Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Biol Chem ; 295(22): 7653-7668, 2020 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-32321761

RESUMO

The erythropoietin-producing human hepatocellular receptor EPH receptor B6 (EPHB6) is a receptor tyrosine kinase that has been shown previously to control catecholamine synthesis in the adrenal gland chromaffin cells (AGCCs) in a testosterone-dependent fashion. EPHB6 also has a role in regulating blood pressure, but several facets of this regulation remain unclear. Using amperometry recordings, we now found that catecholamine secretion by AGCCs is compromised in the absence of EPHB6. AGCCs from male knockout (KO) mice displayed reduced cortical F-actin disassembly, accompanied by decreased catecholamine secretion through exocytosis. This phenotype was not observed in AGCCs from female KO mice, suggesting that testosterone, but not estrogen, contributes to this phenotype. Of note, reverse signaling from EPHB6 to ephrin B1 (EFNB1) and a 7-amino acid-long segment in the EFNB1 intracellular tail were essential for the regulation of catecholamine secretion. Further downstream, the Ras homolog family member A (RHOA) and FYN proto-oncogene Src family tyrosine kinase (FYN)-proto-oncogene c-ABL-microtubule-associated monooxygenase calponin and LIM domain containing 1 (MICAL-1) pathways mediated the signaling from EFNB1 to the defective F-actin disassembly. We discuss the implications of EPHB6's effect on catecholamine exocytosis and secretion for blood pressure regulation.


Assuntos
Glândulas Suprarrenais/enzimologia , Catecolaminas/metabolismo , Células Cromafins/enzimologia , Exocitose , Receptor EphB6/metabolismo , Transdução de Sinais , Glândulas Suprarrenais/citologia , Animais , Catecolaminas/genética , Células Cromafins/citologia , Efrina-B1/genética , Efrina-B1/metabolismo , Feminino , Masculino , Camundongos , Camundongos Knockout , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Oxigenases de Função Mista/genética , Oxigenases de Função Mista/metabolismo , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-fyn/genética , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Receptor EphB6/genética , Caracteres Sexuais , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo
2.
J Biol Chem ; 294(17): 6871-6887, 2019 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-30824540

RESUMO

EPHB6 is a member of the erythropoietin-producing hepatocellular kinase (EPH) family and a receptor tyrosine kinase with a dead kinase domain. It is involved in blood pressure regulation and adrenal gland catecholamine (CAT) secretion, but several facets of EPHB6-mediated CAT regulation are unclear. In this study, using biochemical, quantitative RT-PCR, immunoblotting, and gene microarray assays, we found that EPHB6 up-regulates CAT biosynthesis in adrenal gland chromaffin cells (AGCCs). We observed that epinephrine content is reduced in the AGCCs from male Ephb6-KO mice, caused by decreased expression of tyrosine hydroxylase, the rate-limiting enzyme in CAT biosynthesis. We demonstrate that the signaling pathway from EPHB6 to tyrosine hydroxylase expression in AGCCs involves Rac family small GTPase 1 (RAC1), MAP kinase kinase 7 (MKK7), c-Jun N-terminal kinase (JNK), proto-oncogene c-Jun, activator protein 1 (AP1), and early growth response 1 (EGR1). On the other hand, signaling via extracellular signal-regulated kinase (ERK1/2), p38 mitogen-activated protein kinase, and ELK1, ETS transcription factor (ELK1) was not affected by EPHB6 deletion. We further report that EPHB6's effect on AGCCs was via reverse signaling through ephrin B1 and that EPHB6 acted in concert with the nongenomic effect of testosterone to control CAT biosynthesis. Our findings elucidate the mechanisms by which EPHB6 modulates CAT biosynthesis and identify potential therapeutic targets for diseases, such as hypertension, caused by dysfunctional CAT biosynthesis.


Assuntos
Glândulas Suprarrenais/enzimologia , Células Cromafins/enzimologia , Epinefrina/biossíntese , Receptor EphB6/fisiologia , Transcrição Gênica/fisiologia , Tirosina 3-Mono-Oxigenase/genética , Regulação para Cima/fisiologia , Glândulas Suprarrenais/citologia , Animais , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Elementos Facilitadores Genéticos , Epinefrina/metabolismo , Feminino , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor EphB6/genética , Transdução de Sinais , Testosterona/fisiologia , Tirosina 3-Mono-Oxigenase/metabolismo
3.
Brain Res ; 1604: 25-34, 2015 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-25662772

RESUMO

Hypotensive drugs have been used to identify central neurons that mediate compensatory baroreceptor reflex responses. Such drugs also increase blood glucose. Our aim was to identify the neurochemical phenotypes of sympathetic preganglionic neurons (SPN) and adrenal chromaffin cells activated following hydralazine (HDZ; 10mg/kg) administration in rats, and utilize this and SPN target organ destination to ascribe their function as cardiovascular or glucose regulating. Blood glucose was measured and adrenal chromaffin cell activation was assessed using c-Fos immunoreactivity (-ir) and phosphorylation of tyrosine hydroxylase, respectively. The activation and neurochemical phenotype of SPN innervating the adrenal glands and celiac ganglia were determined using the retrograde tracer cholera toxin B subunit, in combination with in situ hybridization and immunohistochemistry. Blood glucose was elevated at multiple time points following HDZ administration but little evidence of chromaffin cell activation was seen suggesting non-adrenal mechanisms contribute to the sustained hyperglycemia. 16±0.1% of T4-T11 SPN contained c-Fos and of these: 24.3±1.4% projected to adrenal glands and 29±5.5% projected to celiac ganglia with the rest innervating other targets. 62.8±1.4% of SPN innervating adrenal glands were activated and 29.9±3.3% expressed PPE mRNA whereas 53.2±8.6% of SPN innervating celiac ganglia were activated and 31.2±8.8% expressed PPE mRNA. CART-ir SPN innervating each target were also activated and did not co-express PPE mRNA. Neurochemical coding reveals that HDZ administration activates both PPE+SPN, whose activity increase glucose mobilization causing hyperglycemia, as well as CART+SPN whose activity drive vasomotor responses mediated by baroreceptor unloading to raise vascular tone and heart rate.


Assuntos
Anti-Hipertensivos/administração & dosagem , Fibras Autônomas Pré-Ganglionares/efeitos dos fármacos , Fenômenos Fisiológicos Cardiovasculares/efeitos dos fármacos , Gânglios Simpáticos/efeitos dos fármacos , Glucose/metabolismo , Hidralazina/farmacologia , Neurônios/efeitos dos fármacos , Medula Suprarrenal/inervação , Animais , Anti-Hipertensivos/farmacologia , Fibras Autônomas Pré-Ganglionares/metabolismo , Glicemia/metabolismo , Células Cromafins/efeitos dos fármacos , Células Cromafins/enzimologia , Células Cromafins/metabolismo , Gânglios Simpáticos/citologia , Gânglios Simpáticos/metabolismo , Masculino , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Sprague-Dawley
4.
J Cell Biol ; 203(2): 283-98, 2013 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-24165939

RESUMO

Several studies have suggested that the V0 domain of the vacuolar-type H(+)-adenosine triphosphatase (V-ATPase) is directly implicated in secretory vesicle exocytosis through a role in membrane fusion. We report in this paper that there was a rapid decrease in neurotransmitter release after acute photoinactivation of the V0 a1-I subunit in neuronal pairs. Likewise, inactivation of the V0 a1-I subunit in chromaffin cells resulted in a decreased frequency and prolonged kinetics of amperometric spikes induced by depolarization, with shortening of the fusion pore open time. Dissipation of the granular pH gradient was associated with an inhibition of exocytosis and correlated with the V1-V0 association status in secretory granules. We thus conclude that V0 serves as a sensor of intragranular pH that controls exocytosis and synaptic transmission via the reversible dissociation of V1 at acidic pH. Hence, the V-ATPase membrane domain would allow the exocytotic machinery to discriminate fully loaded and acidified vesicles from vesicles undergoing neurotransmitter reloading.


Assuntos
Exocitose , Neurônios/enzimologia , Vesículas Secretórias/enzimologia , Transmissão Sináptica , Vesículas Sinápticas/enzimologia , ATPases Vacuolares Próton-Translocadoras/metabolismo , Animais , Catecolaminas/metabolismo , Bovinos , Células Cromafins/enzimologia , Células Cromafins/metabolismo , Exocitose/efeitos dos fármacos , Exocitose/efeitos da radiação , Concentração de Íons de Hidrogênio , Cinética , Luz , Fusão de Membrana , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/efeitos da radiação , Células PC12 , Estrutura Terciária de Proteína , Interferência de RNA , Ratos , Proteínas Recombinantes de Fusão/metabolismo , Vesículas Secretórias/efeitos dos fármacos , Vesículas Secretórias/metabolismo , Vesículas Secretórias/efeitos da radiação , Potenciais Sinápticos , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/efeitos da radiação , Vesículas Sinápticas/efeitos dos fármacos , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/efeitos da radiação , Transfecção , ATPases Vacuolares Próton-Translocadoras/genética
5.
J Neurosci ; 33(8): 3545-56, 2013 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-23426682

RESUMO

Calcium-regulated exocytosis in neuroendocrine cells and neurons is accompanied by the redistribution of phosphatidylserine (PS) to the extracellular space, leading to a disruption of plasma membrane asymmetry. How and why outward translocation of PS occurs during secretion are currently unknown. Immunogold labeling on plasma membrane sheets coupled with hierarchical clustering analysis demonstrate that PS translocation occurs at the vicinity of the secretory granule fusion sites. We found that altering the function of the phospholipid scramblase-1 (PLSCR-1) by expressing a PLSCR-1 calcium-insensitive mutant or by using chromaffin cells from PLSCR-1⁻/⁻ mice prevents outward translocation of PS in cells stimulated for exocytosis. Remarkably, whereas transmitter release was not affected, secretory granule membrane recapture after exocytosis was impaired, indicating that PLSCR-1 is required for compensatory endocytosis but not for exocytosis. Our results provide the first evidence for a role of specific lipid reorganization and calcium-dependent PLSCR-1 activity in neuroendocrine compensatory endocytosis.


Assuntos
Células Cromafins/metabolismo , Endocitose/fisiologia , Células Neuroendócrinas/metabolismo , Fosfatidilserinas/metabolismo , Proteínas de Transferência de Fosfolipídeos/metabolismo , Animais , Transporte Biológico Ativo/fisiologia , Bovinos , Membrana Celular/metabolismo , Células Cromafins/enzimologia , Exocitose/fisiologia , Feminino , Metabolismo dos Lipídeos/fisiologia , Masculino , Camundongos , Camundongos Transgênicos , Células Neuroendócrinas/enzimologia , Células PC12 , Ratos
6.
Mol Pharmacol ; 80(2): 304-13, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21540292

RESUMO

Treatment of cultured bovine adrenal chromaffin cells with the catecholamine transport blocker reserpine was shown previously to increase enkephalin levels severalfold. To explore the biochemical mechanism of this effect, we examined the effect of reserpine treatment on the activities of three different peptide precursor processing enzymes: carboxypeptidase E (CPE) and the prohormone convertases (PCs) PC1/3 and PC2. Reserpine treatment increased both CPE and PC activity in extracts of cultured chromaffin cells; total protein levels were unaltered for any enzyme. Further analysis showed that the increase in CPE activity was due to an elevated V(max), with no change in the K(m) for substrate hydrolysis or the levels of CPE mRNA. Reserpine activation of endogenous processing enzymes was also observed in extracts prepared from PC12 cells stably expressing PC1/3 or PC2. In vitro experiments using purified enzymes showed that catecholamines inhibited CPE, PC1/3, and PC2, with dopamine quinone the most potent inhibitor (IC(50) values of ∼50-500 µM); dopamine, norepinephrine, and epinephrine exhibited inhibition in the micromolar range. The inhibition of purified CPE with catecholamines was time-dependent and, for dopamine quinone, dilution-independent, suggesting covalent modification of the protein by the catecholamine. Because the catecholamine concentrations found to be inhibitory to PC1/3, PC2, and CPE are well within the physiological range found in chromaffin granules, we conclude that catecholaminergic transmitter systems have the potential to exert considerable dynamic influence over peptidergic transmitter synthesis by altering the activity of peptide processing enzymes.


Assuntos
Carboxipeptidase H/fisiologia , Catecolaminas/fisiologia , Células Cromafins/enzimologia , Neuropeptídeos/metabolismo , Pró-Proteína Convertase 1/fisiologia , Pró-Proteína Convertase 2/fisiologia , Processamento de Proteína Pós-Traducional/fisiologia , Animais , Carboxipeptidase H/antagonistas & inibidores , Catecolaminas/farmacologia , Bovinos , Células Cultivadas , Células Cromafins/efeitos dos fármacos , Células PC12 , Pró-Proteína Convertase 1/antagonistas & inibidores , Pró-Proteína Convertase 2/antagonistas & inibidores , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Ratos , Reserpina/farmacologia
7.
Cell Mol Neurobiol ; 30(8): 1441-9, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21107678

RESUMO

Pituitary adenylate cyclase-activating polypeptide (PACAP) is a co-transmitter with acetylcholine at the adrenomedullary synapse, mediating sustained hormone secretion and regulation of cellular plasticity in response to stress at the level of gene transcription. Here we have extended our investigation of PACAP-regulated neuroendocrine cell-specific genes from PC12 cells to PC12 cells expressing physiological levels of the PAC1hop receptor found on chromaffin cells in vivo. PACAP induces in these PC12_bPAC1hop cells an additional cohort of genes, compared to PC12 cells, enriched in informational molecules including cytokines, neuropeptides, and growth factors. Using two newly developed microarray platforms for expressed bovine transcripts, we further examined PACAP-induced genes in bovine chromaffin cells during a period of exposure (6 h) corresponding to a period of prolonged metabolic or psychogenic stress in vivo during which PACAP is released from the splanchnic nerve onto chromaffin cells. As in PC12_bPAC1hop cells, PACAP induced in bovine chromaffin cells a cohort of genes encoding secretory proteins, identified by tiling for cellular localization using Ingenuity Pathway Analysis, which were highly enriched in informational molecules (secreted proteins acting at extracellular receptors). These included cytokines, growth factors and hormones, as well as converting enzymes, or protease inhibitors modulating converting enzyme function. Several neuropeptide prohormone transcripts not previously shown to be PACAP-regulated in chromaffin cells, such as thyrotropin-releasing hormone, and tachykinin precursor 1, were identified. Identification of this cohort of informational molecule-encoding transcripts suggests a wider, more integrative role for PACAP as a co-transmitter specific to stress transduction in the adrenal medulla.


Assuntos
Células Cromafins/metabolismo , Citocinas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neurotransmissores/farmacologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/farmacologia , Estresse Fisiológico/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Animais , Bovinos , Células Cromafins/efeitos dos fármacos , Células Cromafins/enzimologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Citocinas/genética , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/genética , Células PC12 , Ratos
8.
Horm Res Paediatr ; 73(2): 135-9, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20190551

RESUMO

BACKGROUND/AIMS: To describe the management of a subject with multiple chromaffin tumours found to have a novel succinate dehydrogenase D (SDHD) mutation. CASE: A 15-year-old boy with marked hypertension was found to have elevated urinary catecholamines and initial imaging thought to represent bilateral adrenal phaeochromocytomas. An adrenal venous catheter was required to clarify a right adrenal phaeochromocytoma and a left abdominal paraganglioma, distinct from the left adrenal gland. Excision of these tumours, with preservation of the left adrenal gland, provided a cure for this subject without the need for lifelong steroid replacement. Genetic analysis revealed a novel SDHD mutation (c. 169 + 1 G>A) which was shown to result in loss of the 5' splice site and exclusion of exon 2 during splicing. This suggests the likely pathogenicity of this mutation. Disease surveillance in this subject and genetic screening of first degree relatives is ongoing. CONCLUSIONS: Genetic testing should be considered in all subjects presenting with a chromaffin tumour. In certain circumstances an adrenal venous sampling catheter for catecholamines may clarify diagnostic uncertainty. The complex management issues raised in the care of these subjects requires the involvement of a multidisciplinary team with the relevant expertise.


Assuntos
Neoplasias Abdominais/genética , Neoplasias das Glândulas Suprarrenais/genética , Catecolaminas/genética , Neoplasias Primárias Múltiplas/genética , Paraganglioma/genética , Feocromocitoma/genética , Succinato Desidrogenase/genética , Neoplasias Abdominais/diagnóstico , Neoplasias Abdominais/fisiopatologia , Neoplasias Abdominais/cirurgia , Adolescente , Neoplasias das Glândulas Suprarrenais/diagnóstico , Neoplasias das Glândulas Suprarrenais/fisiopatologia , Neoplasias das Glândulas Suprarrenais/cirurgia , Pressão Sanguínea , Catecolaminas/sangue , Catecolaminas/urina , Cateterismo/métodos , Células Cromafins/enzimologia , Células Cromafins/patologia , Éxons , Humanos , Masculino , Mutação , Neoplasias Primárias Múltiplas/diagnóstico , Neoplasias Primárias Múltiplas/fisiopatologia , Neoplasias Primárias Múltiplas/cirurgia , Paraganglioma/diagnóstico , Paraganglioma/fisiopatologia , Paraganglioma/cirurgia , Feocromocitoma/diagnóstico , Feocromocitoma/fisiopatologia , Feocromocitoma/cirurgia , Sítios de Splice de RNA , Splicing de RNA
9.
J Neuroendocrinol ; 22(2): 83-91, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20025629

RESUMO

Urotensin II (U-II), initially identified as a cyclic peptide from fish urophysis, acts both as a strong vasoconstrictor and vasodilator in the vasculature via its receptor, G-protein coupled receptor 14. In addition, U-II and its receptor are co-expressed in the adrenal medulla, as well as in human pheochromocytomas, suggesting that this peptide may have some function in chromaffin cells. However, the precise role of U-II in these cells is unknown. In the present study, we initially demonstrate that U-II and its receptors mRNA are co-expressed in the rat pheochromocytoma cell line PC12. Moreover, U-II has not effect on tyrosine hydroxylase (TH), the rate-limiting enzyme involved in the biosynthesis of catecholamine, in terms of enzyme activity or at the mRNA level. However, U-II does induce an increase in the phosphorylation of TH specifically at Ser31 without affecting phosphorylation at the two other sites (Ser19 and Ser40). U-II also markedly activates extracellular signal-regulated kinases (ERKs) and p38, but not Jun N-terminal kinase. Blockade of the epidermal growth factor (EGF) receptor by AG1478 significantly reduces activation of ERK, suggesting that EGF receptor transactivation could act upstream of the ERK pathway in PC12 cells. Furthermore, U-II significantly increases dopamine secretion from PC12 cells. Finally, we show that U-II induced significant DNA synthesis in a ERKs and P38 mitogen-activated protein kinase-dependent manner. The results obtained indicate that U-II may exert its effects as a neuromodulator in chromaffin cells.


Assuntos
Células Cromafins/metabolismo , Urotensinas/metabolismo , Sequência de Aminoácidos , Animais , Proliferação de Células , Células Cromafins/efeitos dos fármacos , Células Cromafins/enzimologia , DNA/biossíntese , DNA/metabolismo , Dopamina/metabolismo , Inibidores Enzimáticos/farmacologia , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Células PC12 , Fosforilação , Quinazolinas , RNA Mensageiro/metabolismo , Ratos , Receptores Acoplados a Proteínas G/metabolismo , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo , Tirfostinas/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
10.
Am J Physiol Regul Integr Comp Physiol ; 296(1): R133-40, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18987290

RESUMO

We tested the hypothesis that endogenously produced hydrogen sulfide (H(2)S) can potentially contribute to the adrenergic stress response in rainbow trout by initiating catecholamine secretion from chromaffin cells. During acute hypoxia (water Po(2) = 35 mmHg), plasma H(2)S levels were significantly elevated concurrently with a rise in circulating catecholamine concentrations. Tissues enriched with chromaffin cells (posterior cardinal vein and anterior kidney) produced H(2)S in vitro when incubated with l-cysteine. In both tissues, the production of H(2)S was eliminated by adding the cystathionine beta-synthase inhibitor, aminooxyacetate. Cystathionine beta-synthase and cystathionine gamma-lyase were cloned and sequenced and the results of real-time PCR demonstrated that with the exception of white muscle, mRNA for both enzymes was broadly distributed within the tissues that were examined. Electrical field stimulation of an in situ saline-perfused posterior cardinal vein preparation caused the appearance of H(2)S and catecholamines in the outflowing perfusate. Perfusion with the cholinergic receptor agonist carbachol (1 x 10(-6) M) or depolarizing levels of KCl (1 x 10(-2) M) caused secretion of catecholamines without altering H(2)S output, suggesting that neuronal excitation is required for H(2)S release. Addition of H(2)S (at concentrations exceeding 5 x 10(-7) M) to the perfusion fluid resulted in a marked stimulation of catecholamine secretion that was not observed when Ca(2+)-free perfusate was used. These data, together with the finding that H(2)S-induced catecholamine secretion was unaltered by the nicotinic receptor blocker hexamethonium, suggest that H(2)S is able to directly elicit catecholamine secretion via membrane depolarization followed by Ca(2+)-mediated exocytosis.


Assuntos
Células Cromafins/metabolismo , Epinefrina/metabolismo , Sulfeto de Hidrogênio/metabolismo , Hipóxia/metabolismo , Norepinefrina/metabolismo , Oncorhynchus mykiss/metabolismo , Estresse Fisiológico , Animais , Sinalização do Cálcio , Agonistas Colinérgicos/farmacologia , Células Cromafins/efeitos dos fármacos , Células Cromafins/enzimologia , Clonagem Molecular , Cistationina beta-Sintase/antagonistas & inibidores , Cistationina beta-Sintase/genética , Cistationina beta-Sintase/metabolismo , Cistationina gama-Liase/genética , Cistationina gama-Liase/metabolismo , Cisteína/metabolismo , Estimulação Elétrica , Inibidores Enzimáticos/farmacologia , Epinefrina/sangue , Exocitose , Feminino , Proteínas de Peixes/antagonistas & inibidores , Proteínas de Peixes/genética , Proteínas de Peixes/metabolismo , Regulação Enzimológica da Expressão Gênica , Sulfeto de Hidrogênio/sangue , Hipóxia/sangue , Hipóxia/fisiopatologia , Masculino , Potenciais da Membrana , Antagonistas Nicotínicos/farmacologia , Norepinefrina/sangue , Oncorhynchus mykiss/sangue , Perfusão , Filogenia , RNA Mensageiro/metabolismo , Fatores de Tempo
11.
Circulation ; 118(3): 247-57, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18591442

RESUMO

BACKGROUND: Chromogranin A (CHGA) triggers catecholamine secretory granule biogenesis, and its catestatin fragment inhibits catecholamine release. We approached catestatin heritability using twin pairs, coupled with genome-wide linkage, in a series of twin and sibling pairs from 2 continents. METHODS AND RESULTS: Hypertensive patients had elevated CHGA coupled with reduction in catestatin, suggesting diminished conversion of precursor to catestatin. Heritability for catestatin in twins was 44% to 60%. Six hundred fifteen nuclear families yielded 870 sib pairs for linkage, with significant logarithm of odds peaks on chromosomes 4p, 4q, and 17q. Because acidification of catecholamine secretory vesicles determines CHGA trafficking and processing to catestatin, we genotyped at positional candidate ATP6N1, bracketed by peak linkage markers on chromosome 17q, encoding a subunit of vesicular H(+)-translocating ATPase. The minor allele diminished CHGA secretion and processing to catestatin. The ATP6N1 variant also influenced blood pressure in 1178 individuals with the most extreme blood pressure values in the population. In chromaffin cells, inhibition of H(+)-ATPase diverted CHGA from regulated to constitutive secretory pathways. CONCLUSIONS: We established heritability of catestatin in twins from 2 continents. Linkage identified 3 regions contributing to catestatin, likely novel determinants of sympathochromaffin exocytosis. At 1 such positional candidate (ATP6N1), variation influenced CHGA secretion and processing to catestatin, confirming the mechanism of a novel trans-QTL for sympathochromaffin activity and blood pressure.


Assuntos
Cromogranina A/genética , Ligação Genética , Genoma Humano , Fragmentos de Peptídeos/genética , Característica Quantitativa Herdável , Gêmeos/genética , Alelos , Austrália , Pressão Sanguínea/genética , Células Cromafins/enzimologia , Células Cromafins/metabolismo , Cromogranina A/sangue , Cromogranina A/metabolismo , Mapeamento Cromossômico , Cromossomos Humanos Par 17 , Meio Ambiente , Exocitose , Feminino , Variação Genética , Humanos , Hipertensão/genética , Masculino , Pessoa de Meia-Idade , Fragmentos de Peptídeos/sangue , ATPases Translocadoras de Prótons/metabolismo , Irmãos , Estados Unidos
12.
Proc Natl Acad Sci U S A ; 105(24): 8434-9, 2008 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-18550821

RESUMO

Exocytosis of neurotransmitters and hormones occurs through the fusion of secretory vesicles with the plasma membrane. This highly regulated process involves key proteins, such as SNAREs, and specific lipids at the site of membrane fusion. Phospholipase D (PLD) has recently emerged as a promoter of membrane fusion in various exocytotic events potentially by providing fusogenic cone-shaped phosphatidic acid. We show here that PLD1 is regulated by ribosomal S6 kinase 2 (RSK2)-dependent phosphorylation. RSK2 is activated by a high K(+)-induced rise in cytosolic calcium. Expression of inactive RSK2 mutants or selective knockdown of endogenous RSK2 dramatically affects the different kinetic components of the exocytotic response in chromaffin cells. RSK2 physically interacts with and stimulates PLD activity through the phosphorylation of Thr-147 in the PLD1 amino-terminal phox homology domain. Expression of PLD1 phosphomimetic mutants fully restores secretion in cells depleted of RSK2, suggesting that RSK2 is a critical upstream signaling element in the activation of PLD1 to produce the lipids required for exocytosis. We propose that PLD-related defects in neuronal and endocrine activities could contribute to the effect observed after the loss-of-function mutations in Rsk2 that lead to Coffin-Lowry syndrome, an X-linked form of growth and mental retardation.


Assuntos
Cálcio/metabolismo , Células Cromafins/metabolismo , Síndrome de Coffin-Lowry/enzimologia , Exocitose , Fosfolipase D/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Animais , Células Cromafins/enzimologia , Síndrome de Coffin-Lowry/genética , Exocitose/genética , Células PC12 , Ácidos Fosfatídicos/metabolismo , Fosforilação , Ratos , Proteínas Quinases S6 Ribossômicas 90-kDa/genética
13.
J Neurochem ; 106(1): 384-91, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18410501

RESUMO

Neuropeptide Y (NPY) functions as a peptide neurotransmitter and as a neuroendocrine hormone. The active NPY peptide is generated in secretory vesicles by proteolytic processing of proNPY. Novel findings from this study show that cathepsin L participates as a key proteolytic enzyme for NPY production in secretory vesicles. Notably, NPY levels in cathepsin L knockout (KO) mice were substantially reduced in brain and adrenal medulla by 80% and 90%, respectively. Participation of cathepsin L in producing NPY predicts their colocalization in secretory vesicles, a primary site of NPY production. Indeed, cathepsin L was colocalized with NPY in brain cortical neurons and in chromaffin cells of adrenal medulla, demonstrated by immunofluorescence confocal microscopy. Immunoelectron microscopy confirmed the localization of cathepsin L with NPY in regulated secretory vesicles of chromaffin cells. Functional studies showed that coexpression of proNPY with cathepsin L in neuroendocrine PC12 cells resulted in increased production of NPY. Furthermore, in vitro processing indicated cathepsin L processing of proNPY at paired basic residues. These findings demonstrate a role for cathepsin L in the production of NPY from its proNPY precursor. These studies illustrate the novel biological role of cathepsin L in the production of NPY, a peptide neurotransmitter, and neuroendocrine hormone.


Assuntos
Medula Suprarrenal/enzimologia , Encéfalo/enzimologia , Catepsinas/genética , Células Cromafins/enzimologia , Cisteína Endopeptidases/genética , Neuropeptídeo Y/biossíntese , Vesículas Secretórias/enzimologia , Medula Suprarrenal/ultraestrutura , Sequência de Aminoácidos/fisiologia , Animais , Encéfalo/ultraestrutura , Catepsina L , Catepsinas/fisiologia , Células Cultivadas , Células Cromafins/ultraestrutura , Cisteína Endopeptidases/fisiologia , Imunofluorescência , Regulação Enzimológica da Expressão Gênica/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Confocal , Microscopia Imunoeletrônica , Neuropeptídeo Y/metabolismo , Sistemas Neurossecretores/enzimologia , Sistemas Neurossecretores/ultraestrutura , Células PC12 , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/metabolismo , Ratos , Vesículas Secretórias/metabolismo , Vesículas Secretórias/ultraestrutura
14.
Am J Physiol Cell Physiol ; 293(5): C1509-22, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17686997

RESUMO

Neurons and neuroendocrine cells must retrieve plasma membrane excess and refill vesicle pools depleted by exocytosis. To perform these tasks cells can use different endocytosis/recycling mechanisms whose selection will impact on vesicle recycling time and secretion performance. We used FM1-43 to evaluate in the same experiment exocytosis, endocytosis, and recovery of releasable vesicles on mouse chromaffin cells. Various exocytosis levels were induced by a variety of stimuli, and we discriminated the resultant endocytosis-recycling responses according to their ability to rapidly generate releasable vesicles. Exocytosis of < or =20% of plasma membrane (provoked by nicotine/acetylcholine) was followed by total recovery of releasable vesicles. If a stronger stimulus (50 mM K(+) and 2 mM Ca(2+)) provoking intense exocytosis (51 +/- 7%) was applied, endocytosis still retrieved all the fused membrane, but only a fraction (19 +/- 2%) was releasable by a second stimulus. Using ADVASEP-7 or bromophenol blue to quickly eliminate fluorescence from noninternalized FM1-43, we determined that this fraction became releasable in <2 min. The remaining nonreleasable fraction was distributed mainly as fluorescent spots ( approximately 0.7 microm) selectively labeled by 40- to 70-kDa dextrans and was suppressed by a phosphatidylinositol-3-phosphate kinase inhibitor, suggesting that it had been formed by a bulk retrieval mechanism. We concluded that chromaffin cells can rapidly recycle significant fractions of their total vesicle population, and that this pathway prevails when cholinergic agonists are used as secretagogues. When exocytosis exceeded approximately 20% of plasma membrane, an additional mechanism was activated, which was unable to produce secretory vesicles in our experimental time frame but appeared crucial to maintaining membrane surface homeostasis under extreme conditions.


Assuntos
Glândulas Suprarrenais/metabolismo , Células Cromafins/metabolismo , Endocitose , Endossomos/metabolismo , Exocitose , Vesículas Transportadoras/metabolismo , Acetilcolina/farmacologia , Glândulas Suprarrenais/citologia , Glândulas Suprarrenais/efeitos dos fármacos , Animais , Azul de Bromofenol/química , Cálcio/metabolismo , Células Cultivadas , Agonistas Colinérgicos/farmacologia , Células Cromafins/efeitos dos fármacos , Células Cromafins/enzimologia , Ciclodextrinas/química , Endocitose/efeitos dos fármacos , Endossomos/efeitos dos fármacos , Exocitose/efeitos dos fármacos , Corantes Fluorescentes/química , Homeostase , Fusão de Membrana , Camundongos , Nicotina/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Potássio/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Compostos de Piridínio/química , Compostos de Amônio Quaternário/química , Coloração e Rotulagem/métodos , Fatores de Tempo , Vesículas Transportadoras/efeitos dos fármacos
15.
Toxicol Lett ; 168(3): 286-91, 2007 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-17184936

RESUMO

Chromaffin cells in culture show high neuropathy target esterase (NTE) activity. It is well known that inhibition and specific modification of NTE by some organophosphorus (OPs) compounds induces a neurodegenerative neuropathy. It has been suggested that NTE is responsible for phosphatidylcholine homeostasis, although its role in neuropathy induction remains unclear. The cDNA of human NTE (4.4kbp) was inserted into an adenoviral vector. Bovine chromaffin cells cultured at 50,000 cells/well were incubated with the vector for 2h and after removing the volume of infection, cells were maintained in the incubator. After 24h, NTE activity was 6.8+/-0.5mU/10(6) cells in untreated cells and 14.8+/-1.5mU/10(6) cells, 19.3+/-2.9mU/10(6) cells, 24.8+/-0.9mU/10(6) cells and 30.9+/-1.0mU/10(6) cells in cells incubated with 2, 4, 8 and 16microl of vector, respectively. After 60min of inhibition with mipafox increased concentrations, the calculated I(50) (60min) values were 5.5, 6.2 and 6.6microM for cells infected with 0, 2 and 10microl of vector preparation. We confirm that the adenoviral vector containing the human NTE gene is active in bovine chromaffin cells in culture and that the NTE activity expressed by the vector shows the same inhibition pattern by the neuropathic OP mipafox as the NTE activity of bovine chromaffin cells and cells remained viable after the high NTE activity expression.


Assuntos
Adenoviridae/genética , Hidrolases de Éster Carboxílico/metabolismo , Células Cromafins/enzimologia , Animais , Hidrolases de Éster Carboxílico/antagonistas & inibidores , Bovinos , Células Cultivadas , Vetores Genéticos , Humanos , Isoflurofato/análogos & derivados , Isoflurofato/toxicidade , Paraoxon/toxicidade , Transfecção , Valeratos/metabolismo
16.
J Biol Chem ; 281(26): 17644-51, 2006 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-16644734

RESUMO

Tyrosine hydroxylase (TH) is the rate-limiting enzyme in the biosynthesis of the catecholamines dopamine, noradrenaline, and adrenaline. In response to short term stimuli TH activity is primarily controlled by phosphorylation of serine 40. We have previously shown that phosphorylation of serine 19 in TH can indirectly activate TH via a hierarchical mechanism by increasing the rate of phosphorylation of serine 40. Here we show that phosphorylation of serine 31 in rat TH increases the rate of serine 40 phosphorylation 9-fold in vitro. Phosphorylation of serine 31 in intact bovine chromaffin cells potentiated the forskolin-induced increase in serine 40 phosphorylation and TH activity more than 2-fold. Humans are unique in that they contain four TH isoforms but to date no significant differences have been shown in the regulation of these isoforms. Phosphorylation of the human TH isoform 1 at serine 31 by extracellular signal-regulated protein kinase (ERK) also produced a 9-fold increase in the rate of phosphorylation of serine 40, whereas little effect was seen in the TH isoforms 3 and 4. ERK did not phosphorylate human TH isoform 2. The effect of serine 19 phosphorylation on serine 40 (44 in TH2) phosphorylation is stronger in TH2 than in TH1. Thus hierarchical phosphorylation provides a mechanism whereby the two major human TH isoforms (1 and 2) can be differentially regulated with only isoform 1 responding to the ERK pathway, whereas isoform 2 is more sensitive to calcium-mediated events.


Assuntos
Células Cromafins/enzimologia , Isoenzimas/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo , Sequência de Aminoácidos , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Catecolaminas/biossíntese , Bovinos , Colforsina/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Isoenzimas/genética , Dados de Sequência Molecular , Fosforilação , Ratos , Serina/metabolismo , Tirosina 3-Mono-Oxigenase/genética
17.
Biol Chem ; 386(9): 931-40, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16164418

RESUMO

The regulated secretory pathway of neurons is the major source of extracellular A beta that accumulates in Alzheimer's disease (AD). Extracellular A beta secreted from that pathway is generated by beta-secretase processing of amyloid precursor protein (APP). Previously, cysteine protease activity was demonstrated as the major beta-secretase activity in regulated secretory vesicles of neuronal chromaffin cells. In this study, the representative cysteine protease activity in these secretory vesicles was purified and identified as cathepsin B by peptide sequencing. Immunoelectron microscopy demonstrated colocalization of cathepsin B with A beta in these vesicles. The selective cathepsin B inhibitor, CA074, blocked the conversion of endogenous APP to A beta in isolated regulated secretory vesicles. In chromaffin cells, CA074Me (a cell permeable form of CA074) reduced by about 50% the extracellular A beta released by the regulated secretory pathway, but CA074Me had no effect on A beta released by the constitutive pathway. Furthermore, CA074Me inhibited processing of APP into the COOH-terminal beta-secretase-like cleavage product. These results provide evidence for cathepsin B as a candidate beta-secretase in regulated secretory vesicles of neuronal chromaffin cells. These findings implicate cathepsin B as beta-secretase in the regulated secretory pathway of brain neurons, suggesting that inhibitors of cathepsin B may be considered as therapeutic agents to reduce A beta in AD.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/biossíntese , Catepsina B/antagonistas & inibidores , Células Cromafins/metabolismo , Endopeptidases/metabolismo , Neurônios/metabolismo , Vesículas Secretórias/metabolismo , Doença de Alzheimer/enzimologia , Sequência de Aminoácidos , Secretases da Proteína Precursora do Amiloide , Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/metabolismo , Animais , Catepsina B/isolamento & purificação , Catepsina B/metabolismo , Bovinos , Células Cromafins/efeitos dos fármacos , Células Cromafins/enzimologia , Cisteína Endopeptidases/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , Dados de Sequência Molecular , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Vesículas Secretórias/enzimologia
18.
Cell Tissue Res ; 322(3): 443-53, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16047163

RESUMO

Expression of the noradrenaline transporter (NAT) was examined in normal human adrenal medulla and phaeochromocytoma by using immunohistochemistry and confocal microscopy. The enzymes tyrosine hydroxylase (TH) and phenylethanolamine N-methyltransferase (PNMT) were used as catecholamine biosynthetic markers and chromogranin A (CGA) as a marker for secretory granules. Catecholamine content was measured by using high performance liquid chromatography (HPLC). In normal human adrenal medulla (n=5), all chromaffin cells demonstrated strong TH, PNMT and NAT immunoreactivity. NAT was co-localized with PNMT and was located within the cytoplasm with a punctate appearance. Human phaeochromocytomas demonstrated strong TH expression (n=20 samples tested) but variable NAT and PNMT expression (n=24). NAT immunoreactivity ranged from absent (n=3) to weak (n=10) and strong (n=11) and, in some cases, occupied an apparent nuclear location. Unlike the expression seen in normal human adrenal medullary tissue, NAT expression was not consistently co-localized with PNMT. PNMT also showed highly variable expression that was poorly correlated with tumour adrenaline content. Immunoreactivity for CGA was colocalized with NAT within the cytoplasm of normal human chromaffin cells (n=4). This co-localization was not consistent in phaeochromocytoma tumour cells (n=7). The altered pattern of expression for both NAT and PNMT in phaeochromocytoma indicates a significant disruption in the regulation and possibly in the function of these proteins in adrenal medullary tumours.


Assuntos
Medula Suprarrenal/metabolismo , Células Cromafins/metabolismo , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/biossíntese , Feniletanolamina N-Metiltransferase/biossíntese , Feocromocitoma/metabolismo , Medula Suprarrenal/enzimologia , Catecolaminas/biossíntese , Células Cromafins/enzimologia , Humanos , Imuno-Histoquímica , Microscopia Confocal , Feniletanolamina N-Metiltransferase/metabolismo , Feocromocitoma/enzimologia
19.
J Physiol ; 564(Pt 1): 161-72, 2005 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15695243

RESUMO

Adrenal medullary (AM) cells are exposed to high concentrations of cortical hormones, one of which is a ouabain-like substance. Thus, the effects of ouabain on catecholamine secretion and distribution of Na+,K+-ATPase alpha and beta subunits in rat and guinea-pig AM cells were examined using amperometry and immunological techniques. While exposure to 1 microm ouabain did not have a marked effect on resting secretion, it induced an increase in secretion due to mobilization of Ca2+ ions that were stored during a 4 min interval between muscarine applications. Immunocytochemistry revealed that Na+,K+-ATPase alpha1 subunit-like and beta3 subunit-like immunoreactive (IR) materials were distributed ubiquitously at the cell periphery, whereas alpha2- and beta2-like IR materials were present in restricted parts of the cell periphery. The alpha1 and alpha2 subunits were mainly immunoprecipitated from AM preparations by anti-beta3 and anti-beta2 antisera, respectively. Peripheral BODIPY-FL-InsP3 binding sites were localized below membrane domains with alpha2- and beta2-like IR materials. The results indicate that in AM cells, alpha1beta3 isozymes of Na+,K+-ATPase were present ubiquitously in the plasma membrane, while alpha2beta2 isozymes were in the membrane domain closely associated with peripheral Ca2+ store sites. This close association of the alpha2beta2 isozyme with peripheral Ca2+ store sites may account for the facilitation of mobilization-dependent secretion in the presence of 1 microm ouabain.


Assuntos
Medula Suprarrenal/enzimologia , Células Cromafins/enzimologia , Subunidades Proteicas/química , Subunidades Proteicas/fisiologia , ATPase Trocadora de Sódio-Potássio/química , ATPase Trocadora de Sódio-Potássio/fisiologia , Medula Suprarrenal/efeitos dos fármacos , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Células Cromafins/efeitos dos fármacos , Cobaias , Isoenzimas/química , Isoenzimas/fisiologia , Ouabaína/farmacologia , Ratos
20.
Eur J Pharmacol ; 485(1-3): 137-40, 2004 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-14757133

RESUMO

Bovine chromaffin cells contain an ecto-ATPase (K(m)=1.57 +/- 0.27 x 10(-4) M) which can hydrolyze ATP present in the culture media. ARL 67156 is a competitive inhibitor of this ATPase (K(i)=2.55 +/- 1.36 x 10(-7) M). A small increase in potency (threefold) is seen when ARL 67156 is included during measurement of ATP-stimulated inositol phosphate formation. ARL 67156 also acts on chromaffin cell P2Y receptors to increase inositol phosphate formation (EC(50)=4.9 x 10(-5) M). It is useful as an ecto-ATPase inhibitor in studies with bovine chromaffin cells since it exhibits a 300-fold selectivity for the ecto-ATPase versus the P2Y receptor.


Assuntos
Adenosina Trifosfatases/antagonistas & inibidores , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/farmacologia , Células Cromafins/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Adenosina Trifosfatases/metabolismo , Animais , Bovinos , Células Cultivadas , Células Cromafins/enzimologia , Relação Dose-Resposta a Droga
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA