Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Sci Adv ; 6(14): eaay9572, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32270040

RESUMO

The endoplasmic reticulum (ER) is a highly dynamic network of membranes. Here, we combine live-cell microscopy with in situ cryo-electron tomography to directly visualize ER dynamics in several secretory cell types including pancreatic ß-cells and neurons under near-native conditions. Using these imaging approaches, we identify a novel, mobile form of ER, ribosome-associated vesicles (RAVs), found primarily in the cell periphery, which is conserved across different cell types and species. We show that RAVs exist as distinct, highly dynamic structures separate from the intact ER reticular architecture that interact with mitochondria via direct intermembrane contacts. These findings describe a new ER subcompartment within cells.


Assuntos
Vesículas Citoplasmáticas/metabolismo , Retículo Endoplasmático/metabolismo , Ribossomos/metabolismo , Animais , Transporte Biológico , Microscopia Crioeletrônica , Vesículas Citoplasmáticas/ultraestrutura , Retículo Endoplasmático/ultraestrutura , Complexo de Golgi/metabolismo , Complexo de Golgi/ultraestrutura , Camundongos , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Imagem Molecular , Especificidade de Órgãos , Ratos , Ribossomos/ultraestrutura , Estresse Fisiológico
2.
Elife ; 82019 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-31855182

RESUMO

mRNA translation and decay appear often intimately linked although the rules of this interplay are poorly understood. In this study, we combined our recent P-body transcriptome with transcriptomes obtained following silencing of broadly acting mRNA decay and repression factors, and with available CLIP and related data. This revealed the central role of GC content in mRNA fate, in terms of P-body localization, mRNA translation and mRNA stability: P-bodies contain mostly AU-rich mRNAs, which have a particular codon usage associated with a low protein yield; AU-rich and GC-rich transcripts tend to follow distinct decay pathways; and the targets of sequence-specific RBPs and miRNAs are also biased in terms of GC content. Altogether, these results suggest an integrated view of post-transcriptional control in human cells where most translation regulation is dedicated to inefficiently translated AU-rich mRNAs, whereas control at the level of 5' decay applies to optimally translated GC-rich mRNAs.


Assuntos
Composição de Bases/genética , Estabilidade de RNA/genética , RNA Mensageiro Estocado/genética , RNA Mensageiro/genética , Regulação da Expressão Gênica/genética , Humanos , MicroRNAs/química , MicroRNAs/genética , Biossíntese de Proteínas/genética , RNA Mensageiro/química , RNA Mensageiro Estocado/química , Transcriptoma/genética
3.
Am J Hum Genet ; 105(3): 509-525, 2019 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-31422817

RESUMO

The human RNA helicase DDX6 is an essential component of membrane-less organelles called processing bodies (PBs). PBs are involved in mRNA metabolic processes including translational repression via coordinated storage of mRNAs. Previous studies in human cell lines have implicated altered DDX6 in molecular and cellular dysfunction, but clinical consequences and pathogenesis in humans have yet to be described. Here, we report the identification of five rare de novo missense variants in DDX6 in probands presenting with intellectual disability, developmental delay, and similar dysmorphic features including telecanthus, epicanthus, arched eyebrows, and low-set ears. All five missense variants (p.His372Arg, p.Arg373Gln, p.Cys390Arg, p.Thr391Ile, and p.Thr391Pro) are located in two conserved motifs of the RecA-2 domain of DDX6 involved in RNA binding, helicase activity, and protein-partner binding. We use functional studies to demonstrate that the first variants identified (p.Arg373Gln and p.Cys390Arg) cause significant defects in PB assembly in primary fibroblast and model human cell lines. These variants' interactions with several protein partners were also disrupted in immunoprecipitation assays. Further investigation via complementation assays included the additional variants p.Thr391Ile and p.Thr391Pro, both of which, similarly to p.Arg373Gln and p.Cys390Arg, demonstrated significant defects in P-body assembly. Complementing these molecular findings, modeling of the variants on solved protein structures showed distinct spatial clustering near known protein binding regions. Collectively, our clinical and molecular data describe a neurodevelopmental syndrome associated with pathogenic missense variants in DDX6. Additionally, we suggest DDX6 join the DExD/H-box genes DDX3X and DHX30 in an emerging class of neurodevelopmental disorders involving RNA helicases.


Assuntos
RNA Helicases DEAD-box/genética , Deficiência Intelectual/genética , Mutação de Sentido Incorreto , Proteínas Proto-Oncogênicas/genética , RNA/genética , Humanos
5.
Mol Cell ; 68(1): 144-157.e5, 2017 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-28965817

RESUMO

Within cells, soluble RNPs can switch states to coassemble and condense into liquid or solid bodies. Although these phase transitions have been reconstituted in vitro, for endogenous bodies the diversity of the components, the specificity of the interaction networks, and the function of the coassemblies remain to be characterized. Here, by developing a fluorescence-activated particle sorting (FAPS) method to purify cytosolic processing bodies (P-bodies) from human epithelial cells, we identified hundreds of proteins and thousands of mRNAs that structure a dense network of interactions, separating P-body from non-P-body RNPs. mRNAs segregating into P-bodies are translationally repressed, but not decayed, and this repression explains part of the poor genome-wide correlation between RNA and protein abundance. P-bodies condense thousands of mRNAs that strikingly encode regulatory processes. Thus, we uncovered how P-bodies, by condensing and segregating repressed mRNAs, provide a physical substrate for the coordinated regulation of posttranscriptional mRNA regulons.


Assuntos
Regulação da Expressão Gênica , Proteoma/genética , RNA Mensageiro/genética , Regulon , Ribonucleoproteínas/genética , Fracionamento Celular , Citoplasma/metabolismo , Grânulos Citoplasmáticos/química , Grânulos Citoplasmáticos/metabolismo , Ontologia Genética , Células HEK293 , Células HeLa , Humanos , Anotação de Sequência Molecular , Transição de Fase , Biossíntese de Proteínas , Proteoma/metabolismo , Estabilidade de RNA , RNA Mensageiro/metabolismo , Ribonucleoproteínas/metabolismo
6.
Sci Rep ; 7(1): 5172, 2017 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-28701771

RESUMO

Hormone secretion relies on secretory granules which store hormones in endocrine cells and release them upon cell stimulation. The molecular events leading to hormone sorting and secretory granule formation at the level of the TGN are still elusive. Our proteomic analysis of purified whole secretory granules or secretory granule membranes uncovered their association with the actomyosin components myosin 1b, actin and the actin nucleation complex Arp2/3. We found that myosin 1b controls the formation of secretory granules and the associated regulated secretion in both neuroendocrine cells and chromogranin A-expressing COS7 cells used as a simplified model of induced secretion. We show that F-actin is also involved in secretory granule biogenesis and that myosin 1b cooperates with Arp2/3 to recruit F-actin to the Golgi region where secretory granules bud. These results provide the first evidence that components of the actomyosin complex promote the biogenesis of secretory granules and thereby regulate hormone sorting and secretion.


Assuntos
Actinas/genética , Miosina Tipo I/genética , Vesículas Secretórias/metabolismo , Actinas/metabolismo , Animais , Transporte Biológico , Células COS , Proteínas de Transporte , Chlorocebus aethiops , Complexo de Golgi/metabolismo , Camundongos , Miosina Tipo I/metabolismo , Células Neuroendócrinas/metabolismo , Sistemas Neurossecretores/metabolismo , Células PC12 , Ligação Proteica , Ratos
7.
Eur J Cancer ; 50(17): 3039-49, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25307750

RESUMO

AIM: In prostate cancer (PCa), neuroendocrine differentiation (NED) is commonly observed in relapsing, hormone therapy-resistant tumours after androgen deprivation. However, the molecular mechanisms involved in the NED of PCa cells remain poorly understood. In this study, we investigated the expression of the neuroendocrine secretory protein secretogranin II (SgII) in PCa, and its potential involvement in the progression of this cancer as a granulogenic factor promoting NED. METHODS: We have examined SgII immunoreactivity in 25 benign prostate hyperplasia and 32 PCa biopsies. In vitro experiments were performed to investigate the involvement of SgII in the neuroendocrine differentiation and the proliferation of PCa cell lines. RESULTS: We showed that immunoreactive SgII intensity correlates with tumour grade in PCa patients. Using the androgen-dependent lymph node cancer prostate cells (LNCaP) cells, we found that NED triggered by androgen deprivation is associated with the induction of SgII expression. In addition, forced expression of SgII in LNCaP cells implemented a regulated secretory pathway by triggering the formation of secretory granule-like structures competent for hormone storage and regulated release. Finally, we found that SgII promotes prostate cancer (CaP) cell proliferation. CONCLUSION: The present data show that SgII is highly expressed in advanced PCa and may contribute to the neuroendocrine differentiation by promoting the formation of secretory granules and the proliferation of PCa cells.


Assuntos
Neoplasias da Próstata/metabolismo , Secretogranina II/metabolismo , Antagonistas de Androgênios/farmacologia , Androgênios/farmacologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Meios de Cultura/farmacologia , Progressão da Doença , Humanos , Masculino , Neuropeptídeo Y/farmacologia , Antígeno Prostático Específico/metabolismo , Neoplasias da Próstata/patologia , Esteroides/farmacologia
8.
Eur J Cancer ; 50(12): 2126-33, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24910418

RESUMO

AIM: In the present study, we have examined the presence of orexins and their receptors in prostate cancer (CaP) and investigated their effects on the apoptosis of prostate cancer cells. METHODS: We have localised the orexin type 1 and 2 receptors (OX1R and OX2R) and orexin A (OxA) in CaP sections of various grades and we have quantified tumour cells containing OX1R. Expression of OX1R was evaluated in the androgeno-dependent (AD) LNCaP and the androgeno-independent (AI) DU145 prostate cancer cells submitted or not to a neuroendocrine differentiation. The effects of orexins on the apoptosis and viability of DU145 cells were also investigated. RESULTS: OX1R is strongly expressed in carcinomatous foci exhibiting a neuroendocrine differentiation, and the number of OX1R-stained cancer cells increases with the grade of the CaP. In contrast, OX2R is only detected in scattered malignant cells in high grade CaP. OX1R is expressed in the AI DU145 cells but is undetectable in the LNCaP cells. Acquisition of a neuroendocrine phenotype by the DU145 cells is associated with an overexpression of OX1R. Orexins induce the apoptosis of DU145 cells submitted to a neuroendocrine differentiation. CONCLUSION: The present data indicate that OX1R-driven apoptosis is overexpressed in AI CaP exhibiting a neuroendocrine differentiation opening a gate for novel therapies for these aggressive cancers which are incurable until now.


Assuntos
Células Neuroendócrinas , Receptores de Orexina/fisiologia , Neoplasias da Próstata/metabolismo , Apoptose/fisiologia , Proliferação de Células , Sobrevivência Celular/fisiologia , Humanos , Imuno-Histoquímica , Masculino , Células Neuroendócrinas/metabolismo , Células Neuroendócrinas/patologia , Receptores de Orexina/genética , Neoplasias da Próstata/patologia , RNA Mensageiro/metabolismo , Células Tumorais Cultivadas
9.
J Clin Endocrinol Metab ; 98(11): 4346-54, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24001749

RESUMO

CONTEXT: A number of incidentally discovered pheochromocytomas are not associated with hypertension. The characteristics of normotensive incidentally discovered pheochromocytomas (NIPs) are poorly known. OBJECTIVE: The purpose of this work was to assess the clinical, hormonal, histological, and molecular features of NIPs. DESIGN: This was a retrospective cohort recruited from 2001 to 2011 in 2 tertiary care medical departments. PATIENTS AND METHODS: Clinical, biological, and radiological investigations performed in 96 consecutive patients with sporadic unilateral pheochromocytomas were examined; 47 patients had overt pheochromocytomas responsible for hypertension. Among the patients with incidental pheochromocytomas, 28 had hypertension and 21 were normotensive (NIPs). A total of 62 tumors were examined to determine the Pheochromocytoma of the Adrenal Gland Scale Score, and 29 were studied for the expression of 16 genes involved in chromaffin cell function. RESULTS: Tumor size and metaiodobenzylguanidine (MIBG) scintigraphy results were similar for hypertensive pheochromocytomas (HPs) and NIPs. Patients with NIPs displayed reduced summed levels of urinary catecholamines and metanephrines and, more specifically, reduced levels of adrenaline and metadrenaline compared with those of patients with HPs (P < .001). Urinary metanephrines had 98% diagnostic sensitivity in patients with HPs and only 75% in patients with NIPs (P < .01). Tumor diameter positively correlated with the total amount of urinary concentrations of metanephrines in patients with HPs (P < .001) but not in patients with NIPs. NIPs displayed global decreased chromaffin gene expression (reaching significance for 5 of them) and 2 corresponding proteins (phenylethanolamine N-methyltransferase and secretogranin II) and a significant increase in the cellularity, mitotic activity, and presence of atypical mitosis (P < .05). CONCLUSIONS: NIPs differ from pheochromocytomas responsible for hypertension and display features of altered chromaffin differentiation. These tumors may be misdiagnosed with the use of the usual biological diagnostic tools.


Assuntos
Neoplasias das Glândulas Suprarrenais , Células Cromafins/diagnóstico por imagem , Células Cromafins/fisiologia , Regulação Neoplásica da Expressão Gênica , Feocromocitoma , 3-Iodobenzilguanidina , Neoplasias das Glândulas Suprarrenais/diagnóstico por imagem , Neoplasias das Glândulas Suprarrenais/genética , Neoplasias das Glândulas Suprarrenais/metabolismo , Células Cromafins/patologia , Feminino , Humanos , Hipertensão/diagnóstico por imagem , Hipertensão/genética , Hipertensão/metabolismo , Achados Incidentais , Masculino , Pessoa de Meia-Idade , Feocromocitoma/diagnóstico por imagem , Feocromocitoma/genética , Feocromocitoma/metabolismo , Cintilografia , Compostos Radiofarmacêuticos , Estudos Retrospectivos , Transcriptoma
10.
Hum Mol Genet ; 22(18): 3624-40, 2013 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-23674521

RESUMO

Hypertension is a common hereditary syndrome with unclear pathogenesis. Chromogranin A (Chga), which catalyzes formation and cargo storage of regulated secretory granules in neuroendocrine cells, contributes to blood pressure homeostasis centrally and peripherally. Elevated Chga occurs in spontaneously hypertensive rat (SHR) adrenal glands and plasma, but central expression is unexplored. In this report, we measured SHR and Wistar-Kyoto rat (control) Chga expression in central and peripheral nervous systems, and found Chga protein to be decreased in the SHR brainstem, yet increased in the adrenal and the plasma. By re-sequencing, we systematically identified five promoter, two coding and one 3'-untranslated region (3'-UTR) polymorphism at the SHR (versus WKY or BN) Chga locus. Using HXB/BXH recombinant inbred (RI) strain linkage and correlations, we demonstrated genetic determination of Chga expression in SHR, including a cis-quantitative trait loci (QTLs) (i.e. at the Chga locus), and such expression influenced biochemical determinants of blood pressure, including a cascade of catecholamine biosynthetic enzymes, catecholamines themselves and steroids. Luciferase reporter assays demonstrated that the 3'-UTR polymorphism (which disrupts a microRNA miR-22 motif) and promoter polymorphisms altered gene expression consistent with the decline in SHR central Chga expression. Coding region polymorphisms did not account for changes in Chga expression or function. Thus, we hypothesized that the 3'-UTR and promoter mutations lead to dysregulation (diminution) of Chga in brainstem cardiovascular control nuclei, ultimately contributing to the pathogenesis of hypertension in SHR. Accordingly, we demonstrated that in vivo administration of miR-22 antagomir to SHR causes substantial (∼18 mmHg) reductions in blood pressure, opening a novel therapeutic avenue for hypertension.


Assuntos
Cromogranina A/genética , Cromogranina A/metabolismo , Hipertensão/genética , MicroRNAs/genética , Regiões Promotoras Genéticas , Regiões 3' não Traduzidas , Glândulas Suprarrenais/metabolismo , Animais , Pressão Sanguínea/genética , Tronco Encefálico/metabolismo , Linhagem Celular Tumoral , Cromogranina A/sangue , Cromogranina A/química , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Ligação Genética , Humanos , Hipertensão/tratamento farmacológico , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Masculino , MicroRNAs/metabolismo , Células PC12 , Polimorfismo Genético , Estrutura Secundária de Proteína , Locos de Características Quantitativas , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Alinhamento de Sequência , Transcrição Gênica
11.
Eur J Cancer ; 49(2): 511-9, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22863147

RESUMO

AIM: Accumulating data suggest that neuropeptides produced by neuroendocrine cells play a crucial role in the progression and aggressiveness of hormone refractory prostate cancer (CaP). In this study, we have investigated the presence and function of the neuropeptide 26RFa in CaP. METHODS: We have localised and quantified tumour cells containing 26RFa and its receptor, GPR103, in CaP sections of various grades. In vitro experiments were performed to investigate the effects of 26RFa on the migration, proliferation and neuroendocrine differentiation of the androgeno-independent (AI) prostate cancer cell line DU145. RESULTS: 26RFa and GPR103 are present in carcinomatous foci exhibiting a neuroendocrine differentiation, and the number of 26RFa and GPR103-immunoreactive cancer cells increases with the grade of CaP. 26RFa stimulated the migration of native or transdifferentiated AI DU145 cells, but had no effect on their proliferation. Furthermore, 26RFa induced the neuroendocrine differentiation of DU145 cells as assessed by the occurrence of neurite-like extensions and the increase of the expression of the neuroendocrine marker chromogranin A. CONCLUSION: The present data indicate that 26RFa may participate to the development of CaP at the AI state by promoting the neuroendocrine differentiation and the migration of cancer cells via autocrine/paracrine mechanisms.


Assuntos
Neuropeptídeos/biossíntese , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Androgênios/metabolismo , Diferenciação Celular/fisiologia , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Progressão da Doença , Humanos , Imuno-Histoquímica , Masculino , Neoplasias Hormônio-Dependentes/metabolismo , Neoplasias Hormônio-Dependentes/patologia , Células Neuroendócrinas/metabolismo , Células Neuroendócrinas/patologia , Receptores Acoplados a Proteínas G/biossíntese
12.
Endocrinology ; 153(9): 4444-56, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22851679

RESUMO

Chromogranins are a family of acidic glycoproteins that play an active role in hormone and neuropeptide secretion through their crucial role in secretory granule biogenesis in neuroendocrine cells. However, the molecular mechanisms underlying their granulogenic activity are still not fully understood. Because we previously demonstrated that the expression of the major component of secretory granules, chromogranin A (CgA), is able to induce the formation of secretory granules in nonendocrine COS-7 cells, we decided to use this model to dissect the mechanisms triggered by CgA leading to the biogenesis and trafficking of such granules. Using quantitative live cell imaging, we first show that CgA-induced organelles exhibit a Ca(2+)-dependent trafficking, in contrast to native vesicle stomatitis virus G protein-containing constitutive vesicles. To identify the proteins that confer such properties to the newly formed granules, we developed CgA-stably-expressing COS-7 cells, purified their CgA-containing granules by subcellular fractionation, and analyzed the granule proteome by liquid chromatography-tandem mass spectrometry. This analysis revealed the association of several cytosolic proteins to the granule membrane, including GTPases, cytoskeleton-based molecular motors, and other proteins with actin- and/or Ca(2+)-binding properties. Furthermore, disruption of cytoskeleton affects not only the distribution and the transport but also the Ca(2+)-evoked exocytosis of the CgA-containing granules, indicating that these granules interact with microtubules and cortical actin for the regulated release of their content. These data demonstrate for the first time that the neuroendocrine factor CgA induces the recruitment of cytoskeleton-, GTP-, and Ca(2+)-binding proteins in constitutively secreting COS-7 cells to generate vesicles endowed with typical dynamics and exocytotic properties of neuroendocrine secretory granules.


Assuntos
Actinas/metabolismo , Cálcio/metabolismo , Cromogranina A/farmacologia , Vesículas Secretórias/efeitos dos fármacos , Vesículas Secretórias/metabolismo , Actinas/ultraestrutura , Animais , Western Blotting , Células COS , Chlorocebus aethiops , Eletroforese em Gel de Poliacrilamida , Exocitose/efeitos dos fármacos , Microscopia Eletrônica , Microscopia de Fluorescência , Vesículas Secretórias/ultraestrutura , Espectrometria de Massas em Tandem
13.
Circ Cardiovasc Genet ; 4(4): 381-9, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21558123

RESUMO

BACKGROUND: The catecholamine release-inhibitor catestatin and its precursor chromogranin A (CHGA) may constitute "intermediate phenotypes" in the analysis of genetic risk for cardiovascular disease such as hypertension. Previously, the vacuolar H(+)-ATPase subunit gene ATP6V0A1 was found within the confidence interval for linkage with catestatin secretion in a genome-wide study, and its 3'-UTR polymorphism T+3246C (rs938671) was associated with both catestatin processing from CHGA and population blood pressure. We explored the molecular mechanism of this effect by experiments with transfected chimeric photoproteins in chromaffin cells. METHODS AND RESULTS: Placing the ATP6V0A1 3'-UTR downstream of a luciferase reporter, we found that the C (variant) allele decreased overall gene expression. The 3'-UTR effect was verified by coupled in vitro transcription/translation of the entire/intact human ATP6V0A1 mRNA. Chromaffin granule pH, monitored by fluorescence of CHGA/EGFP chimera during vesicular H(+)-ATPase inhibition by bafilomycin A1, was more easily perturbed during coexpression of the ATP6V0A1 3'-UTR C-allele than the T-allele. After bafilomycin A1 treatment, the ratio of CHGA precursor to its catestatin fragments in PC12 cells was substantially diminished, though the qualitative composition of such fragments was not affected (on immunoblot or matrix-assisted laser desorption ionization (MALDI) mass spectrometry). Bafilomycin A1 treatment also decreased exocytotic secretion from the regulated pathway, monitored by a CHGA chimera tagged with embryonic alkaline phosphatase. 3'-UTR T+3246C created a binding motif for micro-RNA hsa-miR-637; cotransfection of hsa-miR-637 precursor or antagomir/inhibitor oligonucleotides yielded the predicted changes in expression of luciferase reporter/ATP6V0A1-3'-UTR plasmids varying at T+3246C. CONCLUSIONS: The results suggest a series of events whereby ATP6V0A1 3'-UTR variant T+3246C functioned: ATP6V0A1 expression probably was affected through differential micro-RNA effects, altering vacuolar pH and consequently CHGA processing and exocytotic secretion.


Assuntos
Regiões 3' não Traduzidas/genética , Cromogranina A/metabolismo , Variação Genética , Hipertensão/genética , MicroRNAs , ATPases Vacuolares Próton-Translocadoras/genética , Sítios de Ligação , Exocitose , Humanos
14.
Cell Mol Neurobiol ; 30(8): 1189-95, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21046450

RESUMO

Chromogranin A (CgA) is a soluble glycoprotein stored along with hormones and neuropeptides in secretory granules of endocrine cells. In the last four decades, intense efforts have been concentrated to characterize the structure and the biological function of CgA. Besides, CgA has been widely used as a diagnostic marker for tumors of endocrine origin, essential hypertension, various inflammatory diseases, and neurodegenerative disorders such as amyotrophic lateral sclerosis and Alzheimer's disease. CgA displays peculiar structural features, including numerous multibasic cleavage sites for prohormone convertases as well as a high proportion of acidic residues. Thus, it has been proposed that CgA represents a precursor of biologically active peptides, and a "granulogenic protein" that plays an important role as a chaperone for catecholamine storage in adrenal chromaffin cells. The widespread distribution of CgA throughout the neuroendocrine system prompted several groups to investigate the role of CgA in peptide hormone sorting to the regulated secretory pathway. This review summarizes the findings and theoretical concepts around the molecular machinery used by CgA to exert this putative intracellular function. Since CgA terminal regions exhibited strong sequence conservation through evolution, our work focused on the implication of these domains as potential functional determinants of CgA. Characterization of the molecular signals implicating CgA in the intracellular traffic of hormones represents a major biological issue that may contribute to unraveling the mechanisms defining the secretory competence of neuroendocrine cells.


Assuntos
Cromogranina A/metabolismo , Hormônios Peptídicos/metabolismo , Vesículas Secretórias/metabolismo , Animais , Humanos , Modelos Biológicos , Hormônios Peptídicos/química , Estrutura Quaternária de Proteína , Transporte Proteico
15.
J Biol Chem ; 285(13): 10030-10043, 2010 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-20061385

RESUMO

Processes underlying the formation of dense core secretory granules (DCGs) of neuroendocrine cells are poorly understood. Here, we present evidence that DCG biogenesis is dependent on the secretory protein secretogranin (Sg) II, a member of the granin family of pro-hormone cargo of DCGs in neuroendocrine cells. Depletion of SgII expression in PC12 cells leads to a decrease in both the number and size of DCGs and impairs DCG trafficking of other regulated hormones. Expression of SgII fusion proteins in a secretory-deficient PC12 variant rescues a regulated secretory pathway. SgII-containing dense core vesicles share morphological and physical properties with bona fide DCGs, are competent for regulated exocytosis, and maintain an acidic luminal pH through the V-type H(+)-translocating ATPase. The granulogenic activity of SgII requires a pH gradient along this secretory pathway. We conclude that SgII is a critical factor for the regulation of DCG biogenesis in neuroendocrine cells, mediating the formation of functional DCGs via its pH-dependent aggregation at the trans-Golgi network.


Assuntos
Catecolaminas/metabolismo , Secretogranina II/metabolismo , Vesículas Secretórias/metabolismo , Animais , Células COS , Chlorocebus aethiops , Grânulos Cromafim/metabolismo , Inativação Gênica , Vetores Genéticos , Concentração de Íons de Hidrogênio , Células Neuroendócrinas/metabolismo , Células PC12 , RNA Interferente Pequeno/metabolismo , Ratos , Proteínas Recombinantes de Fusão/metabolismo
16.
J Am Soc Nephrol ; 20(7): 1623-32, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19520754

RESUMO

Chromogranin A (CHGA), a protein released from secretory granules of chromaffin cells and sympathetic nerves, triggers endothelin-1 release from endothelial cells. CHGA polymorphisms associate with an increased risk for ESRD, but whether altered CHGA-endothelium interactions may explain this association is unknown. Here, CHGA led to the release of endothelin-1 and Weibel-Palade body exocytosis in cultured human umbilical vein endothelial cells. In addition, CHGA triggered secretion of endothelin-1 from glomerular endothelial cells and TGF-beta1 from mesangial cells cocultured with glomerular endothelial cells. In humans, plasma CHGA correlated positively with endothelin-1 and negatively with GFR. GFR was highly heritable in twin pairs, and common promoter haplotypes of CHGA predicted GFR. In patients with progressive hypertensive renal disease, a CHGA haplotype predicted rate of GFR decline. In conclusion, these data suggest that CHGA acts through the glomerular endothelium to regulate renal function.


Assuntos
Cromogranina A/metabolismo , Endotélio/metabolismo , Exocitose/fisiologia , Taxa de Filtração Glomerular/fisiologia , Glomérulos Renais/metabolismo , Corpos de Weibel-Palade/metabolismo , Animais , Células Cultivadas , Cromogranina A/genética , Cromogranina A/farmacologia , Doença Crônica , Técnicas de Cocultura , Relação Dose-Resposta a Droga , Endotelinas/metabolismo , Endotélio/citologia , Endotélio/efeitos dos fármacos , Humanos , Nefropatias/metabolismo , Nefropatias/patologia , Falência Renal Crônica/metabolismo , Falência Renal Crônica/patologia , Glomérulos Renais/citologia , Glomérulos Renais/efeitos dos fármacos , Camundongos , Polimorfismo Genético/genética , Fatores de Risco , Fatores de Tempo , Fator de Crescimento Transformador beta1/metabolismo
17.
Endocrinology ; 150(8): 3547-57, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19372204

RESUMO

Chromogranin A (CgA), the major soluble protein in chromaffin granules, is proteolytically processed to generate biologically active peptides including the catecholamine release inhibitory peptide catestatin. Here we sought to determine whether cysteine protease cathepsin L (CTSL), a novel enzyme for proteolytic processing of neuropeptides, acts like the well-established serine proteases [prohormone convertase (PC)1/3 or PC2] to generate catestatin by proteolytic processing of CgA. We found that endogenous CTSL colocalizes with CgA in the secretory vesicles of primary rat chromaffin cells. Transfection of PC12 cells with an expression plasmid encoding CTSL directed expression of CTSL toward secretory vesicles. Deconvolution fluorescence microscopy suggested greater colocalization of CTSL with CgA than the lysosomal marker LGP110. The overexpression of CTSL in PC12 cells caused cleavage of full-length CgA. CTSL also cleaved CgA in vitro, in time- and dose-dependent fashion, and specificity of the process was documented through E64 (thiol reagent) inhibition. Mass spectrometry on CTSL-digested recombinant CgA identified a catestatin-region peptide, corresponding to CgA(360-373). The pool of peptides generated from the CTSL cleavage of CgA inhibited nicotine-induced catecholamine secretion from PC12 cells. CTSL processing in the catestatin region was diminished by naturally occurring catestatin variants, especially Pro370Leu and Gly364Ser. Among the CTSL-generated peptides, a subset matched those found in the catestatin region in vivo. These findings indicate that CgA can be a substrate for the cysteine protease CTSL both in vitro and in cella, and their colocalization within chromaffin granules in cella suggests the likelihood of an enzyme/substrate relationship in vivo.


Assuntos
Catepsinas/metabolismo , Células Cromafins/metabolismo , Cromogranina A/metabolismo , Cisteína Endopeptidases/metabolismo , Animais , Catecolaminas/metabolismo , Catepsina L , Células Cultivadas , Eletroforese em Gel de Poliacrilamida , Feminino , Imunofluorescência , Humanos , Immunoblotting , Células PC12 , Peptídeos/síntese química , Peptídeos/química , Peptídeos/metabolismo , Ratos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
18.
Biochemistry ; 47(27): 7167-78, 2008 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-18549247

RESUMO

Secretion of proteins and peptides from eukaryotic cells takes place by both constitutive and regulated pathways. Regulated secretion may involve interplay of proteins that are currently unknown. Recent studies suggest an important role of chromogranin A (CHGA) in the regulated secretory pathway in neuroendocrine cells, but the mechanism by which CHGA enters the regulated pathway, or even triggers the formation of the pathway, remains unclear. In this study, we used a transcriptome/proteome-wide approach, to discover binding partners for CHGA, by employing a phage display cDNA library method. Several proteins within or adjacent to the secretory pathway were initially detected as binding partners of recombinant human CHGA. We then focused on the trans-Golgi protein SCLIP (STMN3) and its stathmin paralog SCG10 (STMN2) for functional study. Co-immunoprecipitation experiments confirmed the interaction of each of these two proteins with CHGA in vitro. SCLIP and SCG10 were colocalized to the Golgi apparatus of chromaffin cells in vivo and shared localization with CHGA as it transited the Golgi. Downregulation of either SCLIP or SCG10 by synthetic siRNAs virtually abolished chromaffin cell secretion of a transfected CHGA-EAP chimera (expressing CHGA fused to an enzymatic reporter, and trafficked to the regulated pathway). SCLIP siRNA also decreased the level of secretion of endogenous CHGA and SCG2, as well as transfected human growth hormone, while SCG10 siRNA decreased the level of regulated secretion of endogenous CHGB. Moreover, a dominant negative mutant of SCG10 (Cys 22,Cys 24-->Ala 22,Ala 24) significantly blocked secretion of the transfected CHGA-EAP chimera. A decrease in the buoyant density of chromaffin granules was observed after downregulation of SCG10 by siRNA, suggesting participation of these stathmins in granule formation or maturation. We conclude that SCLIP and SCG10 interact with CHGA, share partial colocalization in the Golgi apparatus, and may be necessary for typical transmitter storage and release from chromaffin cells.


Assuntos
Cromogranina A/metabolismo , Proteínas de Membrana/metabolismo , Sistemas Neurossecretores/metabolismo , Estatmina/metabolismo , Rede trans-Golgi/metabolismo , Animais , Células Cromafins/metabolismo , Grânulos Cromafim/metabolismo , Cromogranina B/metabolismo , Regulação para Baixo , Inativação Gênica , Genes Dominantes , Hormônio do Crescimento/metabolismo , Humanos , Imunoprecipitação , Espaço Intracelular/metabolismo , Proteínas Mutantes/metabolismo , Células PC12 , Ligação Proteica , Transporte Proteico , RNA Interferente Pequeno/metabolismo , Ratos , Proteínas Recombinantes de Fusão/metabolismo , Transfecção
19.
J Biol Chem ; 283(17): 11807-22, 2008 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-18299326

RESUMO

Secretogranin II (SgII) belongs to the granin family of prohormones widely distributed in dense-core secretory granules (DCGs) of endocrine, neuroendocrine, and neuronal cells, including sympathoadrenal chromaffin cells. The mechanisms by which secretory proteins, and granins in particular, are sorted into the regulated secretory pathway are unsettled. We designed a strategy based on novel chimeric forms of human SgII fused to fluorescent (green fluorescent protein) or chemiluminescent (embryonic alkaline phosphatase) reporters to identify trafficking determinants mediating DCG targeting of SgII in sympathoadrenal cells. Three-dimensional deconvolution fluorescence microscopy and secretagogue-stimulated release studies demonstrate that SgII chimeras are correctly targeted to DCGs and released by exocytosis in PC12 and primary chromaffin cells. Results from a Golgi-retained mutant form of SgII suggest that sorting of SgII into DCGs depends on a saturable sorting machinery at the trans-Golgi/trans-Golgi network. Truncation analyses reveal the presence of DCG-targeting signals within both the N- and C-terminal regions of SgII, with the putative alpha-helix-containing SgII-(25-41) and SgII-(334-348) acting as sufficient, independent sorting domains. This study defines sequence features of SgII mediating vesicular targeting in sympathoadrenal cells and suggests a mechanism by which discrete domains of the molecule function in sorting, perhaps by virtue of a particular arrangement in tertiary structure and/or interaction with a specific component of the DCG membrane.


Assuntos
Regulação da Expressão Gênica , Secretogranina II/metabolismo , Fosfatase Alcalina/metabolismo , Animais , Membrana Celular/metabolismo , Exocitose , Complexo de Golgi/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Células PC12 , Conformação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Ratos , Proteínas Recombinantes de Fusão/química , Transdução de Sinais
20.
J Biol Chem ; 281(49): 38038-51, 2006 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-17032650

RESUMO

Chromogranin A (CgA) may be critical for secretory granule biogenesis in sympathoadrenal cells. We found that silencing the expression of CgA reduced the number of secretory granules in normal sympathoadrenal cells (PC12), and we therefore questioned whether a discrete domain of CgA might promote the formation of a regulated secretory pathway in variant sympathoadrenal cells (A35C) devoid of such a phenotype. The secretory granule-forming activity of a series of human CgA domains labeled with a hemagglutinin epitope, green fluorescent protein, or embryonic alkaline phosphatase was assessed in A35C cells by deconvolution and electron microscopy and by secretagogue-stimulated release assays. Expression of CgA in A35C cells induced the formation of vesicular organelles throughout the cytoplasm, whereas two constitutive secretory pathway markers accumulated in the Golgi complex. The lysosome-associated membrane protein LGP110 did not co-localize with CgA, consistent with non-lysosomal targeting of the granin in A35C cells. Thus, CgA-expressing A35C cells showed electron-dense granules approximately 180-220 nm in diameter, and secretagogue-stimulated exocytosis of CgA from A35C cells suggested that expression of the granin may be sufficient to restore a regulated secretory pathway and thereby rescue the sorting of other secretory proteins. We show that the formation of vesicular structures destined for regulated exocytosis may be mediated by a determinant located within the CgA N-terminal region (CgA-(1-115), with a necessary contribution of CgA-(40-115)), but not the C-terminal region (CgA-(233-439)) of the protein. We propose that CgA promotes the biogenesis of secretory granules by a mechanism involving a granulogenic determinant located within CgA-(40-115) of the mature protein.


Assuntos
Cromogranina A/fisiologia , Vesículas Secretórias/metabolismo , Vesículas Secretórias/ultraestrutura , Glândulas Suprarrenais/fisiologia , Glândulas Suprarrenais/ultraestrutura , Animais , Sequência de Bases , Linhagem Celular , Cromogranina A/antagonistas & inibidores , Cromogranina A/química , Cromogranina A/genética , Humanos , Microscopia Eletrônica , Microscopia de Fluorescência , Células PC12 , RNA Interferente Pequeno/genética , Ratos , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA