Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Cell Sci ; 135(5)2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-34981808

RESUMO

High-density lipoproteins (HDLs) prevent cell death induced by a variety of cytotoxic drugs. The underlying mechanisms are however still poorly understood. Here, we present evidence that HDLs efficiently protect cells against thapsigargin (TG), a sarco/endoplasmic reticulum (ER) Ca2+-ATPase (SERCA) inhibitor, by extracting the drug from cells. Drug efflux could also be triggered to some extent by low-density lipoproteins and serum. HDLs did not reverse the non-lethal mild ER stress response induced by low TG concentrations or by SERCA knockdown, but HDLs inhibited the toxic SERCA-independent effects mediated by high TG concentrations. HDLs could extract other lipophilic compounds, but not hydrophilic substances. This work shows that HDLs utilize their capacity of loading themselves with lipophilic compounds, akin to their ability to extract cellular cholesterol, to reduce the cell content of hydrophobic drugs. This can be beneficial if lipophilic xenobiotics are toxic but may be detrimental to the therapeutic benefit of lipophilic drugs such as glibenclamide.


Assuntos
Lipoproteínas HDL , Preparações Farmacêuticas , Cálcio/metabolismo , Retículo Endoplasmático/metabolismo , Estresse do Retículo Endoplasmático , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , Tapsigargina/farmacologia
2.
FEBS J ; 289(3): 808-831, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34528388

RESUMO

The human genome contains 11 APOBEC (apolipoprotein B mRNA editing catalytic polypeptide-like) cytidine deaminases classified into four families. These proteins function mainly in innate antiviral immunity and can also restrict endogenous retrotransposable element multiplication. The present study focuses on APOBEC3C (A3C), a member of the APOBEC3 subfamily. Some APOBEC3 proteins use their enzymatic activity on genomic DNA, inducing mutations and DNA damage, while other members facilitate DNA repair. Our results show that A3C is highly expressed in cells treated with DNA-damaging agents. Its expression is regulated by p53. Depletion of A3C slightly decreases proliferation and does not affect DNA repair via homologous recombination or nonhomologous end joining. The A3C interactomes obtained from control cells and cells exposed to the genotoxin etoposide indicated that A3C is a nucleolar protein. This was confirmed by the detection of either endogenous or ectopic A3C in nucleoli. Interestingly, we show that A3C is excluded from areas of DNA breaks in live cells. Our data also indicate that the C-terminal part of A3C is responsible for its nucleolar localization and exclusion from DNA damage sites.


Assuntos
Citidina Desaminase/genética , Reparo do DNA por Junção de Extremidades/genética , Recombinação Homóloga/genética , Proteína Supressora de Tumor p53/genética , Nucléolo Celular/genética , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/genética , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/genética , Etoposídeo/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Genoma Humano/genética , Humanos , Família Multigênica/genética , Mutagênicos/farmacologia , Mutação/genética
3.
Elife ; 102021 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-34713805

RESUMO

Cell-penetrating peptides (CPPs) allow intracellular delivery of bioactive cargo molecules. The mechanisms allowing CPPs to enter cells are ill-defined. Using a CRISPR/Cas9-based screening, we discovered that KCNQ5, KCNN4, and KCNK5 potassium channels positively modulate cationic CPP direct translocation into cells by decreasing the transmembrane potential (Vm). These findings provide the first unbiased genetic validation of the role of Vm in CPP translocation in cells. In silico modeling and live cell experiments indicate that CPPs, by bringing positive charges on the outer surface of the plasma membrane, decrease the Vm to very low values (-150 mV or less), a situation we have coined megapolarization that then triggers formation of water pores used by CPPs to enter cells. Megapolarization lowers the free energy barrier associated with CPP membrane translocation. Using dyes of varying dimensions in CPP co-entry experiments, the diameter of the water pores in living cells was estimated to be 2 (-5) nm, in accordance with the structural characteristics of the pores predicted by in silico modeling. Pharmacological manipulation to lower transmembrane potential boosted CPP cellular internalization in zebrafish and mouse models. Besides identifying the first proteins that regulate CPP translocation, this work characterized key mechanistic steps used by CPPs to cross cellular membranes. This opens the ground for strategies aimed at improving the ability of cells to capture CPP-linked cargos in vitro and in vivo.


Before a drug can have its desired effect, it must reach its target tissue or organ, and enter its cells. This is not easy because cells are surrounded by the plasma membrane, a fat-based barrier that separates the cell from its external environment. The plasma membrane contains proteins that act as channels, shuttling specific molecules in and out of the cell, and it also holds charge, with its inside surface being more negatively charged than its outside surface. Cell-penetrating peptides are short sequences of amino acids (the building blocks that form proteins) that carry positive charges. These positive charges allow them to cross the membrane easily, but it is not well understood how. To find out how cell-penetrating peptides cross the membrane, Trofimenko et al. attached them to dyes of different sizes. This revealed that the cell-penetrating peptides enter the cell through temporary holes called water pores, which measure about two nanometres across. The water pores form when the membrane becomes 'megapolarized', this is, when the difference in charge between the inside and the outside of the membrane becomes greater than normal. This can happen when the negative charge on the inside surface or the positive charge on the outer surface of the membrane increase. Megapolarization depends on potassium channels, which transport positive potassium ions outside the cell, making the outside of the membrane positive. When cell-penetrating peptides arrive at the outer surface of the cell near potassium channels, they make it even more positive. This increases the charge difference between the inside and the outside of the cell, allowing water pores to form. Once the peptides pass through the pores, the charge difference between the inside and the outside of the cell membrane dissipates, and the pores collapse. Drug developers are experimenting with attaching cell-penetrating peptides to drugs to help them get inside their target cells. Currently there are several experimental medications of this kind in clinical trials. Understanding how these peptides gain entry, and what size of molecule they could carry with them, provides solid ground for further drug development.


Assuntos
Peptídeos Penetradores de Células/genética , Canais de Potássio/genética , Animais , Linhagem Celular , Peptídeos Penetradores de Células/química , Peptídeos Penetradores de Células/metabolismo , Células HeLa , Humanos , Potenciais da Membrana , Camundongos , Camundongos Endogâmicos C57BL , Canais de Potássio/metabolismo , Transporte Proteico , Ratos , Ratos Sprague-Dawley , Peixe-Zebra
4.
Sci Rep ; 10(1): 8354, 2020 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-32415214

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

5.
Sci Rep ; 10(1): 6413, 2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32286361

RESUMO

Long non-coding RNAs (lncRNAs) contribute to diverse cellular functions and the dysregulation of their expression or function can contribute to diseases, including diabetes. The contributions of lncRNAs to ß-cell development, function and survival has been extensively studied in vitro. However, very little is currently known on the in vivo roles of lncRNAs in the regulation of glucose and insulin homeostasis. Here we investigated the impact of loss-of-function in mice of the lncRNA A830019P07Rik, hereafter P07Rik, which was previously reported to be associated with reduced plasma insulin levels. Compared with wild-type littermates, male and female P07Rik mutant mice did not show any defect in glycaemia and plasma insulin levels in both fed and fasted state. Furthermore, P07Rik mutant mice displayed similar glucose and insulin levels in response to an intra-peritoneal glucose tolerance test. Ex vivo, islets from mutant P07Rik released similar amount of insulin in response to increased glucose concentration as wildtype littermates. In contrast with previous reports, our characterization of P07Rik mouse mutants revealed that loss of function of this lncRNA does not affect glucose and insulin homeostasis in mice.


Assuntos
Secreção de Insulina/genética , Insulina/metabolismo , RNA Longo não Codificante/metabolismo , Animais , Glicemia/metabolismo , Peso Corporal , Sequência Conservada/genética , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/genética , Regulação para Baixo/genética , Jejum/sangue , Comportamento Alimentar , Feminino , Homeostase , Insulina/sangue , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos Obesos , RNA Longo não Codificante/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
6.
Cell Signal ; 62: 109347, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31229616

RESUMO

High-density lipoproteins (HDLs) can protect cells against a variety of death-inducing stresses. This is often accompanied by activation of the anti-apoptotic Akt kinase but whether this activation mediates the protective functions of HDLs is still unclear. In this study, we evaluated the roles of PI3K/Akt signaling in endoplasmic reticulum (ER) stress- and starvation-induced cell death using pharmacological and genetic approaches to gain a better understanding of the relationship between Akt- and HDL-mediated protection. Three cell models were used for this purpose, a primary endothelial cell line, an insulinoma cell line and a colon adenocarcinoma cell line. Our results show that HDLs indeed elicited mild Akt activation in all the tested cellular models. PI3K is one of the main upstream proteins involved in Akt stimulation. In the three cellular models, LY294002, a PI3K inhibitor, only slightly blunted HDLs protection, indicating that HDLs induce PI3K-independent cell protection. Furthermore, genetic ablation or silencing of Akt did not abolish the protective effects of HDLs. This study demonstrates that the PI3K-Akt signaling pathway is not the main mediator of the cell protective functions of HDLs. Further investigation is therefore needed to identify the intrinsic mechanism of HDL-mediated cell protection.


Assuntos
Citoproteção/genética , Lipoproteínas HDL/genética , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , Apoptose/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Cromonas/farmacologia , Citoproteção/efeitos dos fármacos , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/genética , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Humanos , Lipoproteínas HDL/antagonistas & inibidores , Morfolinas/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição CHOP
7.
Oncotarget ; 10(66): 7058-7070, 2019 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-31903165

RESUMO

Sorafenib is the first-line drug used for patients with advanced hepatocellular carcinoma (HCC). However, acquired sorafenib resistance in cancer patients limits its efficacy. Here, we performed the first genome-wide CRISPR/Cas9-based screening on sorafenib-treated HCC cells to identify essential genes for non-mutational mechanisms related to acquired sorafenib resistance and/or sensitivity in HCC cells. KEAP1 was identified as the top candidate gene by Model-based Analysis of Genome-wide CRISPR/Cas9 Knockout (MAGeCK). KEAP1 disrupted HCC cells were less sensitive than wild-type cells in short- and long-term sorafenib treatments. Compared to wild-type cells, KEAP1-disrupted cells showed lower basal and sorafenib-induced reactive oxygen species (ROS) levels and were more resistant to oxidative stress-induced cell death. The absence of KEAP1 led to increased activity of Nrf2, a key transcription factor controlling antioxidant responses, as further evidenced by increased expression of Nrf2-controlled genes including NQO1, GPX2 and TXNRD1, which were positively associated with chemoresistance. In addition, KEAP1 disruption counteracted the reduction of cell viability and the elevation of ROS caused by lenvatinib, a drug that recently showed clinical efficacy as a first-line treatment for unresectable HCC. Finally, Keap1 disruption also increased the resistance of cells to regorafenib, a recently approved drug to treat HCC as a second line therapy. Taken together, our data indicate that deregulation of the KEAP1/Nrf2 pathway following KEAP1 inactivation contributes to sorafenib, lenvatinib, and regorafenib resistance in human HCC cells through up-regulation of Nrf2 downstream genes and decreased ROS levels.

8.
Cell Signal ; 26(12): 2992-7, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25246356

RESUMO

The caspase-3/p120 RasGAP module acts as a stress sensor that promotes pro-survival or pro-death signaling depending on the intensity and the duration of the stressful stimuli. Partial cleavage of p120 RasGAP generates a fragment, called fragment N, which protects stressed cells by activating Akt signaling. Akt family members regulate many cellular processes including proliferation, inhibition of apoptosis and metabolism. These cellular processes are regulated by three distinct Akt isoforms: Akt1, Akt2 and Akt3. However, which of these isoforms are required for fragment N mediated protection have not been defined. In this study, we investigated the individual contribution of each isoform in fragment N-mediated cell protection against Fas ligand induced cell death. To this end, DLD1 and HCT116 isogenic cell lines lacking specific Akt isoforms were used. It was found that fragment N could activate Akt1 and Akt2 but that only the former could mediate the protective activity of the RasGAP-derived fragment. Even overexpression of Akt2 or Akt3 could not rescue the inability of fragment N to protect cells lacking Akt1. These results demonstrate a strict Akt isoform requirement for the anti-apoptotic activity of fragment N.


Assuntos
Apoptose , Caspase 3/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Estresse Fisiológico , Proteína p120 Ativadora de GTPase/metabolismo , Sobrevivência Celular , Técnicas de Inativação de Genes , Células HCT116 , Humanos , Isoenzimas/metabolismo , Fosforilação , Especificidade por Substrato
9.
Mol Cell Endocrinol ; 381(1-2): 291-301, 2013 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-23994023

RESUMO

HDLs protect pancreatic beta cells against apoptosis induced by several endoplasmic reticulum (ER) stressors, including thapsigargin, cyclopiazonic acid, palmitate and insulin over-expression. This protection is mediated by the capacity of HDLs to maintain proper ER morphology and ER functions such as protein folding and trafficking. Here, we identified a distinct mode of protection exerted by HDLs in beta cells challenged with tunicamycin (TM), a protein glycosylation inhibitor inducing ER stress. HDLs were found to inhibit apoptosis induced by TM in the MIN6 insulinoma cell line and this correlated with the maintenance of a normal ER morphology. Surprisingly however, this protective response was neither associated with a significant ER stress reduction, nor with restoration of protein folding and trafficking in the ER. These data indicate that HDLs can use at least two mechanisms to protect beta cells against ER stressors. One that relies on the maintenance of ER function and one that operates independently of ER function modulation. The capacity of HDLs to activate several anti-apoptotic pathways in beta cells may explain their ability to efficiently protect these cells against a variety of insults.


Assuntos
Apoptose/efeitos dos fármacos , Estresse do Retículo Endoplasmático , Retículo Endoplasmático/fisiologia , Lipoproteínas HDL/fisiologia , Tunicamicina/farmacologia , Animais , Linhagem Celular Tumoral , Citoproteção , Proteínas de Ligação a DNA/metabolismo , Retículo Endoplasmático/ultraestrutura , Chaperona BiP do Retículo Endoplasmático , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Células HEK293 , Proteínas de Choque Térmico/metabolismo , Humanos , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/fisiologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Sistema de Sinalização das MAP Quinases , Camundongos , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fatores de Transcrição de Fator Regulador X , Tapsigargina/farmacologia , Fator de Transcrição CHOP/metabolismo , Fatores de Transcrição/metabolismo
10.
Mol Cell Biol ; 32(22): 4523-33, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22949508

RESUMO

The ability to generate appropriate defense responses is crucial for the survival of an organism exposed to pathogenesis-inducing insults. However, the mechanisms that allow tissues and organs to cope with such stresses are poorly understood. Here we show that caspase-3-knockout mice or caspase inhibitor-treated mice were defective in activating the antiapoptotic Akt kinase in response to various chemical and environmental stresses causing sunburns, cardiomyopathy, or colitis. Defective Akt activation in caspase-3-knockout mice was accompanied by increased cell death and impaired survival in some cases. Mice homozygous for a mutation in RasGAP that prevents its cleavage by caspase-3 exhibited a similar defect in Akt activation, leading to increased apoptosis in stressed organs, marked deterioration of their physiological functions, and stronger disease development. Our results provide evidence for the relevance of caspase-3 as a stress intensity sensor that controls cell fate by either initiating a RasGAP cleavage-dependent cell resistance program or a cell suicide response.


Assuntos
Cardiomiopatias/enzimologia , Caspase 3/genética , Colite/enzimologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Queimadura Solar/enzimologia , Proteína p120 Ativadora de GTPase/genética , Animais , Sequência de Bases , Cardiomiopatias/induzido quimicamente , Cardiomiopatias/genética , Caspase 3/deficiência , Morte Celular/efeitos dos fármacos , Morte Celular/efeitos da radiação , Colite/induzido quimicamente , Colite/genética , Sulfato de Dextrana , Doxorrubicina , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos da radiação , Hemodinâmica , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Mutação , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transdução de Sinais/efeitos da radiação , Estresse Fisiológico , Queimadura Solar/genética , Raios Ultravioleta , Proteína p120 Ativadora de GTPase/antagonistas & inibidores , Proteína p120 Ativadora de GTPase/deficiência
11.
PLoS One ; 6(7): e22609, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21799917

RESUMO

The caspase-3-generated RasGAP N-terminal fragment (fragment N) inhibits apoptosis in a Ras-PI3K-Akt-dependent manner. Fragment N protects various cell types, including insulin-secreting cells, against different types of stresses. Whether fragment N exerts a protective role during the development of type 1 diabetes is however not known. Non-obese diabetic (NOD) mice represent a well-known model for spontaneous development of type 1 diabetes that shares similarities with the diseases encountered in humans. To assess the role of fragment N in type 1 diabetes development, a transgene encoding fragment N under the control of the rat insulin promoter (RIP) was back-crossed into the NOD background creating the NOD-RIPN strain. Despite a mosaic expression of fragment N in the beta cell population of NOD-RIPN mice, islets isolated from these mice were more resistant to apoptosis than control NOD islets. Islet lymphocytic infiltration and occurrence of a mild increase in glycemia developed with the same kinetics in both strains. However, the period of time separating the mild increase in glycemia and overt diabetes was significantly longer in NOD-RIPN mice compared to the control NOD mice. There was also a significant decrease in the number of apoptotic beta cells in situ at 16 weeks of age in the NOD-RIPN mice. Fragment N exerts therefore a protective effect on beta cells within the pro-diabetogenic NOD background and this prevents a fast progression from mild to overt diabetes.


Assuntos
Apoptose/efeitos dos fármacos , Diabetes Mellitus/patologia , Progressão da Doença , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/patologia , Fragmentos de Peptídeos/farmacologia , Proteínas Ativadoras de ras GTPase/química , Animais , Autoimunidade/efeitos dos fármacos , Linhagem Celular Tumoral , Diabetes Mellitus/imunologia , Diabetes Mellitus/metabolismo , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Feminino , Regulação da Expressão Gênica , Células Secretoras de Insulina/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Fragmentos de Peptídeos/metabolismo , Ratos , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
12.
Diabetes ; 58(11): 2596-606, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19696184

RESUMO

OBJECTIVE: Our laboratory has previously established in vitro that a caspase-generated RasGAP NH(2)-terminal moiety, called fragment N, potently protects cells, including insulinomas, from apoptotic stress. We aimed to determine whether fragment N can increase the resistance of pancreatic beta-cells in a physiological setting. RESEARCH DESIGN AND METHODS: A mouse line, called rat insulin promoter (RIP)-N, was generated that bears a transgene containing the rat insulin promoter followed by the cDNA-encoding fragment N. The histology, functionality, and resistance to stress of RIP-N islets were then assessed. RESULTS: Pancreatic beta-cells of RIP-N mice express fragment N, activate Akt, and block nuclear factor kappaB activity without affecting islet cell proliferation or the morphology and cellular composition of islets. Intraperitoneal glucose tolerance tests revealed that RIP-N mice control their glycemia similarly as wild-type mice throughout their lifespan. Moreover, islets isolated from RIP-N mice showed normal glucose-induced insulin secretory capacities. They, however, displayed increased resistance to apoptosis induced by a series of stresses including inflammatory cytokines, fatty acids, and hyperglycemia. RIP-N mice were also protected from multiple low-dose streptozotocin-induced diabetes, and this was associated with reduced in vivo beta-cell apoptosis. CONCLUSIONS: Fragment N efficiently increases the overall resistance of beta-cells to noxious stimuli without interfering with the physiological functions of the cells. Fragment N and the pathway it regulates represent, therefore, a potential target for the development of antidiabetes tools.


Assuntos
Diabetes Mellitus Experimental/prevenção & controle , Células Secretoras de Insulina/fisiologia , Fragmentos de Peptídeos/genética , Proteínas Ativadoras de ras GTPase/genética , Animais , Apoptose , Glicemia/metabolismo , Encéfalo/fisiologia , Encéfalo/fisiopatologia , Divisão Celular/genética , DNA Complementar/genética , Diabetes Mellitus Experimental/fisiopatologia , Teste de Tolerância a Glucose , Insulina/genética , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Fragmentos de Peptídeos/farmacologia , Regiões Promotoras Genéticas , Ratos
13.
Exp Cell Res ; 315(12): 2081-91, 2009 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-19328779

RESUMO

The specific sensitization of tumor cells to the apoptotic response induced by genotoxins is a promising way of increasing the efficacy of chemotherapies. The RasGAP-derived fragment N2, while not regulating apoptosis in normal cells, potently sensitizes tumor cells to cisplatin- and other genotoxin-induced cell death. Here we show that fragment N2 in living cells is mainly located in the cytoplasm and only minimally associated with specific organelles. The cytoplasmic localization of fragment N2 was required for its cisplatin-sensitization property because targeting it to the mitochondria or the ER abrogated its ability to increase the death of tumor cells in response to cisplatin. These results indicate that fragment N2 requires a spatially constrained cellular location to exert its anti-cancer activity.


Assuntos
Apoptose , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Proteínas Ativadoras de ras GTPase/metabolismo , Motivos de Aminoácidos , Animais , Antineoplásicos/farmacologia , Células Cultivadas , Cisplatino/farmacologia , Humanos , Camundongos , Proteínas Ativadoras de ras GTPase/genética
14.
Cell Signal ; 19(10): 2201-7, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17669625

RESUMO

Islet-Brain 1 (IB1) (also called JNK-interacting protein 1; JIP1) is a scaffold protein that tethers components of the JNK mitogen-activated protein kinase pathway inducing a modulation of the activity and the target specificity of the JNK kinases. Dysfunctions in IB1 have been associated with diseases such as early type II diabetes. To gain more insight in the functions of IB1, its ability to modulate the expression levels of the various JNK proteins was assessed. Each of the three JNK genes gives rise to several splice variants encoding short or long proteins. The expression levels of the short JNK proteins, but not of the long variants, were systematically higher in rat tissues and in transformed cell lines expressing high IB1 levels compared to tissues and cells with no or low IB1 expression. HEK293 cells bearing a tetracycline-inducible IB1 construct showed a specific increase of the short JNK endogenous splice variants in the presence of tetracycline. The augmented expression level of the short JNK splice variants induced by IB1 resulted from an increased stability towards degradation. Modulation of the stability of specific JNK splice variants represents therefore a newly identified mechanism used by IB1 to regulate the JNK MAPK pathway.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Processamento Alternativo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Animais , Linhagem Celular , Linhagem Celular Transformada , Estabilidade Enzimática , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Proteína Quinase 8 Ativada por Mitógeno/genética , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Proteína Quinase 9 Ativada por Mitógeno/genética , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Ratos
15.
Mol Biol Cell ; 16(8): 3511-20, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15901831

RESUMO

RasGAP bears two caspase-3 cleavage sites that are used sequentially as caspase activity increases in cells. When caspase-3 is mildly activated, RasGAP is first cleaved at position 455. This leads to the production of an N-terminal fragment, called fragment N, that activates the Ras-PI3K-Akt pathway and that promotes cell survival. At higher caspase activity, RasGAP is further cleaved at position 157 generating two small N-terminal fragments named N1 and N2. We have now determined the contribution of this second cleavage event in the regulation of apoptosis using cells in which the wild-type RasGAP gene has been replaced by a cDNA encoding a RasGAP mutant that cannot be cleaved at position 157. Our results show that cleavage of fragment N at position 157 leads to a marked reduction in Akt activity. This is accompanied by efficient processing of caspase-3 that favors cell death in response to various apoptotic stimuli. In nontumorigenic cells, fragments N1 and N2 do not modulate apoptosis. Therefore, the role of the second caspase-mediated cleavage of RasGAP is to allow the inactivation of the antiapoptotic function of fragment N so that caspases are no longer hampered in their ability to kill cells.


Assuntos
Apoptose , Caspases/metabolismo , Mutação/genética , Proteínas Ativadoras de ras GTPase/genética , Proteínas Ativadoras de ras GTPase/metabolismo , Animais , Ácido Aspártico/genética , Ácido Aspártico/metabolismo , Caspase 3 , Linhagem Celular , Regulação para Baixo , Ativação Enzimática , Fibroblastos , Camundongos , Camundongos Knockout , Proteínas Ativadoras de ras GTPase/deficiência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...