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











Base de dados
Intervalo de ano de publicação
1.
J Biol Chem ; 295(23): 7849-7864, 2020 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-32317279

RESUMO

Activation of the T cell receptor (TCR) results in binding of the adapter protein Nck (noncatalytic region of tyrosine kinase) to the CD3ϵ subunit of the TCR. The interaction was suggested to be important for the amplification of TCR signals and is governed by a proline-rich sequence (PRS) in CD3ϵ that binds to the first Src homology 3 (SH3) domain of Nck (Nck-SH3.1). Inhibition of this protein/protein interaction ameliorated inflammatory symptoms in mouse models of multiple sclerosis, psoriasis, and asthma. A small molecule, AX-024, was reported to inhibit the Nck/CD3ϵ interaction by physically binding to the Nck1-SH3.1 domain, suggesting a route to develop an inhibitor of the Nck1/CD3ϵ interaction for modulating TCR activity in autoimmune and inflammatory diseases. We show here that AX-024 reduces T cell proliferation upon weak TCR stimulation but does not significantly affect phosphorylation of Zap70 (ζ chain of T cell receptor-associated protein kinase 70). We also find that AX-024 is likely not involved in modulating the Nck/TCR interaction but probably has other targets in T cells. An array of biophysical techniques did not detect a direct interaction between AX-024 and Nck-SH3.1 in vitro Crystal structures of the Nck-SH3.1 domain revealed its binding mode to the PRS in CD3ϵ. The SH3 domain tends to generate homodimers through a domain swap. Domain swaps observed previously in other SH3 domains indicate a general propensity of this protein fold to exchange structural elements. The swapped form of Nck-SH3.1 is unable to bind CD3ϵ, possibly representing an inactive form of Nck in cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Complexo CD3/metabolismo , Proteínas Oncogênicas/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Linfócitos T/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Humanos , Células Jurkat , Modelos Moleculares , Domínios de Homologia de src
2.
J Biol Chem ; 293(49): 18864-18878, 2018 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-30291141

RESUMO

The inflammasome is a critical molecular complex that activates interleukin-1 driven inflammation in response to pathogen- and danger-associated signals. Germline mutations in the inflammasome sensor NLRP1 cause Mendelian systemic autoimmunity and skin cancer susceptibility, but its endogenous regulation remains less understood. Here we use a proteomics screen to uncover dipeptidyl dipeptidase DPP9 as a novel interacting partner with human NLRP1 and a related inflammasome regulator, CARD8. DPP9 functions as an endogenous inhibitor of NLRP1 inflammasome in diverse primary cell types from human and mice. DPP8/9 inhibition via small molecule drugs and CRISPR/Cas9-mediated genetic deletion specifically activate the human NLRP1 inflammasome, leading to ASC speck formation, pyroptotic cell death, and secretion of cleaved interleukin-1ß. Mechanistically, DPP9 interacts with a unique autoproteolytic domain (Function to Find Domain (FIIND)) found in NLRP1 and CARD8. This scaffolding function of DPP9 and its catalytic activity act synergistically to maintain NLRP1 in its inactive state and repress downstream inflammasome activation. We further identified a single patient-derived germline missense mutation in the NLRP1 FIIND domain that abrogates DPP9 binding, leading to inflammasome hyperactivation seen in the Mendelian autoinflammatory disease Autoinflammation with Arthritis and Dyskeratosis. These results unite recent findings on the regulation of murine Nlrp1b by Dpp8/9 and uncover a new regulatory mechanism for the NLRP1 inflammasome in primary human cells. Our results further suggest that DPP9 could be a multifunctional inflammasome regulator involved in human autoinflammatory diseases.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Dipeptidil Peptidases e Tripeptidil Peptidases/metabolismo , Inflamassomos/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Reguladoras de Apoptose/antagonistas & inibidores , Proteínas Reguladoras de Apoptose/genética , Ácidos Borônicos/farmacologia , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Dipeptídeos/farmacologia , Dipeptidil Peptidases e Tripeptidil Peptidases/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Mutação em Linhagem Germinativa , Células HEK293 , Humanos , Inflamação/genética , Mutação de Sentido Incorreto , Proteínas NLR , Proteínas de Neoplasias/metabolismo , Ligação Proteica , Domínios Proteicos
3.
Ann Rheum Dis ; 72(6): 1044-52, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23117241

RESUMO

OBJECTIVES: Tumour necrosis factor (TNF) receptor-associated periodic syndrome (TRAPS) is caused by TNFRSF1A mutations, known to induce intracellular retention of the TNFα receptor 1 (TNFR1) protein, defective TNFα-induced apoptosis, and production of reactive oxygen species. As downregulation of autophagy, the main cellular pathway involved in insoluble aggregate elimination, has been observed to increase the inflammatory response, we investigated whether it plays a role in TRAPS pathogenesis. METHODS: The possible link between TNFRSF1A mutations and inflammation in TRAPS was studied in HEK-293T cells, transfected with expression constructs for wild-type and mutant TNFR1 proteins, and in monocytes derived from patients with TRAPS, by investigating autophagy function, NF-κB activation and interleukin (IL)-1ß secretion. RESULTS: We found that autophagy is responsible for clearance of wild-type TNFR1, but when TNFR1 is mutated, the autophagy process is defective, probably accounting for mutant TNFR1 accumulation as well as TRAPS-associated induction of NF-κB activity and excessive IL-1ß secretion, leading to chronic inflammation. Autophagy inhibition due to TNFR1 mutant proteins can be reversed, as demonstrated by the effects of the antibiotic geldanamycin, which was found to rescue the membrane localisation of mutant TNFR1 proteins, reduce their accumulation and counteract the increased inflammation by decreasing IL-1ß secretion. CONCLUSIONS: Autophagy appears to be an important mechanism in the pathogenesis of TRAPS, an observation that provides a rationale for the most effective therapy in this autoinflammatory disorder. Our findings also suggest that autophagy could be proposed as a novel therapeutic target for TRAPS and possibly other similar diseases.


Assuntos
Autofagia/genética , Doenças Hereditárias Autoinflamatórias/genética , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Adolescente , Adulto , Autofagia/fisiologia , Estudos de Casos e Controles , Criança , Pré-Escolar , Células HEK293 , Humanos , Interleucina-1beta/metabolismo , Pessoa de Meia-Idade , NF-kappa B/fisiologia , Receptores Tipo I de Fatores de Necrose Tumoral/fisiologia , Síndrome
4.
Pigment Cell Melanoma Res ; 25(1): 5-8, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22117610

RESUMO

Some familial forms of the dermatological condition vitiligo have recently been linked to polymorphisms in the innate immunity gene, NLRP1. Here, we review what is currently known about the mechanisms that regulate activation of the NLRP1 protein and the downstream effects of NLRP1 on pathways impacting inflammation and apoptosis. How polymorphic variants of the NLRP1 gene contribute to the pathogenesis of vitiligo remains mysterious, requiring further investigation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Proteínas Reguladoras de Apoptose/fisiologia , Imunidade Inata/genética , Vitiligo/genética , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Proteínas Reguladoras de Apoptose/deficiência , Proteínas Reguladoras de Apoptose/genética , Caspases/metabolismo , Ativação Enzimática , Humanos , Camundongos , Família Multigênica , Proteínas NLR , Polimorfismo Genético , Vitiligo/imunologia , Vitiligo/fisiopatologia
5.
PLoS One ; 6(11): e27396, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22087307

RESUMO

The "Function to Find Domain" (FIIND)-containing proteins CARD8 (Cardinal; Tucan) and NLRP1 (NALP1; NAC) are well known components of inflammasomes, multiprotein complexes responsible for activation of caspase-1, a regulator of inflammation and innate immunity. Although identified many years ago, the role of the FIIND is unknown. Here, we report that CARD8 and NLRP1 undergo autoproteolytic cleavage at a conserved SF/S motif within the FIIND. Using bioinformatics and computational modeling approaches, we detected striking structural similarity between the FIIND and the ZU5-UPA domain present in the autoproteolytic protein PIDD. This allowed us to generate a three-dimensional model and to gain insights in the molecular mechanism of the cleavage. Site-directed mutagenesis experiments revealed that the second serine of the SF/S motif is required for CARD8 and NLRP1 autoproteolysis. Furthermore, we discovered an important function for conserved glutamic acid and histidine residues, located in proximity of the cleavage site in regulating the autoprocessing efficiency. Altogether, these results identify a function for the FIIND and show that CARD8 and NLRP1 are ZU5-UPA domain-containing autoproteolytic proteins, thus suggesting a novel mechanism for regulating innate immune responses.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Proteínas de Neoplasias/metabolismo , Peptídeo Hidrolases/metabolismo , Sítios de Ligação , Linhagem Celular , Humanos , Imunidade Inata , Inflamassomos , Complexos Multiproteicos/imunologia , Proteínas NLR , Estrutura Terciária de Proteína
6.
Arthritis Rheum ; 56(9): 3138-48, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17763411

RESUMO

OBJECTIVE: To examine the synthesis, processing, and secretion of interleukin-1beta (IL-1beta), as well as the clinical and biologic effects of IL-1 blockade, in patients with chronic infantile neurologic, cutaneous, articular (CINCA) syndrome and Muckle-Wells syndrome (MWS), in an effort to understand the molecular mechanisms linking mutations of the CIAS1 gene and IL-1beta hypersecretion, and the underlying response to IL-1 receptor antagonist (IL-1Ra). METHODS: Six patients with CINCA syndrome or MWS were treated with IL-1Ra and followed up longitudinally. Monocytes obtained from the patients and from 24 healthy donors were activated with lipopolysaccharide (LPS) for 3 hours, and intracellular and secreted IL-1beta levels were determined by Western blotting and enzyme-linked immunosorbent assay before and after exposure to exogenous ATP. RESULTS: LPS-induced IL-1beta secretion was markedly increased in monocytes from patients with CIAS1 mutations. However, unlike in healthy subjects, secretion of IL-1beta was not induced by exogenous ATP. Treatment with IL-1Ra resulted in a dramatic clinical improvement, which was paralleled by an early and strong down-regulation of LPS-induced IL-1beta secretion by the patients' cells in vitro. CONCLUSION: Our results showed that the requirements of ATP stimulation for IL-1beta release observed in healthy individuals are bypassed in patients bearing CIAS1 mutations. This indicates that cryopyrin is the direct target of ATP and that the mutations release the protein from the requirement of ATP for activation. In addition, the dramatic amelioration induced by IL-1Ra treatment is at least partly due to the strong decrease in IL-1beta secretion that follows the first injections of the antagonist. These findings may have implications for other chronic inflammatory conditions characterized by increased IL-1beta.


Assuntos
Dor Abdominal/genética , Dor Abdominal/imunologia , Artrite/genética , Artrite/imunologia , Proteínas de Transporte/genética , Febre/genética , Febre/imunologia , Proteína Antagonista do Receptor de Interleucina 1/farmacologia , Interleucina-1/antagonistas & inibidores , Interleucina-1beta/metabolismo , Mutação , Doenças do Sistema Nervoso/genética , Doenças do Sistema Nervoso/imunologia , Urticária/genética , Urticária/imunologia , Trifosfato de Adenosina/administração & dosagem , Adolescente , Adulto , Criança , Pré-Escolar , Doença Crônica , Feminino , Humanos , Lipopolissacarídeos/administração & dosagem , Estudos Longitudinais , Masculino , Proteína 3 que Contém Domínio de Pirina da Família NLR , Síndrome
7.
Scand J Gastroenterol ; 41(4): 430-6, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16635911

RESUMO

OBJECTIVE: Hyper-IgD syndrome (HIDS) is a rare autosomal recessive disease characterized by recurrent fever, lymphadenopathy, diarrhoea, abdominal pain, headache, arthralgia and skin rash. Abdominal symptomatology may mimic inflammatory bowel disease. We report on four patients with HIDS who had been previously investigated for Crohn's disease (CD). The levels of IgD were measured in a series of patients with CD to evaluate the specificity of this assay in the differential diagnosis between the two conditions. MATERIAL AND METHODS: Diagnosis of HIDS was based on clinical criteria as well as immunological or genetic data. IgD levels were measured in the four subjects affected by HIDS, in 59 patients with CD and in a group of 160 healthy controls. RESULT: All patients underwent a variety of gastroenterological investigations because inflammatory bowel disease was suspected. Ultrasonography was pathologic in all the patients, showing enlargement of mesenteric lymph nodes. Abdominal leucocyte scintigraphy displayed diffuse signals of mild to moderate degree. IgD and IgA levels were elevated in three out of four patients. No difference in IgD values was found in CD patients as compared to the control group. CONCLUSIONS: Gastrointestinal complaints associated with recurrent fever and mesenteric adenopathy warrant genetic investigation for HIDS, in order to avoid unnecessary invasive investigations and treatment.


Assuntos
Hipergamaglobulinemia/diagnóstico , Imunoglobulina D , Criança , Pré-Escolar , Doença de Crohn/diagnóstico , Diagnóstico Diferencial , Feminino , Gastroenteropatias/complicações , Humanos , Hipergamaglobulinemia/diagnóstico por imagem , Imunoglobulina D/sangue , Inflamação/diagnóstico , Linfonodos/diagnóstico por imagem , Masculino , Ultrassonografia
8.
Arthritis Rheum ; 54(3): 998-1008, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16508982

RESUMO

OBJECTIVE: To explore tumor necrosis factor (TNF)-induced apoptosis in neutrophils from patients with TNF receptor-associated periodic syndrome (TRAPS) and to correlate the results with the different kinds of TNFRSF1A mutations. METHODS: Two hundred sixty-five patients with clinically suspected inherited autoinflammatory syndrome were screened for mutations of the TNFRSF1A gene. Neutrophils were isolated from heparinized blood by dextran sedimentation and incubated with and without cycloheximide (CHX) and TNFalpha. Cell apoptosis was assessed by human annexin V binding, and caspase 8 activation was assessed by flow cytometry. RESULTS: Twenty-one patients were found to carry a variant of the TNFRSF1A gene: 13 patients had an R92Q substitution, and 8 patients presented other missense substitutions, 1 splicing mutation, and 1 in-frame interstitial deletion. Neutrophil stimulation with TNF and CHX was associated with induction of apoptosis in 12 normal controls and in 10 subjects with the R92Q mutation. Conversely, neutrophils from 8 TRAPS patients with mutations of cysteine or threonine residues or interstitial deletion did not show any induction of apoptosis after stimulation. The incidence of the R92Q mutation among patients with recurrent autoinflammatory syndromes was similar to that observed in the normal population. CONCLUSION: Resistance to TNF-mediated apoptosis is a feature in TRAPS patients who have mutations of cysteine residues or interstitial deletion, and may play a pathogenic role. The R92Q mutation does not appear to be significantly associated with TRAPS.


Assuntos
Apoptose/fisiologia , Mutação , Neutrófilos/fisiologia , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Fator de Necrose Tumoral alfa/fisiologia , Adulto , Anexina A5/metabolismo , Caspase 8 , Caspases/farmacologia , Células Cultivadas , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Síndrome
9.
Eur J Hum Genet ; 13(3): 314-20, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15536479

RESUMO

Autosomal recessive autoinflammatory disorder caused by mutations of the mevalonate kinase gene (MVK), leading to mild, incomplete MK enzyme deficiency (MKD), has been known so far as Hyper-IgD and periodic fever syndrome (HIDS) and regarded as mostly occurring in Northern Europe. Here we report the results of the molecular characterization of the first Italian series of patients affected with autoinflammatory disorders and periodic fever. A total of 13 different mutations, scattered throughout the MVK coding region, were identified in either homozygous or compound heterozygous state in 15 patients. The mutation leading to the V377I amino-acid change, already described also in other series, resulted the most common with a frequency of 50% of all MKD alleles. Among the other mutations, eight had never been described before, including an interstitial deletion of 19 nucleotides in exon 2. In addition to these nucleotide changes, private and polymorphic MVK variants have been detected in the patients under analysis and checked also in a set of control individuals. Clinical features are reported for each of the 15 MKD patients, and life-threatening infections and systemic amyloidosis presented as unexpected MKD-related complications. Our study demonstrates that MKD is a common cause of recurrent fever also in the Italian population, where it is associated with both a wide spectrum of previously unreported MVK mutations and peculiar phenotypic features.


Assuntos
Febre Familiar do Mediterrâneo/genética , Fosfotransferases (Aceptor do Grupo Álcool)/deficiência , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Adolescente , Adulto , Amiloidose/etiologia , Criança , Pré-Escolar , Análise Mutacional de DNA , Febre Familiar do Mediterrâneo/complicações , Feminino , Humanos , Hipergamaglobulinemia/complicações , Hipergamaglobulinemia/genética , Imunoglobulina D/sangue , Lactente , Itália , Masculino , Mutação
10.
Arthritis Rheum ; 50(9): 2966-9, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15457465

RESUMO

Systemic reactive (AA) amyloidosis, leading to renal failure, is a severe complication of most hereditary periodic fever syndromes. The risk of developing this life-threatening condition varies widely among these disorders, being higher for patients affected by familial Mediterranean fever and tumor necrosis factor receptor-associated periodic syndrome. In spite of an acute-phase response during attacks, amyloidosis has never, to date, been described in patients affected with the hyperimmunoglobulinemia D with periodic fever syndrome (HIDS). This is the first report to describe the occurrence of renal AA amyloidosis causing severe nephrotic syndrome in a young Italian man affected with HIDS. The diagnosis of HIDS was established according to clinical, laboratory, and genetic criteria as required by the international Nijmegen HIDS registry. In this patient, 2 mutations in the mevalonate kinase gene were identified, one of which, the leucine-to-arginine substitution at codon 265, is novel.


Assuntos
Amiloidose/imunologia , Febre Familiar do Mediterrâneo/genética , Febre Familiar do Mediterrâneo/imunologia , Hipergamaglobulinemia/imunologia , Adulto , Amiloidose/etiologia , Febre Familiar do Mediterrâneo/complicações , Humanos , Hipergamaglobulinemia/complicações , Imunoglobulina D/imunologia , Masculino , Síndrome Nefrótica/etiologia , Síndrome Nefrótica/fisiopatologia , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Proteína Amiloide A Sérica/imunologia
11.
Anticancer Res ; 23(5A): 3727-34, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14666670

RESUMO

Mammalian cells transfected with the S. cerevisiae APN1 protein acquire resistance to oxidizing agents, the damage of which is mainly repaired via DNA base excision repair (BER). We have recently hypothesized that this effect might be linked to the possible capacity of APN1 to accelerate mammalian BER by its 3' diesterase activity. We have investigated here the effect of pure APN1 protein on BER performed by mouse embryonic fibroblast extracts. No significant acceleration was observed in the repair of either a single AP site cleaved by the bifunctional glycosylase NTH of E. coli or the repair of a single 8-oxoguanine, initiated by the bifunctional glycosylase OGG1. Similarly, no significant effect was observed on the repair of a single U (initiated by the monofunctional glycosylase U DNA glycosylase) or the repair of a single natural abasic site. The inability of APN1 to increase the efficiency of BER initiated by bifunctional glycosylases indicates that removal of 3' blocking fragments is not the rate-limiting step of this repair pathway.


Assuntos
Reparo do DNA/efeitos dos fármacos , Endodesoxirribonucleases/farmacologia , Guanina/análogos & derivados , Proteínas de Saccharomyces cerevisiae/farmacologia , Animais , Células Cultivadas , DNA Glicosilases/metabolismo , Reparo do DNA/fisiologia , Enzimas Reparadoras do DNA , Endodesoxirribonucleases/genética , Endodesoxirribonucleases/isolamento & purificação , Endodesoxirribonucleases/metabolismo , Fibroblastos , Glutationa Transferase/genética , Glutationa Transferase/isolamento & purificação , Glutationa Transferase/metabolismo , Glutationa Transferase/farmacologia , Guanina/metabolismo , Camundongos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/isolamento & purificação , Proteínas de Saccharomyces cerevisiae/metabolismo
12.
Teratog Carcinog Mutagen ; Suppl 1: 113-21, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12616602

RESUMO

8-oxo-7,8-dihydroguanine (8-oxoG) is a potent mutagenic lesion that forms at elevated levels in cellular DNA and is repaired with low efficiency in human cells. Unlike its human counterpart, the Drosophila S3 ribosomal/repair protein is endowed with a vigorous 8 oxoG repair activity that is associated to beta,delta-elimination AP lyase activity. We have recently observed that pure GST-tagged Drosophila S3 protein can significantly accelerate the in vitro repair of 8 oxoG performed by human and mouse cell extracts [Cappelli et al., unpublished data]. In this work, we have transfected Chinese hamster cells with mammalian expression vectors containing the Drosophila S3 cDNA. The cells synthesized both S3 mRNA and protein but no improved repair of 8 oxoguanine was observed. Factors important for the proper expression of Drosophila genes in mammalian cells are discussed.


Assuntos
Reparo do DNA/genética , Proteínas de Drosophila/genética , Guanina/análogos & derivados , Guanina/metabolismo , Proteínas Ribossômicas/genética , Animais , Células CHO , Clonagem Molecular , Sequência Consenso , Cricetinae , DNA Complementar/biossíntese , DNA Complementar/genética , Proteínas de Drosophila/biossíntese , Vetores Genéticos , Proteínas Ribossômicas/biossíntese , Transfecção
13.
Anticancer Res ; 22(5): 2797-804, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12529999

RESUMO

Mammalian cells transfected with the S. cerevisiae APN1 protein acquire resistance to oxidizing agents, the damage of which are mainly repaired via DNA base excision repair (BER). We have recently hypothesized that this effect might be linked to the possible capacity of APN1 to accelerate mammalian BER by its 3' diesterase activity. We have investigated here the effect of pure APN1 protein on BER performed by mouse embryonic fibroblast extracts. No significant acceleration was observed in the repair of either a single AP site cleaved by the bifunctional glycosylase NTH of E. coli or the repair of a single 8-oxoguanine, initiated by the bifunctional glycosylase OGG1. Similarly, no significant effect was observed on the repair of a single U (initiated by the monofunctional glycosylase U DNA glycosylase) or the repair of a single natural abasic site. The inability of APN1 to increase the efficiency of BER initiated by bifunctional glycosylases indicates that removal of 3' blocking fragments is not the rate limiting step of this repair pathway.


Assuntos
Reparo do DNA/efeitos dos fármacos , Endodesoxirribonucleases/farmacologia , Proteínas de Saccharomyces cerevisiae/farmacologia , Animais , Extratos Celulares/química , Reparo do DNA/fisiologia , Enzimas Reparadoras do DNA , DNA-Formamidopirimidina Glicosilase , Endodesoxirribonucleases/biossíntese , Endodesoxirribonucleases/genética , Endodesoxirribonucleases/isolamento & purificação , Fibroblastos/química , Glutationa Transferase/biossíntese , Glutationa Transferase/genética , Glutationa Transferase/isolamento & purificação , Glutationa Transferase/farmacologia , Camundongos , N-Glicosil Hidrolases/metabolismo , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/farmacologia , Proteínas de Saccharomyces cerevisiae/biossíntese , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/isolamento & purificação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA