Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Pharmacol Ther ; 230: 107968, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34391789

RESUMO

Our understanding of the progression and mechanisms underlying the onset of Parkinson's disease (PD) has grown enormously in the past few decades. There is growing evidence suggesting that poly (ADP-ribose) polymerase 1 (PARP-1) hyperactivation is involved in various neurodegenerative disorders, including PD, and that poly (ADP-ribose) (PAR)-dependent cell death is responsible for neuronal loss. In this review, we discuss the contribution of PARP-1 and PAR in the pathological process of PD. We describe the potential pathways regulated by the enzyme, review clinically relevant PARP-1 inhibitors as potential disease-modifying therapeutics for PD, and outline important factors that need to be considered for repurposing PARP-1 inhibitors for use in PD.


Assuntos
Doenças Neurodegenerativas , Doença de Parkinson , Morte Celular , Humanos , Doenças Neurodegenerativas/metabolismo , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/metabolismo , Poli Adenosina Difosfato Ribose/metabolismo , Poli Adenosina Difosfato Ribose/farmacologia , Poli Adenosina Difosfato Ribose/uso terapêutico , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases/uso terapêutico
2.
Biochemistry ; 57(51): 6923-6926, 2018 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-30540446

RESUMO

TAR DNA-binding protein of 43 kDa (TDP-43) forms granulo-filamentous aggregates in affected brain regions of >95% of patients with ALS and ∼50% of patients with frontotemporal degeneration (FTD). Furthermore, in disease, TDP-43 becomes N-terminally truncated resulting in protein deposits that are mainly composed of the C-terminal prion-like domain (PrLD). The PrLD is inherently aggregation-prone and is hypothesized to drive protein aggregation of TDP-43 in disease. Here, we establish that the N-terminal region of the protein is critical for rapid TDP-43 granulo-filamentous aggregation. We show that the biopolymer poly(ADP-ribose), or PAR, inhibits granulo-filamentous aggregation of TDP-43 by engaging PAR-binding motifs (PBMs) embedded in the TDP-43 nuclear-localization sequence. We demonstrate that progressive N-terminal truncation of TDP-43 can decelerate aggregation kinetics and promote formation of thread-like filaments. Thus, the N-terminal region and the PBMs of TDP-43 promote rapid granulo-filamentous aggregation and antagonize formation of thread-like fibrils. These findings illustrate the complexity of TDP-43 aggregation trajectories.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Poli Adenosina Difosfato Ribose/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/metabolismo , Encéfalo/metabolismo , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Degeneração Lobar Frontotemporal/genética , Degeneração Lobar Frontotemporal/metabolismo , Humanos , Técnicas In Vitro , Cinética , Sinais de Localização Nuclear/genética , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Poli Adenosina Difosfato Ribose/farmacologia , Agregados Proteicos/efeitos dos fármacos , Agregação Patológica de Proteínas/genética , Agregação Patológica de Proteínas/metabolismo , Agregação Patológica de Proteínas/prevenção & controle , Domínios Proteicos
3.
Org Biomol Chem ; 16(46): 8904-8907, 2018 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-30203829

RESUMO

Poly(ADP-ribose) polymerase-1 (PARP-1) is an important target in cancer therapy. We present the synthesis of novel disaccharide nucleoside analogues that resemble the central motif of poly(ADP-ribose) and test their inhibitory effects on human PARP-1. Some compounds show inhibition of enzymatic activity in vitro and thus might be interesting for further investigations.


Assuntos
Poli(ADP-Ribose) Polimerase-1/antagonistas & inibidores , Poli Adenosina Difosfato Ribose/análogos & derivados , Poli Adenosina Difosfato Ribose/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases/química , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Técnicas de Química Sintética , Dissacarídeos/síntese química , Dissacarídeos/química , Dissacarídeos/farmacologia , Descoberta de Drogas , Humanos , Poli(ADP-Ribose) Polimerase-1/metabolismo , Poli Adenosina Difosfato Ribose/síntese química , Poli Adenosina Difosfato Ribose/química , Inibidores de Poli(ADP-Ribose) Polimerases/síntese química
4.
Bioorg Med Chem Lett ; 26(15): 3395-403, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27318540

RESUMO

Poly(ADP-ribose) (PAR) is an important biopolymer, which is involved in various life processes such as DNA repair and replication, modulation of chromatin structure, transcription, cell differentiation, and in pathogenesis of various diseases such as cancer, diabetes, ischemia and inflammations. PAR is the most electronegative biopolymer and this property is essential for its binding with a wide range of proteins. Understanding of PAR functions in cell on molecular level requires chemical synthesis of regular PAR oligomers. Recently developed methodologies for chemical synthesis of PAR oligomers, will facilitate the study of various cellular processes, involving PAR.


Assuntos
Desenho de Fármacos , Inibidores Enzimáticos/farmacologia , Poli Adenosina Difosfato Ribose/farmacologia , Poli(ADP-Ribose) Polimerases/metabolismo , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Humanos , Estrutura Molecular , Poli Adenosina Difosfato Ribose/síntese química , Poli Adenosina Difosfato Ribose/química , Relação Estrutura-Atividade
5.
Chem Biol ; 22(4): 446-452, 2015 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-25865309

RESUMO

Poly(ADP-ribose) polymerase 1 (PARP1) synthesizes poly(ADP-ribose) (PAR), an essential post-translational modification whose function is important in many cellular processes including DNA damage signaling, cell death, and inflammation. All known PAR biology is intracellular, but we suspected it might also play a role in cell-to-cell communication during inflammation. We found that PAR activated cytokine release in human and mouse macrophages, a hallmark of innate immune activation, and determined structure-activity relationships. PAR was rapidly internalized by murine macrophages, while the monomer, ADP-ribose, was not. Inhibitors of Toll-like receptor 2 (TLR2) and TLR4 signaling blocked macrophage responses to PAR, and PAR induced TLR2 and TLR4 signaling in reporter cell lines suggesting it was recognized by these TLRs, much like bacterial pathogens. We propose that PAR acts as an extracellular damage associated molecular pattern that drives inflammatory signaling.


Assuntos
Poli Adenosina Difosfato Ribose/farmacologia , Poli(ADP-Ribose) Polimerases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Linhagem Celular , Citocinas/metabolismo , Dimerização , Humanos , Macrófagos/citologia , Macrófagos/metabolismo , Camundongos , Microscopia Confocal , Poli Adenosina Difosfato Ribose/química , Relação Estrutura-Atividade , Receptor 2 Toll-Like/antagonistas & inibidores , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/antagonistas & inibidores , Receptor 4 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
6.
Chem Biol ; 22(4): 432-433, 2015 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-25910238

RESUMO

The inflammatory response is a critical component of the immune system that is activated by stimuli such as cytokines, foreign DNA, RNA, or other harmful substances. Krukenberg et al. (2015) identify poly(ADP-ribose) as a new signaling molecule that activates inflammation, thus providing yet another mechanism by which PARPs are involved in cellular stress responses.


Assuntos
Poli Adenosina Difosfato Ribose/farmacologia , Poli(ADP-Ribose) Polimerases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Humanos
7.
Free Radic Biol Med ; 50(1): 86-92, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-20977936

RESUMO

Changes in protein turnover are among the dominant metabolic changes during aging. Of special importance is the maintenance of nuclear protein homeostasis to ensure a coordinated cellular metabolism. Therefore, in the nucleus a special PARP-1-mediated mechanism of proteasomal activation exists to ensure a rapid degradation of oxidized nuclear proteins. It was already demonstrated earlier that the cytosolic proteasomal system declines dramatically with aging, whereas the nuclear proteasome remains less affected. We demonstrate here that the stress-mediated proteasomal activation in the nucleus declines during replicative senescence of human fibroblasts. Furthermore, we clearly show that this decline in the PARP-1-mediated proteasomal activation is due to a decline in the expression and activity of PARP-1 in senescent fibroblasts. In a final study we show that this process also happens in vivo, because the protein expression level of PARP-1 is significantly lower in the skin of aged donors compared to that of young ones. Therefore, we conclude that the rate-limiting factor in poly(ADP-ribose)-mediated proteasomal activation in oxidative stress is PARP-1 and not the nuclear proteasome itself.


Assuntos
Núcleo Celular/metabolismo , Senescência Celular/fisiologia , Estresse Oxidativo/fisiologia , Poli(ADP-Ribose) Polimerases/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Envelhecimento/fisiologia , Benzamidas/farmacologia , Biópsia , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Regulação para Baixo , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Peróxido de Hidrogênio/farmacologia , Poli(ADP-Ribose) Polimerase-1 , Poli Adenosina Difosfato Ribose/metabolismo , Poli Adenosina Difosfato Ribose/farmacologia , Inibidores de Poli(ADP-Ribose) Polimerases , Poli(ADP-Ribose) Polimerases/farmacologia
8.
J Pharmacol Exp Ther ; 335(1): 223-30, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20668052

RESUMO

The anthracycline doxorubicin (Dox) is an effective antitumor agent. However, its use is limited because of its toxicity in the heart. N-Benzyladriamycin-14-valerate (AD 198) is a modified anthracycline with antitumor efficacy similar to that of Dox, but with significantly less cardiotoxicity and potentially cardioprotective elements. In the present study, we investigated the possibility of in vivo protective effects of low-dose AD 198 against Dox-induced cardiomyopathy. To do this, rats were divided into four groups: vehicle, Dox (20 mg/kg; single injection day 1), AD 198 (0.3 mg/kg per injection; injections on days 1, 2, and 3), or a combination treatment of Dox + AD 198. Seventy-two hours after beginning treatment, hearts from the Dox group had decreased phosphorylation of AMP kinase and troponin I and reduced poly(ADP-ribose) polymerase, ß-tubulin, and serum albumin expression. Dox also increased the phosphorylation of phospholamban and expression of inducible nitric-oxide synthase in hearts. Each of these Dox-induced molecular changes was attenuated in the Dox + AD 198 group. In addition, excised hearts from rats treated with Dox had a 25% decrease in left ventricular developed pressure (LVDP) and a higher than normal increase in LVDP when perfused with a high extracellular Ca(2+) solution. The Dox-induced decrease in baseline LVDP and hyper-responsiveness to [Ca(2+)] was not observed in hearts from the Dox + AD 198 group. Thus Dox, with well established and efficient antitumor protocols, in combination with low levels of AD 198, to counter anthracycline cardiotoxicity, may be a promising next step in chemotherapy.


Assuntos
Antibióticos Antineoplásicos/antagonistas & inibidores , Antibióticos Antineoplásicos/toxicidade , Cardiomiopatias/induzido quimicamente , Cardiomiopatias/prevenção & controle , Cardiotônicos/farmacologia , Doxorrubicina/antagonistas & inibidores , Doxorrubicina/toxicidade , Quinases Proteína-Quinases Ativadas por AMP , Animais , Western Blotting , Cálcio/farmacologia , Doxorrubicina/análogos & derivados , Doxorrubicina/farmacologia , Eletroforese em Gel de Poliacrilamida , Indicadores e Reagentes , Masculino , Espectrometria de Massas , Miócitos Cardíacos/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/biossíntese , Fosforilação , Poli Adenosina Difosfato Ribose/farmacologia , Proteína Quinase C-épsilon/antagonistas & inibidores , Proteínas Quinases/metabolismo , Ratos , Ratos Wistar
9.
Proc Natl Acad Sci U S A ; 107(28): 12611-6, 2010 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-20616036

RESUMO

Asbestos carcinogenesis has been linked to the release of cytokines and mutagenic reactive oxygen species (ROS) from inflammatory cells. Asbestos is cytotoxic to human mesothelial cells (HM), which appears counterintuitive for a carcinogen. We show that asbestos-induced HM cell death is a regulated form of necrosis that links to carcinogenesis. Asbestos-exposed HM activate poly(ADP-ribose) polymerase, secrete H(2)O(2), deplete ATP, and translocate high-mobility group box 1 protein (HMGB1) from the nucleus to the cytoplasm, and into the extracellular space. The release of HMGB1 induces macrophages to secrete TNF-alpha, which protects HM from asbestos-induced cell death and triggers a chronic inflammatory response; both favor HM transformation. In both mice and hamsters injected with asbestos, HMGB1 was specifically detected in the nuclei, cytoplasm, and extracellular space of mesothelial and inflammatory cells around asbestos deposits. TNF-alpha was coexpressed in the same areas. HMGB1 levels in asbestos-exposed individuals were significantly higher than in nonexposed controls (P < 0.0001). Our findings identify the release of HMGB1 as a critical initial step in the pathogenesis of asbestos-related disease, and provide mechanistic links between asbestos-induced cell death, chronic inflammation, and carcinogenesis. Chemopreventive approaches aimed at inhibiting the chronic inflammatory response, and especially blocking HMGB1, may decrease the risk of malignant mesothelioma among asbestos-exposed cohorts.


Assuntos
Proteína HMGB1/metabolismo , Inflamação/metabolismo , Adenosina Difosfato Ribose/metabolismo , Adenosina Difosfato Ribose/farmacologia , Animais , Amianto/metabolismo , Amianto/farmacologia , Carcinógenos/metabolismo , Carcinógenos/farmacologia , Morte Celular , Núcleo Celular/metabolismo , Células/metabolismo , Cricetinae , Citocinas/metabolismo , Citocinas/farmacologia , Células Epiteliais/metabolismo , Epitélio/efeitos dos fármacos , Epitélio/metabolismo , Feminino , Proteínas HMGB/metabolismo , Proteínas HMGB/farmacologia , Proteína HMGB1/farmacologia , Humanos , Peróxido de Hidrogênio/metabolismo , Peróxido de Hidrogênio/farmacologia , Macrófagos/metabolismo , Mesocricetus , Mesotelioma/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Necrose/metabolismo , Neoplasias Pleurais/metabolismo , Poli Adenosina Difosfato Ribose/farmacologia , Poli(ADP-Ribose) Polimerases/metabolismo , Poli(ADP-Ribose) Polimerases/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Espécies Reativas de Oxigênio/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
10.
J Biol Chem ; 283(29): 19991-8, 2008 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-18495665

RESUMO

Human DNA topoisomerase I plays a dual role in transcription, by controlling DNA supercoiling and by acting as a specific kinase for the SR-protein family of splicing factors. The two activities are mutually exclusive, but the identity of the molecular switch is unknown. Here we identify poly(ADP-ribose) as a physiological regulator of the two topoisomerase I functions. We found that, in the presence of both DNA and the alternative splicing factor/splicing factor 2 (ASF/SF2, a prototypical SR-protein), poly(ADP-ribose) affected topoisomerase I substrate selection and gradually shifted enzyme activity from protein phosphorylation to DNA cleavage. A likely mechanistic explanation was offered by the discovery that poly(ADP-ribose) forms a high affinity complex with ASF/SF2 thereby leaving topoisomerase I available for directing its action onto DNA. We identified two functionally important domains, RRM1 and RS, as specific poly(ADP-ribose) binding targets. Two independent lines of evidence emphasize the potential biological relevance of our findings: (i) in HeLa nuclear extracts, ASF/SF2, but not histone, phosphorylation was inhibited by poly(ADP-ribose); (ii) an in silico study based on gene expression profiling data revealed an increased incidence of alternative splicing within a subset of inflammatory response genes that are dysregulated in cells lacking a functional poly(ADP-ribose) polymerase-1. We propose that poly(ADP-ribose) targeting of topoisomerase I and ASF/SF2 functions may participate in the regulation of gene expression.


Assuntos
DNA Topoisomerases Tipo I/metabolismo , Proteínas Nucleares/metabolismo , Poli Adenosina Difosfato Ribose/metabolismo , Processamento Alternativo/genética , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , DNA Topoisomerases Tipo I/genética , Ativação Enzimática/efeitos dos fármacos , Regulação da Expressão Gênica , Células HeLa , Histonas/metabolismo , Humanos , Proteínas Nucleares/genética , Fosforilação , Poli Adenosina Difosfato Ribose/farmacologia , Ligação Proteica , Proteínas de Ligação a RNA , Fatores de Processamento de Serina-Arginina , Inibidores da Topoisomerase I
11.
Biochem Biophys Res Commun ; 355(2): 451-6, 2007 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-17306228

RESUMO

Poly(ADP-ribose) is a biopolymer synthesized by poly(ADP-ribose) polymerases. Recent findings suggest the possibility for modulation of cellular functions including cell death and mitosis by poly(ADP-ribose). Derivatization of poly(ADP-ribose) may be useful for investigating the effects of poly(ADP-ribose) on various cellular processes. We prepared poly(etheno ADP-ribose) (poly(epsilonADP-ribose)) by converting the adenine moiety of poly(ADP-ribose) to 1-N(6)-etheno adenine residues. Poly(epsilonADP-ribose) is shown to be highly resistant to digestion by poly(ADP-ribose) glycohydrolase (Parg). On the other hand, poly(epsilonADP-ribose) could be readily digested by phosphodiesterase. Furthermore, poly(epsilonADP-ribose) inhibited Parg activity to hydrolyse ribose-ribose bonds of poly(ADP-ribose). This study suggests the possibility that poly(epsilonADP-ribose) might be a useful tool for studying the poly(ADP-ribose) dynamics and function of Parg. This study also implies that modification of the adenine moiety of poly(ADP-ribose) abrogates the susceptibility to digestion by Parg.


Assuntos
Inibidores Enzimáticos/farmacologia , Glicosídeo Hidrolases/antagonistas & inibidores , Poli Adenosina Difosfato Ribose/análogos & derivados , Cromatografia Líquida de Alta Pressão , Células HeLa , Humanos , Poli Adenosina Difosfato Ribose/farmacologia , Poli(ADP-Ribose) Polimerases/genética , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Recombinantes/genética , Espectrometria de Massas por Ionização por Electrospray , Espectrofotometria Ultravioleta
12.
Proc Natl Acad Sci U S A ; 103(48): 18314-9, 2006 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-17116881

RESUMO

Apoptosis-inducing factor (AIF), a mitochondrial oxidoreductase, is released into the cytoplasm to induce cell death in response to poly(ADP-ribose) (PAR) polymerase-1 (PARP-1) activation. How PARP-1 activation leads to AIF release is not known. Here we identify PAR polymer as a cell death signal that induces release of AIF. PAR polymer induces mitochondrial AIF release and translocation to the nucleus. PAR glycohydrolase, which degrades PAR polymer, prevents PARP-1-dependent AIF release. Cells with reduced levels of AIF are resistant to PARP-1-dependent cell death and PAR polymer cytotoxicity. These results reveal PAR polymer as an AIF-releasing factor that plays important roles in PARP-1-dependent cell death.


Assuntos
Fator de Indução de Apoptose/metabolismo , Poli Adenosina Difosfato Ribose/farmacologia , Polímeros/farmacologia , Animais , Morte Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Células Cultivadas , Citosol/efeitos dos fármacos , Humanos , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Transporte Proteico
13.
Biochem Pharmacol ; 70(10): 1458-68, 2005 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-16191427

RESUMO

Thymidylate synthase (TS) is an important target of several chemotherapeutic agents. During TS inhibition, dTTP levels decrease with a subsequent increase in dUTP. Uracil incorporated into the genome is removed by base excision repair (BER). BER has been hypothesized to play a role in the response to thymidylate deprivation, despite a lack of direct evidence. We previously found that beta-pol null murine fibroblasts were approximately six-fold more resistant than wild-type cells to raltitrexed, a folate-based inhibitor specific for TS. In this study, a number of endpoints were determined to understand the influence of BER and beta-pol during raltitrexed treatment. Raltitrexed induced apoptosis in wild-type cells to a greater extent than in beta-pol null cells. A PARP inhibitor decreased the sensitivity to raltitrexed, although the extent was not different between wild-type and beta-pol null cells. No evidence was seen for extensive strand break formation that preceded apoptosis, although raltitrexed induced more sister chromatid exchanges in wild-type cells. Increased levels of uracil in DNA were detected following treatment in wild-type and beta-pol null cells. However, uracil levels were only approximately two-fold higher in DNA from treated cells compared to untreated. Uracil DNA glycosylase activity was slightly higher in beta-pol null cells, although not sufficiently different to explain the difference in sensitivity to raltitrexed. Taken together, the data suggest that the sensitivity of the wild-type cells to raltitrexed is not associated with activation of PARP-1 dependent BER, extensive uracil incorporation into DNA and persistent strand breaks, but rather with changes suggestive of DNA recombination.


Assuntos
Apoptose/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Reparo do DNA/fisiologia , Troca de Cromátide Irmã/efeitos dos fármacos , Timidilato Sintase/antagonistas & inibidores , Uracila/metabolismo , Animais , Apoptose/fisiologia , Linhagem Celular , Células Cultivadas , DNA Polimerase beta/antagonistas & inibidores , DNA Polimerase beta/farmacologia , Reparo do DNA/efeitos dos fármacos , Relação Dose-Resposta a Droga , Embrião de Mamíferos/patologia , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Humanos , Camundongos , Poli Adenosina Difosfato Ribose/antagonistas & inibidores , Poli Adenosina Difosfato Ribose/farmacologia , Quinazolinas/antagonistas & inibidores , Quinazolinas/toxicidade , Troca de Cromátide Irmã/fisiologia , Tiofenos/antagonistas & inibidores , Tiofenos/toxicidade , Timidilato Sintase/efeitos dos fármacos , Fatores de Tempo , Uracila/química , Uracila/farmacologia , Uracila-DNA Glicosidase/química , Uracila-DNA Glicosidase/efeitos dos fármacos , Uracila-DNA Glicosidase/metabolismo
14.
Mutat Res ; 583(1): 36-48, 2005 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-15866464

RESUMO

The boron neutron capture (BNC) reaction results from the interaction of 10B with low-energy thermal neutrons and gives rise to highly damaging lithium and alpha-particles. In this work the genotoxicity caused by the BNC reaction in V79 Chinese hamster cells was evaluated in the presence of poly(ADP-ribosyl)ation inhibitors. Poly(ADP-ribose) polymerase-1 (PARP-1), the most important member of the PARP enzyme family, is considered to be a constitutive factor of the DNA damage surveillance network present in eukaryotic cells, acting through a DNA break sensor function. Inhibition of poly(ADP-ribosyl)ation was achieved with the classical compound 3-aminobenzamide (3-AB), and with two novel and very potent inhibitors, 5-aminoisoquinolinone (5-AIQ) and PJ-34. Dose-response increases in the frequencies of aberrant cells excluding gaps (%ACEG) and chromosomal aberrations excluding gaps per cell (CAEG/cell) were observed for increasing exposures to the BNC reaction. The presence of 3-AB did not increase the %ACEG or CAEG/cell, nor did it change the pattern of the induced chromosomal aberrations. Results with 5-AIQ and PJ-34 were in agreement with the results obtained with 3-AB. We further studied the combined effect of a PARP inhibitor and a DNA-dependent protein kinase (DNA-PK) inhibitors (3-AB and wortmannin, respectively) on the genotoxicity of the BNC reaction, by use of the cytokinesis-block micronucleus assay. DNA-PK is also activated by DNA breaks and binds DNA ends, playing a role of utmost importance in the repair of double-strand breaks. Our results show that the inhibition of poly(ADP-ribosyl)ation does not particularly modify the genotoxicity of the BNC reaction, and that PARP inhibition together with a concomitant inhibition of DNA-PK revealed barely the same sensitizing effect as DNA-PK inhibition per se.


Assuntos
Terapia por Captura de Nêutron de Boro/efeitos adversos , Dano ao DNA , Poli Adenosina Difosfato Ribose/antagonistas & inibidores , Poli Adenosina Difosfato Ribose/farmacologia , Partículas alfa , Animais , Técnicas de Cultura de Células , Cricetinae , Cricetulus , Proteína Quinase Ativada por DNA , Proteínas de Ligação a DNA/farmacologia , Fibroblastos , Humanos , Testes de Mutagenicidade , Proteínas Nucleares , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/farmacologia , Proteínas Serina-Treonina Quinases/farmacologia
15.
DNA Repair (Amst) ; 3(7): 753-67, 2004 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-15177184

RESUMO

Ataxia-telangiectasia mutated (ATM) is a serine/threonine protein kinase that plays a central role in controlling the cellular response to ionizing radiation and other DNA-damaging agents. ATM is a 3056 amino acid polypeptide that is present in low abundance in the nucleus of human cells. Here, we describe the purification and characterization of ATM from the nuclear fraction of HeLa cells. Microgram quantities of highly stable, kinase-active ATM were prepared. Purified ATM was phosphorylated on serine 1981 and was active towards a variety of known ATM substrates, including p53 and the Bloom Syndrome helicase, BLM. The protein kinase activity of ATM was selectively inhibited by wortmannin, caffeine and LY294002 and was stimulated by charged biological polymers, including single-stranded M13 DNA (ssDNA), sheared double-stranded calf thymus DNA, heparin sulfate and poly ADP-ribose (PAR), raising the possibility that charged structures may contribute to regulation of ATM activity. However, chemical inhibition of the formation of poly ADP-ribose in cells had no effect on the activation of ATM-dependent pathways by ionizing radiation. Using gel filtration chromatography, we also show that purified ATM, as well as ATM in crude nuclear extracts from unirradiated and irradiated cells elutes with an estimated native molecular weight of approximately 600 kDa. Moreover, dephosphorylation of serine 1981 did not affect the apparent molecular weight of ATM in irradiated extracts. Our results suggest that phosphorylation of serine 1981 alone may not directly regulate the subunit composition of ATM.


Assuntos
Ataxia Telangiectasia/genética , Proteínas Serina-Treonina Quinases/metabolismo , Sequência de Aminoácidos , Androstadienos/farmacologia , Ataxia Telangiectasia/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia , Cafeína/farmacologia , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Núcleo Celular/química , Cromatografia em Gel , Cromonas/farmacologia , DNA de Neoplasias , DNA de Cadeia Simples , Proteínas de Ligação a DNA , Inibidores Enzimáticos/farmacologia , Células HeLa , Heparina/farmacologia , Humanos , Peso Molecular , Morfolinas/farmacologia , Peptídeos/química , Fosforilação , Poli Adenosina Difosfato Ribose/farmacologia , Proteínas Quinases/efeitos dos fármacos , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/isolamento & purificação , Proteínas Serina-Treonina Quinases/efeitos da radiação , Radiação Ionizante , Serina/química , Serina/metabolismo , Proteínas Supressoras de Tumor , Wortmanina
16.
Toxicology ; 184(1): 1-9, 2003 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-12505371

RESUMO

Mercurial compounds modulate immunologic functions by inducing cytotoxicity. Although mercury chloride (HgCl(2)) is known to induce apoptosis in various immune system cells, the mechanism of the induction of apoptosis is poorly understood. In this study, we examined the activation of caspase-3, an important cysteine aspartic protease, during HgCl(2)-induced apoptosis in a human leukemia cell line (HL-60 cells). Both DNA fragmentation, a characteristic of apoptotic cells, and proteolysis of poly(ADP-ribose) polymerase (PARP), a substrate of caspase-3, occurred at 6 h after HgCl(2) treatment in HL-60 cells. These results suggest that the activation of caspase-3 was involved in HgCl(2)-induced apoptosis. The release of cytochrome c (Cyt c) from mitochondria into the cytosol, which is an initiator of the activation of caspase cascades, was also observed in HgCl(2)-treated HL-60 cells. Moreover, the release of Cyt c from mitochondria was observed in HgCl(2)-treated mitochondria isolated from mice liver, and this was followed by mitochondrial permeability transition (PT). The PT was inhibited by cyclosporin A (CsA), a potent inhibitor of PT. CsA also suppressed the occurrence of DNA fragmentation induced by HgCl(2) treatment in HL-60 cells. Taken together, these findings indicate that HgCl(2) is a potent inducer of apoptosis via Cyt c release from the mitochondria in HL-60 cells.


Assuntos
Apoptose/efeitos dos fármacos , Leucemia Promielocítica Aguda/patologia , Cloreto de Mercúrio/toxicidade , Mitocôndrias/efeitos dos fármacos , Animais , Western Blotting , Sistema Livre de Células , Grupo dos Citocromos c/metabolismo , Fragmentação do DNA/efeitos dos fármacos , Células HL-60 , Humanos , Leucemia Promielocítica Aguda/etnologia , Camundongos , Mitocôndrias/enzimologia , Mitocôndrias/fisiologia , Permeabilidade/efeitos dos fármacos , Poli Adenosina Difosfato Ribose/farmacologia
17.
Biochemistry ; 39(34): 10413-8, 2000 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-10956031

RESUMO

It is well-known that H1-H1 interactions are very important for the induction of 30 nm chromatin fiber and that, among all posttranslational modifications, poly(ADP-ribosyl)ation is one of those capable of modifying chromatin structure, mainly through H1 histone. As this protein can undergo both covalent and noncovalent modifications by poly(ADP-ribosyl)ation, our aim was to investigate whether and how ADP-ribose polymers, by themselves, are able to affect the formation of H1-H1 oligomers, which are normally present in a condensed chromatin structure. The results obtained in our in vitro experimental system indicate that ADP-ribose polymers are involved in chromatin decondensation. This conclusion was reached as the result of two different observations: (a) H1 histone molecules can be hosted in clusters on ADP-ribose polymers, as shown by their ability to be chemically cross-linked, and (b) H1 histone has a higher affinity for ADP-ribose polymers than for DNA; ADP-ribose polymers compete, in fact, with DNA for H1 histone binding.


Assuntos
Histonas/química , Histonas/metabolismo , Poli Adenosina Difosfato Ribose/farmacologia , Animais , Ligação Competitiva , Linhagem Celular , Reagentes de Ligações Cruzadas/farmacologia , DNA/metabolismo , Técnicas In Vitro , Camundongos , Poli Adenosina Difosfato Ribose/metabolismo , Estrutura Quaternária de Proteína
18.
Biochemistry ; 37(26): 9520-7, 1998 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-9649335

RESUMO

In mammalian cells, the formation of DNA strand breaks is accompanied by synthesis of poly(ADP-ribose). This nucleic acid-like homopolymer may modulate protein functions by covalent and/or noncovalent interactions. Here we show that poly(ADP-ribose) binds strongly to the proteins of the myristoylated alanine-rich C kinase substrate (MARCKS) family, MARCKS and MARCKS-related protein (also MacMARCKS or F52). MARCKS proteins are myristoylated proteins associated with membranes and the actin cytoskeleton. As targets for both protein kinase C (PKC) and calmodulin (CaM), MARCKS proteins are thought to mediate cross-talk between these two signal transduction pathways. Dot blot assays show that poly(ADP-ribose) binds to MARCKS proteins at the highly basic effector domain. Complex formation between MARCKS-related protein and CaM as well as phosphorylation of MARCKS-related protein by the catalytic subunit of PKC are strongly inhibited by equimolar amounts of poly(ADP-ribose), suggesting a high affinity of poly(ADP-ribose) for MARCKS-related protein. Binding of MARCKS-related protein to membranes is also inhibited by poly(ADP-ribose). Finally, poly(ADP-ribose) efficiently reverses the actin-filament bundling activity of a peptide corresponding to the effector domain and inhibits the formation of actin filaments in vitro. Our results suggest that MARCKS proteins and actin could be targets of the poly(ADP-ribose) DNA damage signal pathway.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana , Poli Adenosina Difosfato Ribose/farmacologia , Proteína Quinase C/metabolismo , Proteínas/metabolismo , Actinas/antagonistas & inibidores , Actinas/metabolismo , Sequência de Aminoácidos , Animais , Calmodulina/metabolismo , Humanos , Lipídeos de Membrana/antagonistas & inibidores , Lipídeos de Membrana/metabolismo , Camundongos , Dados de Sequência Molecular , Substrato Quinase C Rico em Alanina Miristoilada , Fosforilação/efeitos dos fármacos , Poli Adenosina Difosfato Ribose/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Polímeros/metabolismo , Ligação Proteica/efeitos dos fármacos , Proteínas/antagonistas & inibidores
19.
J Cell Sci ; 111 ( Pt 9): 1217-25, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9547298

RESUMO

To further characterize how gap junction-dependent Ca2+ waves propagate between sheep lens cells, we examined the possible roles of inositol 1,4,5-trisphosphate (IP3), Ca2+ and cyclic ADP-ribose (cADPR) in mediating intercellular Ca2+ waves. Second messengers were microinjected into a single cell in a monolayer of sheep lens cells while monitoring cytosolic Ca2+ with fura-2 and fluorescence microscopy. All three compounds initiated intercellular Ca2+ waves, but more cells responded following the injection of either IP3 or cADPR than responded following the injection of Ca2+. When either IP3 or cADPR was co-injected with the Ca2+ chelator EGTA, cytosolic Ca2+ in the injected cell decreased but cytosolic Ca2+ in the adjacent cells increased, indicating that the intercellular messenger was IP3 or cADPR, rather than Ca2+. The phospholipase C inhibitor U73122 eliminated mechanically initiated intercellular Ca2+ waves, indicating that mechanical initiation probably requires IP3 production. In U73122-treated cells, injected IP3 initiated an intercellular Ca2+ wave in which the number of cells responding increased as the amount of IP3 injected increased, indicating that the distance traveled by the Ca2+ wave was dependent on cell-to-cell diffusion of IP3. In contrast, the ability of cADPR both to increase cytosolic Ca2+ in the injected cell and to initiate intercellular Ca2+ waves was greatly attenuated by U73122. In conclusion, Ca2+, IP3 and cADPR can all mediate intercellular Ca2+ waves by passing through gap junction channels, but both IP3 and cADPR are more effective intercellular messengers than Ca2+.


Assuntos
Cálcio/fisiologia , Junções Comunicantes/fisiologia , Inositol 1,4,5-Trifosfato/fisiologia , Cristalino/citologia , Poli Adenosina Difosfato Ribose/fisiologia , Transdução de Sinais/fisiologia , Animais , Cálcio/farmacologia , Comunicação Celular , Células Cultivadas , Quelantes/farmacologia , Ácido Egtázico/farmacologia , Inibidores Enzimáticos/farmacologia , Estrenos/farmacologia , Inositol 1,4,5-Trifosfato/farmacologia , Transporte de Íons , Cristalino/metabolismo , Microinjeções , Microscopia de Fluorescência , Fosfatidilinositol Diacilglicerol-Liase , Poli Adenosina Difosfato Ribose/farmacologia , Pirrolidinonas/farmacologia , Ovinos , Fosfolipases Tipo C/antagonistas & inibidores , Fosfolipases Tipo C/fisiologia
20.
J Biol Chem ; 264(14): 7850-5, 1989 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-2542248

RESUMO

Poly(ADP-ribose) prepared by incubating NAD+ with rat liver nuclei inhibited the hydroxylation reaction catalyzed by purified prolyl hydroxylase (proline,2-oxoglutarate dioxygenase, EC 1.14.11.2) in vitro. Near complete inhibition of the enzyme was seen in the presence of 6 nM (ADP-Rib)18 with a Ki(app) of 1.5 nM. The monomer unit of poly(ADP-ribose), adenosine diphosphoribose (ADP-Rib), was found to be a weak inhibitor. On the other hand, poly(ADP-ribose)-derived phosphoribosyl-AMP (PRib-AMP) and its dephosphorylated product, ribosyl-ribosyl-adenine (Rib-RibA), inhibited the enzyme in nanomolar concentrations (Ki(app) 16.25 nM). The order of inhibition was (ADP-Rib)18 greater than PRib-AMP, Rib-RibA much greater than ADP-Rib. These results suggested that the 1"----2' ribosyl-ribosyl moiety in these compounds was involved in the inhibition of the enzyme. The possibility that intracellular prolyl hydroxylase is regulated by the involvement of ADP-ribosylation reactions was examined in confluent cultures of skin fibroblast treated with 20 mM lactate. The activity of prolyl hydroxylase was stimulated by 145% over that of untreated cultures. In the lactate-treated cells, the level of NAD+ was lowered and the total ADP-ribosylation of cellular proteins reduced by 40%. These observations imply that the lactate-induced activation of cellular prolyl hydroxylase is mediated by a reduction in ADP-ribosylation and that the synthesis and degradation of ADP-ribose moiety(ies) may possibly regulate prolyl hydroxylase activity in vivo.


Assuntos
Monofosfato de Adenosina/análogos & derivados , Oxigenases de Função Mista , Açúcares de Nucleosídeo Difosfato/farmacologia , Poli Adenosina Difosfato Ribose/farmacologia , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Adenosina/análogos & derivados , Adenosina/farmacologia , Adenosina Difosfato Ribose/farmacologia , Monofosfato de Adenosina/farmacologia , Animais , Núcleo Celular/metabolismo , Embrião de Galinha , Ativação Enzimática/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Hidroxilação , Lactatos/farmacologia , Ácido Láctico , Fígado/metabolismo , NAD/metabolismo , Fosfodiesterase I , Diester Fosfórico Hidrolases/metabolismo , Pró-Colágeno/metabolismo , Coelhos , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA