Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
J Neurochem ; 144(2): 139-151, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29164620

RESUMO

Therapy of malignant glioma relies on treatment with the O6 -methylating agent temozolomide (TMZ) concomitant with ionizing radiation followed by adjuvant TMZ. For the treatment of recurrences, DNA chloroethylating drugs are also used. The main killing lesion induced by these drugs is O6 -alkylguanine. Since this damage is repaired by O6 -methylguanine-DNA methyltransferase (MGMT), the repair enzyme represents a most important factor of drug resistance, limiting the therapy of malignant high-grade gliomas. Although MGMT has been shown to be transcriptionally up-regulated in rodents following genotoxic stress, it is still unclear whether human MGMT is subject to up-regulation. Here, we addressed the question whether MGMT in glioma cells is enhanced following alkylating drugs or ionizing radiation, using promoter assays. We also checked the response of glioma cell lines to dexamethasone. In a series of experiments, we found no evidence that the human MGMT promoter is significantly up-regulated following treatment with TMZ, the chloroethylating agent nimustine or radiation. It was activated, however, by dexamethasone. Using deletion constructs, we further show that the basal level of MGMT is mainly determined by the transcription factor SP1. The high amount of SP1 sites in the MGMT promoter likely prevents transcriptional up-regulation following genotoxic stress by neutralizing inducible signals. The regulation of MGMT by miRNAs plays only a minor role, as shown by DICER knockdown experiments. Since high dose dexamethasone concomitant with temozolomide is frequently used in glioblastoma therapy, induction of the MGMT gene through glucocorticoids in MGMT promoter unmethylated cases might cause further elevation of drug resistance, while radiation and alkylating drugs seem not to induce MGMT at transcriptional level.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Enzimas Reparadoras do DNA/genética , Glucocorticoides/farmacologia , O(6)-Metilguanina-DNA Metiltransferase/genética , Fator de Transcrição Sp1/genética , Temozolomida/farmacologia , Enzimas Reparadoras do DNA/efeitos dos fármacos , Enzimas Reparadoras do DNA/efeitos da radiação , Dexametasona/farmacologia , Indução Enzimática/efeitos dos fármacos , Indução Enzimática/efeitos da radiação , Técnicas de Silenciamento de Genes , Humanos , O(6)-Metilguanina-DNA Metiltransferase/efeitos dos fármacos , O(6)-Metilguanina-DNA Metiltransferase/efeitos da radiação , Regiões Promotoras Genéticas/genética , RNA Mensageiro/farmacologia , Fator de Transcrição Sp1/efeitos dos fármacos , Fator de Transcrição Sp1/efeitos da radiação , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/efeitos da radiação
2.
Nucleic Acids Res ; 44(22): 10727-10743, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27694624

RESUMO

A coordinated and faithful DNA damage response is of central importance for maintaining genomic integrity and survival. Here, we show that exposure of human cells to benzo(a)pyrene 9,10-diol-7,8-epoxide (BPDE), the active metabolite of benzo(a)pyrene (B(a)P), which represents a most important carcinogen formed during food preparation at high temperature, smoking and by incomplete combustion processes, causes a prompt and sustained upregulation of the DNA repair genes DDB2, XPC, XPF, XPG and POLH. Induction of these repair factors on RNA and protein level enhanced the removal of BPDE adducts from DNA and protected cells against subsequent BPDE exposure. However, through the induction of POLH the mutation frequency in the surviving cells was enhanced. Activation of these adaptive DNA repair genes was also observed upon B(a)P treatment of MCF7 cells and in buccal cells of human volunteers after cigarette smoking. Our data provide a rational basis for an adaptive response to polycyclic aromatic hydrocarbons, which occurs however at the expense of mutations that may drive cancer formation.


Assuntos
Apoptose , Reparo do DNA , Ativação Transcricional , 7,8-Di-Hidro-7,8-Di-Hidroxibenzo(a)pireno 9,10-óxido/farmacologia , Sobrevivência Celular , Adutos de DNA/genética , Adutos de DNA/metabolismo , Dano ao DNA , DNA Polimerase Dirigida por DNA/genética , DNA Polimerase Dirigida por DNA/metabolismo , Retroalimentação Fisiológica , Humanos , Células MCF-7 , Mutagênicos/farmacologia , Fator de Transcrição AP-1/fisiologia , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima
3.
Biochim Biophys Acta ; 1833(8): 1832-43, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23578789

RESUMO

To counteract genotoxic stress, DNA repair functions are in effect. Most of them are constitutively expressed while some of them can be up-regulated depending on the level of DNA damage. In human cells, only few DNA repair functions are subject of induction following DNA damage, and thus there is a need to identify and characterize inducible repair functions more thoroughly. Here, we provide evidence that the "three prime exonuclease I" (TREX1) is up-regulated in human fibroblasts and cancer cells on mRNA and protein level. Transcriptional upregulation of TREX1 was observed upon exposure to ultraviolet light and various anticancer drugs in glioma and malignant melanoma cells. Induction of TREX1 was found following treatment with the crosslinking alkylating agents nimustine, carmustine, fotemustine and the topoisomerase I inhibitor topotecan, but not following temozolomide, etoposide and ionizing radiation. Induction of TREX1 following DNA damage requires the AP-1 components c-Jun and c-Fos, as shown by siRNA knockdown, EMSA experiments, ChIP analysis and reporter assays with the TREX1 promoter and constructs harboring mutations in the AP-1 binding site. To analyze whether TREX1 expression impacts the sensitivity of cancer cells to therapeutics, TREX1 expression was down-regulated by siRNA in malignant glioma and melanoma cells. TREX1 knockdown resulted in enhanced cell death following nimustine, fotemustine and topotecan and to a reduced recovery from the anticancer drug induced block to replication. The data revealed that induction of TREX1 is a survival response evoked by various genotoxic anticancer drugs and identified TREX1 as a potential therapeutic target for anticancer therapy.


Assuntos
Antineoplásicos/farmacologia , Dano ao DNA/genética , Exodesoxirribonucleases/genética , Glioma/genética , Melanoma/genética , Fosfoproteínas/genética , Sítios de Ligação/efeitos dos fármacos , Sítios de Ligação/genética , Morte Celular/efeitos dos fármacos , Morte Celular/genética , Linhagem Celular , Linhagem Celular Tumoral , Dano ao DNA/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , Replicação do DNA/genética , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Resistencia a Medicamentos Antineoplásicos , Exodesoxirribonucleases/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes jun/genética , Glioma/tratamento farmacológico , Glioma/metabolismo , Humanos , Melanoma/tratamento farmacológico , Melanoma/metabolismo , Mutação/efeitos dos fármacos , Fosfoproteínas/metabolismo , Regiões Promotoras Genéticas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , RNA Mensageiro/genética , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo , Ativação Transcricional/efeitos dos fármacos , Ativação Transcricional/genética , Regulação para Cima/efeitos dos fármacos
4.
Nucleic Acids Res ; 38(19): 6418-32, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20511593

RESUMO

Cells respond to genotoxic stress with the induction of DNA damage defence functions. Aimed at identifying novel players in this response, we analysed the genotoxic stress-induced expression of DNA repair genes in mouse fibroblasts proficient and deficient for c-Fos or c-Jun. The experiments revealed a clear up-regulation of the three prime exonuclease I (trex1) mRNA following ultraviolet (UV) light treatment. This occurred in the wild-type but not c-fos and c-jun null cells, indicating the involvement of AP-1 in trex1 induction. Trex1 up-regulation was also observed in human cells and was found on promoter, RNA and protein level. Apart from UV light, TREX1 is induced by other DNA damaging agents such as benzo(a)pyrene and hydrogen peroxide. The mouse and human trex1 promoter harbours an AP-1 binding site that is recognized by c-Fos and c-Jun, and its mutational inactivation abrogated trex1 induction. Upon genotoxic stress, TREX1 is not only up-regulated but also translocated into the nucleus. Cells deficient in TREX1 show reduced recovery from the UV and benzo(a)pyrene-induced replication inhibition and increased sensitivity towards the genotoxins compared to the isogenic control. The data revealed trex1 as a novel DNA damage-inducible repair gene that plays a protective role in the genotoxic stress response.


Assuntos
Benzo(a)pireno/toxicidade , Dano ao DNA , Exodesoxirribonucleases/genética , Mutagênicos/toxicidade , Fosfoproteínas/genética , Proteínas Proto-Oncogênicas c-fos/fisiologia , Raios Ultravioleta/efeitos adversos , Transporte Ativo do Núcleo Celular , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Replicação do DNA , Exodesoxirribonucleases/biossíntese , Técnicas de Inativação de Genes , Humanos , Camundongos , Fosfoproteínas/biossíntese , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-fos/genética , RNA Mensageiro/biossíntese , Estresse Fisiológico/genética , Fator de Transcrição AP-1/metabolismo , Ativação Transcricional
5.
Cancer Res ; 79(1): 99-113, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30361254

RESUMO

The DNA-methylating drug temozolomide, which induces cell death through apoptosis, is used for the treatment of malignant glioma. Here, we investigate the mechanisms underlying the ability of temozolomide to induce senescence in glioblastoma cells. Temozolomide-induced senescence was triggered by the specific DNA lesion O6-methylguanine (O6MeG) and characterized by arrest of cells in the G2-M phase. Inhibitor experiments revealed that temozolomide-induced senescence was initiated by damage recognition through the MRN complex, activation of the ATR/CHK1 axis of the DNA damage response pathway, and mediated by degradation of CDC25c. Temozolomide-induced senescence required functional p53 and was dependent on sustained p21 induction. p53-deficient cells, not expressing p21, failed to induce senescence, but were still able to induce a G2-M arrest. p14 and p16, targets of p53, were silenced in our cell system and did not seem to play a role in temozolomide-induced senescence. In addition to p21, the NF-κB pathway was required for senescence, which was accompanied by induction of the senescence-associated secretory phenotype. Upon temozolomide exposure, we found a strong repression of the mismatch repair proteins MSH2, MSH6, and EXO1 as well as the homologous recombination protein RAD51, which was downregulated by disruption of the E2F1/DP1 complex. Repression of these repair factors was not observed in G2-M arrested p53-deficient cells and, therefore, it seems to represent a specific trait of temozolomide-induced senescence. SIGNIFICANCE: These findings reveal a mechanism by which the anticancer drug temozolomide induces senescence and downregulation of DNA repair pathways in glioma cells.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Quinase 1 do Ponto de Checagem/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Enzimas Reparadoras do DNA/antagonistas & inibidores , Reparo do DNA/efeitos dos fármacos , Glioblastoma/patologia , NF-kappa B/metabolismo , Temozolomida/farmacologia , Animais , Antineoplásicos Alquilantes/farmacologia , Apoptose , Proteínas Mutadas de Ataxia Telangiectasia/genética , Ciclo Celular , Proliferação de Células , Senescência Celular , Quinase 1 do Ponto de Checagem/genética , Inibidor de Quinase Dependente de Ciclina p21/genética , Metilação de DNA , Enzimas Reparadoras do DNA/genética , Enzimas Reparadoras do DNA/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , NF-kappa B/genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Nucleic Acids Res ; 34(22): 6530-9, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17130154

RESUMO

Cells deficient in c-Fos are hypersensitive to ultraviolet (UV-C) light. Here we demonstrate that mouse embryonic fibroblasts lacking c-Fos (fos-/-) are defective in the repair of UV-C induced DNA lesions. They show a decreased rate of sealing of repair-mediated DNA strand breaks and are unable to remove cyclobutane pyrimidine dimers from DNA. A search for genes responsible for the DNA repair defect revealed that upon UV-C treatment the level of xpf and xpg mRNA declined but, in contrast to the wild type (wt), did not recover in fos-/- cells. The observed decline in xpf and xpg mRNA is due to impaired re-synthesis, as shown by experiments using actinomycin D. Block of xpf transcription resulted in a lack of XPF protein after irradiation of fos-/- cells, whereas the XPF level normalized quickly in the wt. Although the xpg mRNA level was reduced, the amount of XPG protein was not altered in c-Fos-deficient cells after UV-C, due to higher stability of the XPG protein. The data suggest a new role for c-Fos in cells exposed to genotoxic stress. Being part of the transcription factor AP-1, c-Fos stimulates NER via the upregulation of xpf and thus plays a central role in the recovery of cells from UV light induced DNA damage.


Assuntos
Dano ao DNA , Reparo do DNA , Proteínas de Ligação a DNA/biossíntese , Genes fos/fisiologia , Raios Ultravioleta , Animais , Linhagem Celular , DNA/química , DNA/metabolismo , DNA/efeitos da radiação , Quebras de DNA de Cadeia Simples , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Endonucleases/genética , Endonucleases/metabolismo , Expressão Gênica , Genes fos/genética , Teste de Complementação Genética , Camundongos , Camundongos Knockout , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Dímeros de Pirimidina/metabolismo , Estabilidade de RNA , RNA Mensageiro/metabolismo , Fator de Transcrição AP-1/fisiologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
7.
J Mol Biol ; 315(4): 637-46, 2002 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-11812136

RESUMO

To elucidate the function of the two cytokine-binding modules (CBM) of the leukemia inhibitory factor receptor (LIFR), receptor chimeras of LIFR and the interleukin-6 receptor (IL-6R) were constructed. Either the NH(2)-terminal (chimera RILLIFdeltaI) or the COOH-terminal LIFR CBM (chimera RILLIFdeltaII) were replaced by the structurally related CBM of the IL-6R which does not bind LIF. Chimera RILLIFdeltaI is functionally inactive, whereas RILLIFdeltaII binds LIF and mediates signalling as efficiently as the wild-type LIFR. Deletion mutants of the LIFR revealed that both the NH(2)-terminal CBM and the Ig-like domain of the LIFR are involved in LIF binding, presumably via the LIF site III epitope. The main function of the COOH-terminal CBM of the LIFR is to position the NH(2)-terminal CBM and the Ig-like domain, so that these can bind to LIF. In analogy to a recently published model of the IL-6R complex, a model of the active LIFR complex is suggested which positions the COOH-terminal CBM at LIF site I and the NH(2)-terminal CBM and the Ig-like domain at site III. An additional contact is postulated between the Ig-like domain of gp130 and the NH(2)-terminal CBM of the LIFR.


Assuntos
Inibidores do Crescimento/metabolismo , Imunoglobulinas/química , Interleucina-6 , Linfocinas/metabolismo , Receptores de Citocinas/química , Receptores de Citocinas/metabolismo , Substituição de Aminoácidos/genética , Animais , Sítios de Ligação , Células COS , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Inibidores do Crescimento/farmacologia , Humanos , Fator Inibidor de Leucemia , Subunidade alfa de Receptor de Fator Inibidor de Leucemia , Ligantes , Linfocinas/farmacologia , Oncostatina M , Peptídeos/metabolismo , Fosforilação , Fosfotirosina/metabolismo , Estrutura Terciária de Proteína , Receptores de Citocinas/genética , Receptores de Interleucina-6/química , Receptores de Interleucina-6/genética , Receptores de Interleucina-6/metabolismo , Receptores de OSM-LIF , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Deleção de Sequência/genética , Transfecção
8.
Oncotarget ; 6(32): 33755-68, 2015 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-26418950

RESUMO

The outcome of cancer therapy strongly depends on the complex network of cell signaling pathways, including transcription factor activation following drug exposure. Here we assessed whether and how the MAP kinase (MAPK) cascade and its downstream target, the transcription factor AP-1, influence the sensitivity of malignant glioma cells to the anticancer drugs temozolomide (TMZ) and nimustine (ACNU). Both drugs induce apoptosis in glioma cells at late times following treatment. Activation of the MAPK cascade precedes apoptosis, as shown by phosphorylation of Jun kinase (JNK) and c-Jun, a main component of AP-1. Pharmacological inhibition and siRNA mediated knockdown of JNK and c-Jun reduced the level of apoptosis in LN-229 glioma cells treated with TMZ or ACNU. Analyzing the underlying molecular mechanism, we identified the pro-apoptotic gene BIM as a critical target of AP-1, which is upregulated following TMZ and ACNU. Importantly, shRNA mediated downregulation of BIM in the malignant glioma cell lines LN-229 and U87MG led to an attenuated cleavage of caspase-9 and, consequently, reduced the level of apoptosis following TMZ and ACNU treatment. Overall, we identified JNK/c-Jun activation and BIM induction as a late pro-apoptotic response of glioma cells treated with alkylating anticancer drugs.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Apoptose , Dacarbazina/análogos & derivados , Glioblastoma/patologia , MAP Quinase Quinase 4/metabolismo , Proteínas de Membrana/metabolismo , Nimustina/farmacologia , Proteínas Proto-Oncogênicas c-jun/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Antineoplásicos Alquilantes/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Proteína 11 Semelhante a Bcl-2 , Linhagem Celular Tumoral/efeitos dos fármacos , Núcleo Celular/metabolismo , Ensaio Cometa , Dacarbazina/farmacologia , Regulação Neoplásica da Expressão Gênica , Glioblastoma/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases , Fosforilação , Estrutura Terciária de Proteína , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Temozolomida , Regulação para Cima
9.
DNA Repair (Amst) ; 28: 14-20, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25703834

RESUMO

In both pro- and eukaryotes, the mutagenic and toxic DNA adduct O(6)-methylguanine (O(6)MeG) is subject to repair by alkyltransferase proteins via methyl group transfer. In addition, in prokaryotes, there are proteins with sequence homology to alkyltransferases, collectively designated as alkyltransferase-like (ATL) proteins, which bind to O(6)-alkylguanine adducts and mediate resistance to alkylating agents. Whether such proteins might enable similar protection in higher eukaryotes is unknown. Here we expressed the ATL protein of Escherichia coli (eATL) in mammalian cells and addressed the question whether it is able to protect them against the cytotoxic effects of alkylating agents. The Chinese hamster cell line CHO-9, the nucleotide excision repair (NER) deficient derivative 43-3B and the DNA mismatch repair (MMR) impaired derivative Tk22-C1 were transfected with eATL cloned in an expression plasmid and the sensitivity to N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) was determined in reproductive survival, DNA double-strand break (DSB) and apoptosis assays. The results indicate that eATL expression is tolerated in mammalian cells and conferes protection against killing by MNNG in both wild-type and 43-3B cells, but not in the MMR-impaired cell line. The protection effect was dependent on the expression level of eATL and was completely ablated in cells co-expressing the human O(6)-methylguanine-DNA methyltransferase (MGMT). eATL did not protect against cytotoxicity induced by the chloroethylating agent lomustine, suggesting that O(6)-chloroethylguanine adducts are not target of eATL. To investigate the mechanism of protection, we determined O(6)MeG levels in DNA after MNNG treatment and found that eATL did not cause removal of the adduct. However, eATL expression resulted in a significantly lower level of DSBs in MNNG-treated cells, and this was concomitant with attenuation of G2 blockage and a lower level of apoptosis. The results suggest that eATL confers protection against methylating agents by masking O(6)MeG/thymine mispaired adducts, preventing them from becoming a substrate for mismatch repair-mediated DSB formation and cell death.


Assuntos
Alquil e Aril Transferases/metabolismo , Alquilantes/toxicidade , Adutos de DNA/metabolismo , Proteínas de Escherichia coli/metabolismo , Guanina/análogos & derivados , Alquil e Aril Transferases/genética , Animais , Apoptose , Células CHO , Cricetinae , Cricetulus , DNA/efeitos dos fármacos , DNA/metabolismo , Quebras de DNA de Cadeia Dupla , Metilases de Modificação do DNA/metabolismo , Reparo do DNA , Enzimas Reparadoras do DNA/metabolismo , Proteínas de Escherichia coli/genética , Guanina/metabolismo , Humanos , Metilnitronitrosoguanidina/toxicidade , Transgenes , Proteínas Supressoras de Tumor/metabolismo
10.
Cancer Res ; 74(19): 5585-96, 2014 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-25125662

RESUMO

DNA repair processes are a key determinant of the sensitivity of cancer cells to DNA-damaging chemotherapeutics, which may induce certain repair genes as a mechanism to promote resistance. Here, we report the results of a screen for repair genes induced in cancer cells treated with DNA crosslinking agents, which identified the translesion polymerase η (PolH) as a p53-regulated target acting as one defense against interstrand crosslink (ICL)-inducing agents. PolH was induced by fotemustine, mafosfamide, and lomustine in breast cancer, glioma, and melanoma cells in vitro and in vivo, with similar inductions observed in normal cells such as lymphocytes and diploid fibroblasts. PolH contributions to the protection against ICL-inducing agents were evaluated by its siRNA-mediated attenuation in cells, which elevated sensitivity to these drugs in all tumor cell models. Conversely, PolH overexpression protected cancer cells against these drugs. PolH attenuation reduced repair of ICL lesions as measured by host cell reactivation assays and enhanced persistence of γH2AX foci. Moreover, we observed a strong accumulation of PolH in the nucleus of drug-treated cells along with direct binding to damaged DNA. Taken together, our findings implicated PolH in ICL repair as a mechanism of cancer drug resistance and normal tissue protection.


Assuntos
Antineoplásicos/farmacologia , DNA Polimerase Dirigida por DNA/metabolismo , Regulação para Cima , Animais , Linhagem Celular Tumoral , Replicação do DNA , Resistencia a Medicamentos Antineoplásicos , Humanos , Camundongos , Camundongos Endogâmicos NOD , Proteína Supressora de Tumor p53/fisiologia
11.
Carcinogenesis ; 28(1): 183-90, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16829684

RESUMO

UV light (UV-C) is a potent inducer of the c-fos gene. Cells lacking c-Fos are hypersensitive to the cytotoxic effect of UV-C indicating a protective role of c-fos induction. Here we show that cells deficient in c-Fos (fos-/-) are unable to remove cyclobutane pyrimidine dimers (CPDs) from DNA and undergo apoptosis at high frequency via the Fas pathway. We also show that in c-Fos-deficient cells the activation of c-Jun N-terminal kinase (JNK) by UV-C differs from the wild-type (wt, fos+/+). In wt cells JNK activation is transient, returning to control level 5-8 h after treatment, whereas in c-Fos-deficient cells JNK activation was long-lasting and did not return to control level. Inhibition of early JNK activation by the JNK inhibitor SP600125 attenuated CPD repair and increased UV-C induced apoptosis whereas inhibition of late JNK activation attenuated the apoptotic response of c-Fos-deficient cells. Late and sustained activation of JNK resulted in upregulation of fas (CD95, apo-1) ligand and induction of caspase 8-dependent apoptosis. The data indicate that the immediate-early induction of the JNK/c-fos pathway stimulates the repair of UV-C induced DNA lesions that protects against apoptosis. If this does not occur, cells do not recover from transcription blockage leading, as shown for c-Fos-deficient cells, to a reduced expression of MKP1, sustained JNK activation and fasL overexpression that finally results in activation of the death receptor pathway. The data support the hypothesis that non-repaired DNA damage is the cause for the late and sustained activation of the MAP kinase pathway in response to genotoxins.


Assuntos
Apoptose/efeitos da radiação , Dano ao DNA , Reparo do DNA , Proteínas Proto-Oncogênicas c-jun/fisiologia , Raios Ultravioleta/efeitos adversos , Animais , Southwestern Blotting , Western Blotting , Caspases , Ativação Enzimática/efeitos da radiação , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-jun/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
Biochemistry ; 42(18): 5244-52, 2003 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-12731865

RESUMO

In contrast to other hematopoietic cytokine receptors, the leukemia inhibitory factor receptor (LIFR) possesses two cytokine binding modules (CBMs). Previous studies suggested that the NH(2)-terminal CBM and the Ig-like domain of the LIFR are most important for LIF binding and activity. Using the recently engineered designer cytokine IC7, which induces an active heterodimer of the LIFR and gp130 after binding to the IL-6R, and several receptor chimeras of the LIFR and the interleukin-6 receptor (IL-6R) carrying the CBM of the IL-6R in place of the COOH-terminal LIFR CBM, we could assign individual receptor subdomains to individual binding sites of the ligand. The NH(2)-terminal CBM and the Ig-like domain of the LIFR bind to ligand site III, whereas the COOH-terminal CBM contacts site I. Furthermore, we show that LIFR mutants carrying the IL-6R CBM instead of the COOH-terminal CBM can replace the IL-6R by acting as an alpha-receptor for IL-6. However, in situations where a signaling competent receptor is bound at IL-6 site I, ligand binding to site III is an absolute requirement for participation of the receptor in a signaling heterodimer with gp130; i.e., a functional receptor complex of IL-6 type cytokines cannot be assembled solely via site I and II as in the growth hormone receptor complex.


Assuntos
Imunoglobulinas/química , Receptores de Citocinas/química , Receptores de Citocinas/metabolismo , Receptores de Interleucina-6/metabolismo , Animais , Sítios de Ligação , Células COS , Chlorocebus aethiops , Interleucina-6/metabolismo , Ligantes , Linfocinas/química , Linfocinas/genética , Linfocinas/metabolismo , Mutação/genética , Fosforilação , Fosfotirosina/metabolismo , Receptores de Citocinas/genética , Receptores de Interleucina-6/química , Receptores de Interleucina-6/genética , Receptores de OSM-LIF , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Deleção de Sequência/genética , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA