Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Lett ; 565: 216209, 2023 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-37169162

RESUMO

The development of androgen receptor signaling inhibitor (ARSI) drug resistance in prostate cancer (PC) remains therapeutically challenging. Our group has described the role of sex determining region Y-box 2 (SOX2) overexpression in ARSI-resistant PC. Continuing this work, we report that NR3C1, the gene encoding glucocorticoid receptor (GR), is a novel SOX2 target in PC, positively regulating its expression. Similar to ARSI treatment, SOX2-positive PC cells are insensitive to GR signaling inhibition using a GR modulating therapy. To understand SOX2-mediated nuclear hormone receptor signaling inhibitor (NHRSI) insensitivity, we performed RNA-seq in SOX2-positive and -negative PC cells following NHRSI treatment. RNA-seq prioritized differentially regulated genes mediating the cell cycle, including G2 checkpoint WEE1 Kinase (WEE1) and cyclin-dependent kinase 1 (CDK1). Additionally, WEE1 and CDK1 were differentially expressed in PC patient tumors dichotomized by high vs low SOX2 gene expression. Importantly, pharmacological targeting of WEE1 (WEE1i) in combination with an ARSI or GR modulator re-sensitizes SOX2-positive PC cells to nuclear hormone receptor signaling inhibition in vitro, and WEE1i combined with ARSI significantly slowed tumor growth in vivo. Collectively, our data suggest SOX2 predicts NHRSI resistance, and simultaneously indicates the addition of WEE1i to improve therapeutic efficacy of NHRSIs in SOX2-positive PC.


Assuntos
Antineoplásicos , Neoplasias de Próstata Resistentes à Castração , Masculino , Humanos , Proteína Quinase CDC2/genética , Proteína Quinase CDC2/metabolismo , Transdução de Sinais , Antineoplásicos/farmacologia , Proteínas de Ciclo Celular/metabolismo , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Neoplasias de Próstata Resistentes à Castração/genética , Antagonistas de Receptores de Andrógenos/farmacologia , Receptores Citoplasmáticos e Nucleares , Linhagem Celular Tumoral , Proteínas Tirosina Quinases/metabolismo , Fatores de Transcrição SOXB1/genética
2.
Toxicol Sci ; 159(1): 224-237, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28903499

RESUMO

Superwarfarins are very long-lasting rodenticides effective in warfarin-resistant rodents at extremely low doses. The consequences of chronic superwarfarin levels in tissues, due to biological half-lives on the order of 20 days, have not been examined. We now characterized the neurological effects of brodifacoum (BDF), one of the most widely used superwarfarins, in adult male Sprague Dawley rats. Dosing curves established the acute oral lethal dose for BDF as 221 ± 14 µg/kg. Measurement of tissue BDF levels showed accumulation throughout the body, including the central nervous system, with levels diminishing over several days. Immunocytochemical staining showed that both astrocyte and microglial activation was increased 4 days after BDF administration, as were levels of carbonylated proteins, and neuronal damage assessed by fluorojade B staining. Direct toxic effects of BDF on neurons and glia were observed using enriched cultures of cerebellar neurons and cortical astrocytes. Proteomic analysis of cerebellar lysates revealed that BDF altered expression of 667 proteins in adult rats. Gene ontology and pathway analysis identified changes in several functional pathways including cell metabolism, mitochondria function, and RNA handling with ribosomal proteins comprising the largest group. In vitro studies using primary astrocytes showed that BDF suppressed de novo protein synthesis. These findings demonstrate that superwarfarin accumulation increases indices of neuroinflammation and neuropathology in adult rodents, suggesting that methods which minimize BDF toxicity may not address delayed neurological sequelae.


Assuntos
4-Hidroxicumarinas/toxicidade , Sistema Nervoso/efeitos dos fármacos , Rodenticidas/toxicidade , 4-Hidroxicumarinas/administração & dosagem , 4-Hidroxicumarinas/farmacocinética , Administração Oral , Animais , Cromatografia Líquida de Alta Pressão , Dose Letal Mediana , Masculino , Sistema Nervoso/metabolismo , Sistema Nervoso/patologia , Proteômica , Ratos , Ratos Sprague-Dawley , Rodenticidas/administração & dosagem , Rodenticidas/farmacocinética , Espectrometria de Massas em Tandem , Distribuição Tecidual
3.
Islets ; 5(4): 156-69, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23896637

RESUMO

Adult human pancreatic ß-cells are primarily quiescent (G0) yet the mechanisms controlling their quiescence are poorly understood. Here, we demonstrate, by immunofluorescence and confocal microscopy, abundant levels of the critical negative cell cycle regulators, p27(Kip1) and p18(Ink4c), 2 key members of cyclin-dependent kinase (CDK) inhibitor family, and glycogen synthase kinase-3 (GSK-3), a serine-threonine protein kinase, in islet ß-cells of adult human pancreatic tissue. Our data show that p27(Kip1) localizes primarily in ß-cell nuclei, whereas, p18(Ink4c) is mostly present in ß-cell cytosol. Additionally, p-p27(S10), a phosphorylated form of p27(Kip1), which was shown to interact with and to sequester cyclinD-CDK4/6 in the cytoplasm, is present in substantial amounts in ß-cell cytosol. Our immunofluorescence analysis displays similar distribution pattern of p27(Kip1), p-p27(S10), p18(Ink4c) and GSK-3 in islet ß-cells of adult mouse pancreatic tissue. We demonstrate marked interaction of p27(Kip1) with cyclin D3, an abundant D-type cyclin in adult human islets, and vice versa as well as with its cognate kinase partners, CDK4 and CDK6. Likewise, we show marked interaction of p18(Ink4c) with CDK4. The data collectively suggest that inhibition of CDK function by p27(Kip1) and p18(Ink4c) contributes to human ß-cell quiescence. Consistent with this, we have found by BrdU incorporation assay that combined treatments of small molecule GSK-3 inhibitor and mitogen/s lead to elevated proliferation of human ß-cells, which is caused partly due to p27(Kip1) downregulation. The results altogether suggest that ex vivo expansion of human ß-cells is achievable via increased proliferation for ß-cell replacement therapy in diabetes.


Assuntos
Ciclo Celular/genética , Proliferação de Células , Inibidor de Quinase Dependente de Ciclina p18/fisiologia , Inibidor de Quinase Dependente de Ciclina p27/fisiologia , Quinase 3 da Glicogênio Sintase/fisiologia , Células Secretoras de Insulina/fisiologia , Adulto , Idoso , Animais , Contagem de Células , Células Cultivadas , Quinases Ciclina-Dependentes/metabolismo , Células Secretoras de Glucagon/citologia , Células Secretoras de Glucagon/metabolismo , Glicogênio Sintase Quinase 3 beta , Humanos , Células Secretoras de Insulina/citologia , Camundongos , Pessoa de Meia-Idade
4.
Islets ; 3(1): 21-34, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21278490

RESUMO

Diabetes (T1DM and T2DM) is characterized by a deficit in ß-cell mass. A broader understanding of human ß-cell replication mechanism is thus important to increase ß-cell proliferation for future therapeutic interventions. Here, we show that p27 (Kip1), a CDK inhibitor, is expressed abundantly in isolated adult human islets and interacts with various positive cell cycle regulatory proteins including D-type cyclins (D1, D2 and D3) and their kinase partners, CDK4 and CDK6. Also, we see interaction of cyclin E and its kinase partner, CDK2, with p27 suggesting a critical role of p27 as a negative cell cycle regulator in human islets. Our data demonstrate interaction of p27 with GSK-3 in ß-cells and show, employing rodent ß-cells (INS-1), isolated human islets and purified ß-cells derived from human islets, that siRNA-mediated depletion of GSK-3 or p27 or 1-AKP / BIO - mediated GSK-3 inhibition results in increased ß-cell proliferation. We also see reduction of p27 levels following GSK-3 inactivation or depletion. Our data show that serum induction of quiescent INS-1 cells leads to sequential phosphorylation of p27 on its S10 and T187 residues with faster kinetics for S10 corresponding with the decreased levels of p27. Altogether our findings indicate that p27 levels in ß-cells are stabilized by GSK-3 and thus p27 down regulation following GSK-3 depletion / inactivation plays a critical role in promoting ß-cell replication.


Assuntos
Divisão Celular , Inibidor de Quinase Dependente de Ciclina p27/genética , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/metabolismo , Células Secretoras de Insulina/metabolismo , Adulto , Idoso , Animais , Divisão Celular/genética , Divisão Celular/fisiologia , Linhagem Celular , Inibidor de Quinase Dependente de Ciclina p27/antagonistas & inibidores , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Quinase 3 da Glicogênio Sintase/genética , Humanos , Células Secretoras de Insulina/fisiologia , Masculino , Pessoa de Meia-Idade , RNA Interferente Pequeno/farmacologia , Ratos
5.
Diabetes Res Clin Pract ; 85(3): 252-7, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19595468

RESUMO

Emerging reports on human islets emphasize distinct differences from the widely accepted prototype of rodent islets, raising questions over their suitability for human studies. Here we aim at elucidating architectural differences and similarities of human versus rodent islets. The cellular composition and architecture of human and rodent islets were compared through three-dimensional (3D) reconstructions. Physiological and pathological changes were examined using islets from various mouse models such as non-obese diabetic (NOD), ob/ob, db/db mice and during pregnancy. A subpopulation of human islets is composed of clusters of alpha-cells within the central beta-cell cores, while the overall proportion of alpha-cells varies among islets. In mouse islets under normal conditions, alpha-cells are localized in the islet periphery, but they do not envelop the entire beta-cell core, so that beta-cells are exposed on the outer layer of the islet, as in most human islets. Also, an increased proportion of alpha-cells within the central core is observed in the pancreas of mouse models exhibiting increased demand for insulin. In summary, human and mouse islets share common architectural features as endocrine micro-organs. Since these may hold a key to better understanding islet plasticity, our concept of the prototypic islet should be revised.


Assuntos
Ilhotas Pancreáticas/citologia , Animais , Glicemia/metabolismo , Divisão Celular , Tamanho Celular , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/patologia , Feminino , Células Secretoras de Glucagon/citologia , Células Secretoras de Glucagon/patologia , Humanos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/patologia , Ilhotas Pancreáticas/anatomia & histologia , Ilhotas Pancreáticas/patologia , Camundongos , Camundongos Obesos , Estado Pré-Diabético/patologia , Gravidez , Ratos
6.
Biochem J ; 415(1): 35-43, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18554181

RESUMO

Cellular synthesis of peptide hormones requires PCs (prohormone convertases) for the endoproteolysis of prohormones. Antral G-cells synthesize the most gastrin and express PC1/3, 2 and 5/6 in the rat and human. But the cleavage sites in progastrin for each PC have not been determined. Therefore, in the present study, we measured the concentrations of progastrin, processing intermediates and alpha-amidated gastrins in antral extracts from PC1/3-null mice and compared the results with those in mice lacking PC2 and wild-type controls. The expression of PCs was examined by immunocytochemistry and in situ hybridization of mouse G-cells. Finally, the in vitro effect of recombinant PC5/6 on progastrin and progastrin fragments containing the relevant dibasic cleavage sites was also examined. The results showed that mouse G-cells express PC1/3, 2 and 5/6. The concentration of progastrin in PC1/3-null mice was elevated 3-fold. Chromatography showed that cleavage of the Arg(36)Arg(37) and Arg(73)Arg(74) sites were grossly decreased. Accordingly, the concentrations of progastrin products were markedly reduced, alpha-amidated gastrins (-34 and -17) being 25% of normal. Lack of PC1/3 was without effect on the third dibasic site (Lys(53)Lys(54)), which is the only processing site for PC2. Recombinant PC5/6 did not cleave any of the dibasic processing sites in progastrin and fragments containing the relevant dibasic processing sites. The complementary cleavages of PC1/3 and 2, however, suffice to explain most of the normal endoproteolysis of progastrin. Moreover, the results show that PCs react differently to the same dibasic sequences, suggesting that additional structural factors modulate the substrate specificity.


Assuntos
Gastrinas/metabolismo , Pró-Proteína Convertase 1/metabolismo , Pró-Proteína Convertase 5/metabolismo , Precursores de Proteínas/metabolismo , Sequência de Aminoácidos , Animais , Células CHO , Cricetinae , Cricetulus , Células Secretoras de Gastrina/metabolismo , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Pró-Proteína Convertase 2/metabolismo , Antro Pilórico/química , Proteínas Recombinantes/metabolismo
7.
Biochem J ; 400(1): 209-15, 2006 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-16800814

RESUMO

Understanding the functions of the widely expressed PCs (prohormone/proprotein convertases), including PC5/6, furin and PACE4 (paired basic amino acid cleaving enzyme 4), in animal models is difficult since individual knockouts of these PCs in mice exhibit early embryonic lethality. To investigate the roles of PC5/6 in processing pro-CART (pro-cocaine- and amphetamine-regulated transcript), an important anorexigenic peptide precursor, we have generated GH3 cells silenced for PC5/6 expression by RNAi (RNA interference). We show, following transient knockdown of PC5/6 in these neuroendocrine cells, that generation of the two bioactive forms, CART I (amino acids 42-89/55-102) and CART II (amino acids 49-89/62-102), from pro-CART is impaired due to a lack particularly of the A isoform of PC5/6. The results indicate that PC5/6A shares specificities primarily with PC2 (PC5/6A

Assuntos
Proteínas do Tecido Nervoso/metabolismo , Pró-Proteína Convertase 5/metabolismo , Precursores de Proteínas/metabolismo , Interferência de RNA , Animais , Sequência de Bases , Western Blotting , Linhagem Celular Tumoral , Isoenzimas/genética , Isoenzimas/metabolismo , Pró-Proteína Convertase 5/genética , Isoformas de Proteínas/metabolismo , Processamento de Proteína Pós-Traducional , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
Peptides ; 27(8): 1919-25, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16784796

RESUMO

Cocaine- and amphetamine-regulated transcript (CART) peptides are expressed in several neuroendocrine tissues, including hypothalamus, pituitary, gut, adrenal and pancreas, and are involved in regulating important biological processes including feeding/appetite, drug reward and stress. CART is synthesized as larger, inactive peptide precursors (pro-CART) that require endoproteolytic processing to generate smaller, active forms. Prohormone/proprotein convertases (PCs), a family of calcium-dependent, serine endoproteases, have been shown to cleave many protein precursors in the regulated/constitutive secretory pathway to generate smaller fragments. In our previous studies, we have demonstrated the important roles of the two neuroendocrine-specific PCs, PC2 and PC1/3, in processing the two pro-CART isoforms, long (102aa) and short (89aa), to generate the bioactive CART peptides, I (55-102/42-89) and II (62-102/49-89) as well as the intermediate fragments, 10-89 and 33-102. Our subsequent studies have revealed the participation of another PC family member, PC5/6A (the soluble isoform of a widely expressed PC, PC5/6), in cleaving both precursor isoforms. We conclude that PC5/6A contributes to the normal efficient processing of pro-CART and is functionally more redundant with PC2 than PC1/3 in generating both CART I and II.


Assuntos
Proteínas do Tecido Nervoso/metabolismo , Pró-Proteína Convertases/metabolismo , Precursores de Proteínas/metabolismo , Processamento de Proteína Pós-Traducional , Animais , Humanos , Modelos Biológicos , Proteínas do Tecido Nervoso/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Precursores de Proteínas/química
9.
Endocrinology ; 146(2): 713-27, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15528303

RESUMO

To define the biological significance of the initial cleavage at the proglucagon (PG) interdomain site, K70-R71 downward arrow, we created two interdomain mutants, K70Q-R71Q and R71A. Cotransfection studies in GH4C1 cells show significant amounts of glucagon production by PC2 along with some glicentin, glicentin-related polypeptide-glucagon (GRPP-glucagon) and oxyntomodulin from wild-type PG. In contrast, a larger peptide, PG 33-158, and low amounts of GRPP-glucagon are predominantly generated from interdomain mutants. HPLC analysis shows a 5-fold increase in glucagon production by PC2 from wild-type PG and a corresponding 4-fold lower accumulation and secretion of unprocessed precursor relative to interdomain mutants. PC2 generates significant levels of glucagon from a glicentin (PG 1-69) expression plasmid, whereas PC1/3 produces only modest amounts of oxyntomodulin. Employing a major PG fragment (PG 72-158) expression plasmid, we show that PC1/3 predominantly generates glucagon-like peptide (GLP)-1, whereas PC2 produces only N-terminally extended GLP-1. Surprisingly, production of GLP-1 and GLP-2 by PC1/3 from interdomain mutants, compared with wild-type PG, is not significantly impaired. In addition to PC2 and PC1/3, PC5/6A and furin are also able to cleave the sites, K70-R71 downward arrow and R107-X-R-R110 downward arrow in PG. We show a much greater ability of furin to cleave the monobasic site, R77 downward arrow, than at the dibasic site, R124-R125 downward arrow, which is also weakly processed by PC5/6A, indicating overlapping specificities of these two convertases mainly with PC1/3. We propose here a trimer-like model of the spatial organization of the hormonal sequences within the PG molecule in which the accessibility to prohormone convertase action of most cleavage sites is restricted with the exception of the interdomain site, K70-R71, which is maximally accessible.


Assuntos
Glucagon/biossíntese , Glucagon/metabolismo , Pró-Proteína Convertase 2/genética , Pró-Proteína Convertase 2/metabolismo , Precursores de Proteínas/metabolismo , Animais , Cricetinae , Glucagon/química , Peptídeo 1 Semelhante ao Glucagon , Peptídeo 2 Semelhante ao Glucagon , Modelos Moleculares , Mutagênese , Fragmentos de Peptídeos/metabolismo , Peptídeos/metabolismo , Hipófise/citologia , Proglucagon , Pró-Proteína Convertase 2/química , Precursores de Proteínas/química , Estrutura Terciária de Proteína
10.
J Biol Chem ; 279(30): 31068-75, 2004 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-15143067

RESUMO

The endoproteolytic processing of proproteins in the secretory pathway depends on the expression of selected members of a family of subtilisin-like endoproteases known as the prohormone convertases (PCs). The main PC family members expressed in mammalian neuroendocrine cells are PC2 and PC1/3. The differential processing of proglucagon in pancreatic alpha-cells and intestinal L cells leads to production of distinct hormonal products with opposing physiological effects from the same precursor. Here we describe the establishment and characterization of a novel alpha-cell line (alphaTC-DeltaPC2) derived from PC2 homozygous null animals. The alphaTC-DeltaPC2 cells are shown to be similar to the well characterized alphaTC1-6 cell line in both morphology and overall gene expression. However, the absence of PC2 activity in alphaTC-DeltaPC2 leads to a complete block in the production of mature glucagon. Surprisingly, alphaTC-DeltaPC2 cells are able to efficiently cleave the interdomain site in proglucagon (KR 70-71). Further analysis reveals that alphaTC-DeltaPC2 cells, unlike alphaTC1-6 cells, express low levels of PC1/3 that lead to the generation of glicentin as well as low amounts of oxyntomodulin, GLP-1, truncated GLP-1, and N-terminally extended GLP-2. We conclude that alphaTC-DeltaPC2 cells provide additional evidence for PC2 as the major convertase in alpha-cells leading to mature glucagon production and provide a robust model for further analysis of the mechanisms of proprotein processing by the prohormone convertases.


Assuntos
Glucagon/metabolismo , Ilhotas Pancreáticas/metabolismo , Pró-Proteína Convertase 2/deficiência , Precursores de Proteínas/metabolismo , Animais , Sequência de Bases , Linhagem Celular , Primers do DNA/genética , Perfilação da Expressão Gênica , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/enzimologia , Camundongos , Camundongos Knockout , Microscopia Imunoeletrônica , Modelos Biológicos , Análise de Sequência com Séries de Oligonucleotídeos , Proglucagon , Pró-Proteína Convertase 2/genética , Pró-Proteína Convertase 2/metabolismo , Processamento de Proteína Pós-Traducional
11.
Endocrinology ; 145(4): 1961-71, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-14684599

RESUMO

We investigated the proteolytic processing of mouse pro-GHRH [84 amino acids (aa)] by furin, PC1/3, PC2, and PC5/6A. We created six point mutations in the N- and C-terminal cleavage sites, RXXR decreased and RXRXXR decreased, respectively. The following results were obtained after transient transfection/cotransfection and metabolic pulse-chase labeling studies in several neuroendocrine cells. 1) Furin was the most efficient convertase in cleaving the N-terminal RXXR/RXRR site to generate intermediate I, 12-84aa, whereas PC1/3 was the most potent in processing the C-terminal RXRXXR site to yield mature GHRH, 12-53aa. 2) Both PC1/3 and PC5/6A also processed the N-terminal site but less efficiently than furin. 3) PC2 was much weaker in cleaving the C-terminal site relative to PC1/3 to generate mature GHRH. 4) The Q10R mutant was significantly more susceptible to furin cleavage at the N-terminal site than the wild-type pro-GHRH. And 5) the N- and C-terminal P1 Arg residues, R11 and R54, respectively, were essential for mature GHRH production. We also showed localization of the GHRH immunoreactive peptides in Golgi and secretory granules in neuroendocrine cells by an immunofluorescence assay. We conclude that the efficient production of mature GHRH from pro-GHRH is a stepwise process mediated predominantly by furin at the N-terminal cleavage site followed by PC1/3 at the C terminus.


Assuntos
Furina/metabolismo , Hormônio Liberador de Hormônio do Crescimento/metabolismo , Pró-Proteína Convertase 1/metabolismo , Precursores de Proteínas/metabolismo , Processamento de Proteína Pós-Traducional/fisiologia , Animais , Linhagem Celular , Camundongos , Modelos Biológicos , Sistemas Neurossecretores/citologia , Sistemas Neurossecretores/metabolismo , Fragmentos de Peptídeos/metabolismo , Frações Subcelulares/metabolismo , Distribuição Tecidual
12.
J Biol Chem ; 278(17): 15007-14, 2003 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-12584191

RESUMO

Cocaine and amphetamine-regulated transcript (CART), a neuroendocrine peptide influencing reward, feeding/appetite, and stress responses is derived from two peptide precursors of 129 and 116 amino acid (aa) residues that arise via alternative splicing from a single Cart gene in rats and mice. The signal peptide constitutes the first 27 aa resulting in pro-CART molecules of either 102 or 89 aa. In the present study, we have shown that pro-CART is a substrate for the neuroendocrine subtilisin/kexin-like prohormone convertases, PC2 (SPC2) and PC1/3 (SPC3). By using different neuroendocrine cell lines, with or without endogenous expression of either PC2 or PC1/3 or both enzymes, we have demonstrated through transient transfection studies that long pro-CART gives rise to an intermediate peptide, residues 33-102, and the two major bioactive CART forms, residues 55-102 (I) and 62-102 (II), respectively. Likewise, short pro-CART also generates three peptides, an intermediate, residues 10-89, and the two identical bioactive CART forms. We have confirmed the identities of the bioactive and intermediate CART molecules by microsequencing and/or high performance liquid chromatography and mass spectrometry. We have shown that PC2 is more efficient in generating bioactive CART I compared with PC1/3, whereas the production of the smaller bioactive CART II is exclusively carried out by PC2. PC1/3 is predominantly responsible for generating the intermediate CART fragments, 33-102 and 10-89, from long and short pro-CART, respectively. To compare in vitro and in vivo processing of pro-CART, we have examined its processing in PC2, 7B2, and PC1/3 knock-out mouse hypothalamic extracts and demonstrated that, as in vitro, PC2 is more potent than PC1/3 in generating bioactive CART I whereas bioactive CART II is solely generated by PC2. Also, in vivo, we have shown that PC1/3 is predominantly active in liberating the two intermediate CART fragments, 33-102 and 10-89. These findings confirm the key roles of PC2 and PC1/3 acting together or separately to carry out CART processing in selected sites in vivo.


Assuntos
Ácido Aspártico Endopeptidases/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Pró-Proteína Convertase 1 , Processamento de Proteína Pós-Traducional , Subtilisinas/fisiologia , Animais , Ácido Aspártico Endopeptidases/genética , Western Blotting , Linhagem Celular , Cromatografia Líquida de Alta Pressão , Hipotálamo/química , Hipotálamo/citologia , Camundongos , Camundongos Knockout , Pró-Proteína Convertase 2 , Pró-Proteína Convertases , Isoformas de Proteínas/metabolismo , Subtilisinas/genética , Transfecção
13.
Proc Natl Acad Sci U S A ; 99(16): 10293-8, 2002 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-12145326

RESUMO

The subtilisin-like proprotein convertases PC1/3 (SPC3) and PC2 (SPC2) are believed to be the major endoproteolytic processing enzymes of the regulated secretory pathway. They are expressed together or separately in neuroendocrine cells throughout the brain and dispersed endocrine system in both vertebrates and invertebrates. Disruption of the gene-encoding mouse PC1/3 has now been accomplished and results in a syndrome of severe postnatal growth impairment and multiple defects in processing many hormone precursors, including hypothalamic growth hormone-releasing hormone (GHRH), pituitary proopiomelanocortin to adrenocorticotropic hormone, islet proinsulin to insulin and intestinal proglucagon to glucagon-like peptide-1 and -2. Mice lacking PC1/3 are normal at birth, but fail to grow normally and are about 60% of normal size at 10 weeks. They lack mature GHRH, have low pituitary growth hormone (GH) and hepatic insulin-like growth factor-1 mRNA levels and resemble phenotypically the "little" mouse (Gaylinn, B. D., Dealmeida, V. I., Lyons, C. E., Jr., Wu, K. C., Mayo, K. E. & Thorner, M. O. (1999) Endocrinology 140, 5066-5074) that has a mutant GHRH receptor. Despite a severe defect in pituitary proopiomelanocortin processing to mature adrenocorticotropic hormone, blood corticosterone levels are essentially normal. There is marked hyperproinsulinemia but without impairment of glucose tolerance. In contrast, PC2-null mice lack mature glucagon and are chronically hypoglycemic (Furuta, M., Yano, H., Zhou, A., Rouille, Y., Holst, J., Carroll, R., Ravazzola, M., Orci, L., Furuta, H. & Steiner, D. (1997) Proc. Natl. Acad. Sci. USA 94, 6646-6651). The PC1/3-null mice differ from a human subject reported with compound heterozygosity for defects in this gene, who was of normal stature but markedly obese from early life. The PC1/3-null mice are not obese. The basis for these phenotypic differences is an interesting topic for further study. These findings prove the importance of PC1/3 as a key neuroendocrine convertase.


Assuntos
Ácido Aspártico Endopeptidases/metabolismo , Nanismo/enzimologia , Glucagon/metabolismo , Hormônio Liberador de Hormônio do Crescimento/metabolismo , Fragmentos de Peptídeos/metabolismo , Peptídeos/metabolismo , Pró-Opiomelanocortina/metabolismo , Proinsulina/metabolismo , Pró-Proteína Convertase 1 , Precursores de Proteínas/metabolismo , Processamento de Proteína Pós-Traducional , Hormônio Adrenocorticotrópico/metabolismo , Animais , Ácido Aspártico Endopeptidases/genética , Corticosterona/sangue , Nanismo/genética , Expressão Gênica , Marcação de Genes , Peptídeo 1 Semelhante ao Glucagon , Hormônio do Crescimento/genética , Hormônio Liberador de Hormônio do Crescimento/genética , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso , Sistemas Neurossecretores/metabolismo , Proglucagon , Pró-Proteína Convertases
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...