by Schwann cells, and cytohesin-2 is added to the list of molecules known to underlie PNS myelination.


Assuntos
Proteínas Ativadoras de GTPase/fisiologia , Bainha de Mielina/fisiologia , Animais , Sequência de Bases , Primers do DNA , Feminino , Proteínas Ativadoras de GTPase/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase
7.
Biol Pharm Bull ; 38(8): 1109-12, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26235574

RESUMO

Environmental influences, such as chemical exposure, have long been considered potential risk factors for neurodegenerative disorders, including neuromuscular diseases. However, no definitive links between environmental chemical exposure and a pathogenic mechanism of neurodegenerative disease has yet been established. In this study, we describe that exposure to arsenic, an environmental pollutant naturally found in drinking water, induces neuronal cell death and alteration of morphology, particularly neurite outgrowth and in the cytoskeleton of neurons. Since progressive cell loss accompanied by the alteration of neuronal structures and cytoskeleton is considered the major pathologic feature of neurodegenerative disorders, arsenic-induced neurotoxicity might contribute to an etiologic mechanism of some neurodegenerative diseases. Further, we discuss the importance of in vitro assay, particularly an embryonic toxicity test, for assessing the neurotoxicity of chemicals, because most of chemicals found in our environment remain to be evaluated regarding their neurotoxicity risk for neurodegenerative diseases.


Assuntos
Arsênio/efeitos adversos , Morte Celular , Citoesqueleto/patologia , Poluentes Ambientais/efeitos adversos , Doenças Neurodegenerativas/etiologia , Neurônios/patologia , Síndromes Neurotóxicas/complicações , Animais , Células Cultivadas , Humanos , Neuritos , Doenças Neurodegenerativas/patologia
8.
J Biol Chem ; 288(13): 8910-21, 2013 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-23389032

RESUMO

A K141N missense mutation in heat shock protein (HSP) B8, which belongs to the small HSP family, causes distal hereditary motor neuropathy, which is characterized by the formation of inclusion bodies in cells. Although the HSPB8 gene causes hereditary motor neuropathy, obvious expression of HSPB8 is also observed in other tissues, such as the heart. The effects of a single mutation in HSPB8 upon the heart were analyzed using rat neonatal cardiomyocytes. Expression of HSPB8 K141N by adenoviral infection resulted in increased HSPB8-positive aggregates around nuclei, whereas no aggregates were observed in myocytes expressing wild-type HSPB8. HSPB8-positive aggresomes contained amyloid oligomer intermediates that were detected by a specific anti-oligomer antibody (A11). Expression of HSPB8 K141N induced slight cellular toxicity. Recombinant HSPB8 K141N protein showed reactivity against the anti-oligomer antibody, and reactivity of the mutant HSPB8 protein was much higher than that of wild-type HSPB8 protein. To extend our in vitro study, cardiac-specific HSPB8 K141N transgenic (TG) mice were generated. Echocardiography revealed that the HSPB8 K141N TG mice exhibited mild hypertrophy and apical fibrosis as well as slightly reduced cardiac function, although no phenotype was detected in wild-type HSPB8 TG mice. A single point mutation of HSPB8, such as K141N, can cause cardiac disease.


Assuntos
Cardiomiopatias/metabolismo , Proteínas de Choque Térmico HSP20/genética , Proteínas de Choque Térmico HSP20/fisiologia , Proteínas de Choque Térmico/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/fisiologia , Animais , Cardiomiopatias/genética , Citosol/metabolismo , DNA Complementar/metabolismo , Fibrose/patologia , Imuno-Histoquímica/métodos , Camundongos , Camundongos Transgênicos , Mitocôndrias/metabolismo , Chaperonas Moleculares , Mutação , Mutação de Sentido Incorreto , Fenótipo , Mutação Puntual , Ratos , Proteínas Recombinantes/metabolismo
9.
Biochem Biophys Res Commun ; 452(3): 782-8, 2014 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-25204498

RESUMO

The myelin sheath insulates neuronal axons and markedly increases the nerve conduction velocity. In the peripheral nervous system (PNS), Schwann cell precursors migrate along embryonic neuronal axons to their final destinations, where they eventually wrap around individual axons to form the myelin sheath after birth. ErbB2 and ErbB3 tyrosine kinase receptors form a heterodimer and are extensively expressed in Schwann lineage cells. ErbB2/3 is thought to be one of the primary regulators controlling the entire Schwann cell development. ErbB3 is the bona fide Schwann cell receptor for the neuronal ligand neuregulin-1. Although ErbB2/3 is well known to regulate both Schwann cell precursor migration and myelination by Schwann cells in fishes, it still remains unclear whether in mammals, ErbB2/3 actually regulates Schwann cell precursor migration. Here, we show that knockdown of ErbB3 using a Schwann cell-specific promoter in mice causes delayed migration of Schwann cell precursors. In contrast, littermate control mice display normal migration. Similar results are seen in an in vitro migration assay using reaggregated Schwann cell precursors. Also, ErbB3 knockdown in mice reduces myelin thickness in sciatic nerves, consistent with the established role of ErbB3 in myelination. Thus, ErbB3 plays a key role in migration, as well as in myelination, in mouse Schwann lineage cells, presenting a genetically conservative role of ErbB3 in Schwann cell precursor migration.


Assuntos
Movimento Celular/genética , Regulação da Expressão Gênica no Desenvolvimento , Neurogênese/genética , Receptor ErbB-3/genética , Células de Schwann/metabolismo , Animais , Axônios/metabolismo , Axônios/ultraestrutura , Diferenciação Celular , Embrião de Mamíferos , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Bainha de Mielina/metabolismo , Bainha de Mielina/ultraestrutura , Neuregulina-1/genética , Neuregulina-1/metabolismo , Regiões Promotoras Genéticas , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptor ErbB-3/antagonistas & inibidores , Receptor ErbB-3/metabolismo , Células de Schwann/ultraestrutura , Nervo Isquiático/crescimento & desenvolvimento , Nervo Isquiático/metabolismo , Nervo Isquiático/ultraestrutura , Transdução de Sinais
10.
Biochem Biophys Res Commun ; 449(2): 222-8, 2014 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-24832733

RESUMO

Erythropoietin production has been reported to occur in the peritubular interstitial fibroblasts in the kidney. Since the erythropoietin production in the nephron is controversial, we reevaluated the erythropoietin production in the kidney. We examined mRNA expressions of erythropoietin and HIF PHD2 using high-sensitive in situ hybridization system (ISH) and protein expression of HIF PHD2 using immunohistochemistry in the kidney. We further investigated the mechanism of erythropoietin production by hypoxia in vitro using human liver hepatocell (HepG2) and rat intercalated cell line (IN-IC cells). ISH in mice showed mRNA expression of erythropoietin in proximal convoluted tubules (PCTs), distal convoluted tubules (DCTs) and cortical collecting ducts (CCDs) but not in the peritubular cells under normal conditions. Hypoxia induced mRNA expression of erythropoietin largely in peritubular cells and slightly in PCTs, DCTs, and CCDs. Double staining with AQP3 or AE1 indicated that erythropoietin mRNA expresses mainly in ß-intercalated or non α/non ß-intercalated cells of the collecting ducts. Immunohistochemistry in rat showed the expression of HIF PHD2 in the collecting ducts and peritubular cells and its increase by anemia in peritubular cells. In IN-IC cells, hypoxia increased mRNA expression of erythropoietin, erythropoietin concentration in the medium and protein expression of HIF PHD2. These data suggest that erythropoietin is produced by the cortical nephrons mainly in the intercalated cells, but not in the peritubular cells, in normal hematopoietic condition and by mainly peritubular cells in hypoxia, suggesting the different regulation mechanism between the nephrons and peritubular cells.


Assuntos
Eritropoetina/biossíntese , Néfrons/metabolismo , Animais , Hipóxia Celular , Linhagem Celular , Eritropoetina/genética , Regulação da Expressão Gênica , Células Hep G2 , Humanos , Prolina Dioxigenases do Fator Induzível por Hipóxia/genética , Prolina Dioxigenases do Fator Induzível por Hipóxia/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Rim/citologia , Rim/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Pró-Colágeno-Prolina Dioxigenase/genética , Pró-Colágeno-Prolina Dioxigenase/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual
11.
Liver Transpl ; 20(9): 1127-37, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24838399

RESUMO

The pathogenesis of biliary atresia (BA), which leads to end-stage cirrhosis in most patients, has been thought to inflame and obstruct the intrahepatic and extrahepatic bile ducts. BA is not believed to be caused by abnormalities in parenchymal hepatocytes. However, there has been no report of a detailed analysis of hepatocytes buried in the cirrhotic livers of patients with BA. Therefore, we evaluated the proliferative potential of these hepatocytes in immunodeficient, liver-injured mice [the urokinase-type plasminogen activator (uPA) transgenic NOD/Shi-scid IL2rγnull (NOG); uPA-NOG strain]. We succeeded in isolating viable hepatocytes from the livers of patients with BA who had various degrees of fibrosis. The isolated hepatocytes were intrasplenically transplanted into the livers of uPA-NOG mice. The hepatocytes of only 3 of the 9 BA patients secreted detectable amounts of human albumin in sera when they were transplanted into mice. However, human leukocyte antigen-positive hepatocyte colonies were detected in 7 of the 9 mice with hepatocyte transplants from patients with BA. We demonstrated that hepatocytes buried in the cirrhotic livers of patients with BA retained their proliferative potential. A liver that was reconstituted with hepatocytes from patients with BA was shown to be a functioning human liver with a drug-metabolizing enzyme gene expression pattern that was representative of mature human liver and biliary function, as ascertained by fluorescent dye excretion into the bile canaliculi. These results imply that removing the primary etiology via an earlier portoenterostomy may increase the quantity of functionally intact hepatocytes remaining in a cirrhotic liver and may contribute to improved outcomes.


Assuntos
Atresia Biliar/complicações , Proliferação de Células , Hepatócitos/transplante , Subunidade gama Comum de Receptores de Interleucina/metabolismo , Cirrose Hepática/etiologia , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Adulto , Animais , Bile/metabolismo , Atresia Biliar/enzimologia , Atresia Biliar/imunologia , Atresia Biliar/patologia , Biomarcadores/metabolismo , Criança , Pré-Escolar , Sistema Enzimático do Citocromo P-450/metabolismo , Feminino , Glucuronosiltransferase/metabolismo , Antígenos HLA/imunologia , Eliminação Hepatobiliar , Hepatócitos/enzimologia , Hepatócitos/imunologia , Hepatócitos/patologia , Humanos , Lactente , Subunidade gama Comum de Receptores de Interleucina/genética , Isoenzimas , Cirrose Hepática/enzimologia , Cirrose Hepática/imunologia , Cirrose Hepática/patologia , Testes de Função Hepática , Regeneração Hepática , Masculino , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Fenótipo , Albumina Sérica/metabolismo , Albumina Sérica Humana , Ativador de Plasminogênio Tipo Uroquinase/genética
12.
Pediatr Int ; 56(5): 659-66, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25040584

RESUMO

Pelizaeus-Merzbacher disease (PMD) is a rare leukodystrophy that causes severe dysmyelination in the central nervous system in infancy and early childhood. Many previous studies showed that various proteolipid protein 1 (plp1) mutations, including duplications, point mutations, and deletions, lead to oligodendrocyte dysfunction in patients with PMD. PMD onset and clinical severity range widely, depending on the type of plp1 mutation. Patients with PMD exhibit a delayed mental and physical development phenotype, but specific pharmacological therapy and clinical treatment for PMD are not yet well established. This review describes PMD pathology and establishment of new clinical treatment for PMD. These findings support the development of a new therapy for PMD and these treatments may improve the quality of life in patients with PMD.


Assuntos
Doença de Pelizaeus-Merzbacher/etiologia , Criança , Humanos , Doença de Pelizaeus-Merzbacher/tratamento farmacológico , Doença de Pelizaeus-Merzbacher/genética
13.
Clin Exp Nephrol ; 17(2): 183-90, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22968856

RESUMO

BACKGROUND: Vasopressin V1a receptor null (V1aR(-/-)) mice recently showed incomplete urinary concentration due to higher urine volume during control and water diuresis (euhydration), but showed normal response during dehydration (Aoyagi et al., Am J Physiol 295: F100-7, 2008). METHODS: Water balance, plasma vasopressin, plasma and urine osmolality, and aquaporin 2 (AQP2) expression in the kidney of wild-type (WT) and V1aR(-/-) mice were therefore further examined using improved methods of urine collection (urinary bladder urine). RESULTS: V1aR(-/-) mice demonstrated a lower urine osmolality (3,360 ± 138 vs. 3,610 ± 47 mOsm/kgH2O) and a higher plasma osmolality (354.3 ± 1.3 vs. 342.5 ± 1.5 mOsm/kgH2O) after dehydration for 24 h compared to WT mice (P < 0.05). In contrast, the plasma vasopressin concentration was significantly (P < 0.001) higher in the V1aR(-/-) mice (48.8 ± 4.8 vs. 22.1 ± 2.4 pg/ml). On the other hand, although the AQP2 protein expression in the kidney was increased after dehydration, the basal (control) and dehydration-induced AQP2 protein levels were significantly lower in V1aR(-/-) mice compared to WT mice (by Western blotting). Staining by an anti-AQP2 antibody in the luminal membrane of the collecting ducts was increased in both V1aR(-/-) and WT mice after dehydration, but was relatively weaker in the V1aR(-/-) mice (by immunohistochemistry). Moreover, urinary excretion of AQP2 protein, an index of the luminal AQP2 expression, was significantly (P < 0.05) lower in the V1aR(-/-) mice. CONCLUSION: V1aR signaling may be fundamentally important for the expression of AQP2 in the collecting ducts during control conditions and dehydration.


Assuntos
Aquaporina 2/biossíntese , Túbulos Renais Coletores/metabolismo , Receptores de Vasopressinas/genética , Receptores de Vasopressinas/fisiologia , Animais , Análise Química do Sangue , Western Blotting , Desidratação/genética , Desidratação/metabolismo , Eletroforese em Gel de Poliacrilamida , Imuno-Histoquímica , Rim/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas/isolamento & purificação , Proteínas/metabolismo , Frações Subcelulares/metabolismo , Urinálise
14.
Clin Exp Nephrol ; 17(6): 783-92, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23456233

RESUMO

BACKGROUND: Vasopressin V1a receptor (V1aR) null mice have insufficient acid-base balance, but the target cell for V1aR signaling which results in the urinary acidification has not been identified. METHODS: By using a quantitative in situ hybridization technique and a double-staining technique with an anti-AQP3 antibody in mice, we investigated the axial distribution and acidosis-induced expression of V1aR mRNA along the nephron. We also investigated the acidosis-induced morphological change in the tubule cells from wild-type and V1aR-null (V1aR(-/-)) mice. RESULTS: In the normal condition, V1aR mRNA was moderately expressed in the medullary thick ascending limb (MTAL) and highly expressed in the intercalated cell (IC) throughout the collecting duct (CD). However, no expression was observed in the proximal tubule, thin limbs of Henle's loop, and the principal cell of the CD. Importantly, V1aR mRNA was upregulated significantly both in the TAL and the IC of the CD in the inner stripe of the outer medulla (MTALis and IC of OMCDis, respectively) when mice were treated with NH4Cl (0.28 mol/L) for 6 days. Acidosis-induced hypertrophy, which was completely attenuated in V1aR(-/-) mice, was observed only in the IC of OMCDis (P < 0.005). In addition, urinary excretion of ammonia (NH3/NH4 (+)) was significantly decreased on day 3 (P < 0.05) and day 6 (P < 0.005) in the V1aR(-/-) mice treated with NH4Cl. CONCLUSION: In conclusion, the IC of OMCDis may be the target cell stimulated by the vasopressin V1aR axis and contribute to urinary acidification, at least during metabolic acidosis.


Assuntos
Túbulos Renais Coletores/citologia , Receptores de Vasopressinas/fisiologia , Acidose/fisiopatologia , Acidose/urina , Amônia/urina , Cloreto de Amônio/farmacologia , Animais , Células Epiteliais/metabolismo , Concentração de Íons de Hidrogênio , Hibridização In Situ , Túbulos Renais Coletores/efeitos dos fármacos , Túbulos Renais Coletores/metabolismo , Camundongos , Camundongos Knockout , RNA Mensageiro/metabolismo , Receptores de Vasopressinas/biossíntese , Receptores de Vasopressinas/genética
15.
J Neurosci ; 31(35): 12579-92, 2011 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-21880919

RESUMO

In development of the peripheral nervous system, Schwann cells proliferate, migrate, and ultimately differentiate to form myelin sheath. In all of the myelination stages, Schwann cells continuously undergo morphological changes; however, little is known about their underlying molecular mechanisms. We previously cloned the dock7 gene encoding the atypical Rho family guanine-nucleotide exchange factor (GEF) and reported the positive role of Dock7, the target Rho GTPases Rac/Cdc42, and the downstream c-Jun N-terminal kinase in Schwann cell migration (Yamauchi et al., 2008). We investigated the role of Dock7 in Schwann cell differentiation and myelination. Knockdown of Dock7 by the specific small interfering (si)RNA in primary Schwann cells promotes dibutyryl cAMP-induced morphological differentiation, indicating the negative role of Dock7 in Schwann cell differentiation. It also results in a shorter duration of activation of Rac/Cdc42 and JNK, which is the negative regulator of myelination, and the earlier activation of Rho and Rho-kinase, which is the positive regulator of myelination. To obtain the in vivo evidence, we generated Dock7 short hairpin (sh)RNA transgenic mice. They exhibited a decreased expression of Dock7 in the sciatic nerves and enhanced myelin thickness, consistent with in vitro observation. The effects of the in vivo knockdown on the signals to Rho GTPases are similar to those of the in vitro knockdown. Collectively, the signaling through Dock7 negatively regulates Schwann cell differentiation and the onset of myelination, demonstrating the unexpected role of Dock7 in the interplay between Schwann cell migration and myelination.


Assuntos
Diferenciação Celular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Bainha de Mielina/metabolismo , Células de Schwann/fisiologia , Animais , Animais Recém-Nascidos , Bucladesina/farmacologia , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Células Cultivadas , Embrião de Mamíferos , Feminino , Proteínas Ativadoras de GTPase , Gânglios Espinais/citologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/genética , Fatores de Troca do Nucleotídeo Guanina/genética , Imunoprecipitação/métodos , Indóis , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Bainha de Mielina/ultraestrutura , Neuregulina-1/farmacologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Células de Schwann/efeitos dos fármacos , Células de Schwann/ultraestrutura , Nervo Isquiático/citologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transfecção , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo
16.
Am J Physiol Renal Physiol ; 303(7): F1080-8, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22811487

RESUMO

We previously reported that a deficiency in the vasopressin V1a receptor (V1aR) results in type 4 renal tubular acidosis, which suggests that vasopressin exerts direct effects on the physiological actions of aldosterone. We investigated the role of vasopressin for nucleocytoplasmic transport of mineralocorticoid receptor (MR) in the intercalated cells. Vasopressin V1aR-deficient (V1aR(-/-)) mice showed largely decreased expression of MR and 11ß-hydroxysteroid dehydrogenase type 2 (11ßHSD2) in the medulla of the kidney, which was partially ameliorated by fludrocortisone treatment. The incubation of IN-IC cells, an intercalated cell line established from temperature-sensitive SV40 large T antigen-expressing rats, with aldosterone or vasopressin increased the nuclear-to-cytoplasmic ratio of the MR from 11.2 to 47.2% and from 18.7 to 61.2%, respectively, in 30 min without any changes in MR expression from the whole cell extract. The immunohistochemistry analysis of the IN-IC cells revealed the nuclear accumulation of MRs after a 30-min incubation with aldosterone or vasopressin. These effects were accompanied by an increase in regulator of chromosome condensation-1 (RCC-1) due to aldosterone and a decrease in Ran GTPase-activating protein 1 (Ran Gap1) due to vasopressin. RNA interference against V1aR abolished the nuclear accumulation of MR induced by aldosterone or vasopressin. Vasopressin increased PKCα and -ß(1) expression, and aldosterone increased PKCδ and -ζ expression, but these effects were abolished with a V1aR knockdown. These results suggest that vasopressin directly regulates the nucleocytoplasmic transport of MRs via the V1aR in the intercalated cells of the collecting ducts.


Assuntos
11-beta-Hidroxiesteroide Desidrogenase Tipo 2/metabolismo , Medula Renal/metabolismo , Receptores de Mineralocorticoides/metabolismo , Receptores de Vasopressinas/genética , 11-beta-Hidroxiesteroide Desidrogenase Tipo 2/genética , Animais , Linhagem Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Citoplasma/genética , Citoplasma/metabolismo , Proteínas Ativadoras de GTPase/genética , Proteínas Ativadoras de GTPase/metabolismo , Camundongos , Camundongos Knockout , Transporte Proteico/genética , Interferência de RNA , Ratos , Receptores de Mineralocorticoides/genética , Receptores de Vasopressinas/metabolismo , Vasopressinas/metabolismo
17.
Biochem Biophys Res Commun ; 424(2): 262-8, 2012 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-22750001

RESUMO

Oligodendrocytes (OLs) are myelin-forming glial cells in the central nervous system (CNS) and their dysfunction causes neuropathies such as demyelinating diseases. Proteolipid protein 1 (PLP1) is an oligodendrocyte myelin-rich tetraspan membrane protein and aberration of the plp1 gene is known to be responsible for dysmyelinating Pelizaeus-Merzbacher disease (PMD). Among previously identified gene alternations, multiplication of the plp1 gene causes increased expression of PLP1, resulting in a phenotype with severe dysmyelination in human and also rodent models. Yet little is known about the relationship between increased PLP1 expression and oligodendrocyte precursor cell (OPC) differentiation and the intracellular molecular mechanism. Here we show that expression of PLP1 in OPCs markedly inhibits their differentiation, and that this inhibitory effect is effectively improved by inhibition of extracellular signal-regulated kinase (ERK) activity. Furthermore, in cocultures with dorsal root ganglion (DRG) neurons, ERK inhibition also improves PLP1-induced dysmyelination. Thus, ERK inhibition helps to improve defective OPC differentiation induced by PLP1 expression, suggesting that molecules belonging to ERK signaling cascade may be new PMD therapeutic targets.


Assuntos
Diferenciação Celular , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteína Proteolipídica de Mielina/metabolismo , Oligodendroglia/citologia , Doença de Pelizaeus-Merzbacher/metabolismo , Células-Tronco/citologia , Técnicas de Cocultura , Gânglios Espinais/citologia , Humanos , Bainha de Mielina/metabolismo , Neurônios/citologia , Transdução de Sinais
18.
J Neurosci Res ; 90(4): 721-31, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22109349

RESUMO

The sorting nexins (SNXs) are a family of proteins functioning in diverse processes, including endocytosis, endosomal sorting, and endosomal signaling. Sorting nexin 12 (SNX12) is one of the SNXs family members; however, its function remains unknown. To clarify the function of SNX12, in this study, we first investigated the expression profiles in mice, particularly in the central nervous system (CNS), and then analyzed the functional role on neurite outgrowth. We found that SNX12 was widely expressed in the adult mouse CNS and that its expression level was higher in the cerebral cortex than in other examined regions. SNX12 expression was detected in the neurons but not the glial cells of the adult mouse cerebral cortex. In the fetal brain, SNX12 expression increased during the embryonic stage and gradually decreased after birth. Although the immunoreactivities of SNX12 were widespread in the cerebral cortical cells in the fetal brain, the immunopositive signals of SNX12 were more intense in the postmitotic neurons in the cortical plate than in the proliferating precursor cells in the ventricular zone, suggesting that SNX12 plays critical roles in the postmitotic neurons during cerebral cortical development. Furthermore, in mouse neuroblastoma and N1E-115 cells and rat primary cortical neurons, SNX12 expression was increased as neurite outgrowth progressed and the knockdown of SNX12 attenuated the outgrowth of neurites. These results suggest that SNX12 regulates neurite formation during cerebral cortical development.


Assuntos
Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Neurônios/metabolismo , Nexinas de Classificação/metabolismo , Animais , Bromodesoxiuridina , Linhagem Celular Tumoral , Embrião de Mamíferos , Proteína Glial Fibrilar Ácida/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neuritos/fisiologia , Neuroblastoma/patologia , Neurônios/citologia , Fosfopiruvato Hidratase/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Nexinas de Classificação/genética , Fatores de Tempo , Transfecção
19.
Clin Exp Nephrol ; 16(1): 30-4, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22038263

RESUMO

A prompt rise in blood pressure occurs when arginine-vasopressin is administered in quantities adequate to activate vascular V1a subtype vasopressin receptors. However, it has been controversial whether the endogenous vasopressin-V1a system contributes to the maintenance of basal blood pressure during normal development and aging. Mutant mice lacking the V1a receptor gene (V1a(-/-)) show significantly lower blood pressure compared to control mice, without a notable change in heart rate. In V1a(-/-) mice, arterial baroreceptor reflexes were attenuated due to malfunctioning baroreflex center, and the mice's circulating blood volume was significantly reduced. In line with this reduction in circulating blood volume, adrenocortical hormone release was attenuated; plasma aldosterone levels were reduced and adrenocorticotropic hormone-stimulated corticosteroid release was attenuated. In addition, V1a receptor expression was detected in macula densa cells of the kidneys, which may have facilitated renin production from the juxtaglomerular cells. Deletion of the V1a receptor appears to impact the renin-angiotensin-aldosterone system. Studies on V1a(-/-) mice revealed that non-vascular V1a receptors in the central nervous system and peripheral tissues play critical roles in the maintenance of blood pressure homeostasis.


Assuntos
Pressão Sanguínea/fisiologia , Homeostase/fisiologia , Receptores de Vasopressinas/fisiologia , Hormônio Adrenocorticotrópico , Animais , Antagonistas dos Receptores de Hormônios Antidiuréticos , Benzazepinas/uso terapêutico , Volume Sanguíneo , Insuficiência Cardíaca , Humanos , Hiponatremia/tratamento farmacológico , Camundongos , Receptores de Vasopressinas/deficiência , Sistema Renina-Angiotensina/fisiologia
20.
J Am Soc Nephrol ; 22(4): 673-80, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21415155

RESUMO

Both aldosterone and luminal vasopressin may contribute to the maintenance of acid-base homeostasis, but the functional relationship between these hormones is not well understood. The effects of luminal vasopressin likely result from its interaction with V1a receptors on the luminal membranes of intercalated cells in the collecting duct. Here, we found that mice lacking the V1a receptor exhibit type 4 renal tubular acidosis. The administration of the mineralocorticoid agonist fludrocortisone ameliorated the acidosis by restoring excretion of urinary ammonium via increased expression of Rhcg and H-K-ATPase and decreased expression of H-ATPase. In a cell line of intercalated cells established from transgenic rats expressing the mineralocorticoid and V1a receptors, but not V2 receptors, knockdown of the V1a receptor gene abrogated the effects of aldosterone on H-K-ATPase, Rhcg, and H-ATPase expression. These data suggest that defects in the vasopressin V1a receptor in intercalated cells can cause type 4 renal tubular acidosis and that the tubular effects of aldosterone depend on a functional V1a receptor in the intercalated cells.


Assuntos
Equilíbrio Ácido-Base/fisiologia , Aldosterona/metabolismo , Homeostase/fisiologia , Túbulos Renais Coletores/metabolismo , Receptores de Vasopressinas/metabolismo , Equilíbrio Ácido-Base/efeitos dos fármacos , Aldosterona/farmacologia , Animais , Proteínas de Transporte de Cátions/metabolismo , Linhagem Celular , Fludrocortisona/farmacologia , Homeostase/efeitos dos fármacos , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/efeitos dos fármacos , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Mineralocorticoides/agonistas , Modelos Animais , ATPases Translocadoras de Prótons/metabolismo , Interferência de RNA , Ratos , Ratos Transgênicos , Receptores de Vasopressinas/deficiência , Receptores de Vasopressinas/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa