Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 30(6): 751-6, 2011 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-21057543

RESUMO

Characterization of the direct effects of DNA-damaging agents shows how DNA lesions lead to specific mutations. Yet, serum from Hiroshima survivors, Chernobyl liquidators and radiotherapy patients can induce a clastogenic effect on naive cells, showing indirect induction of genomic instability that persists years after exposure. Such indirect effects are not restricted to ionizing radiation, as chemical genotoxins also induce heritable and transmissible genomic instability phenotypes. Although such indirect induction of genomic instability is well described, the underlying mechanism has remained enigmatic. Here, we show that mouse embryonic stem cells exposed to γ-radiation bear the effects of the insult for weeks. Specifically, conditioned media from the progeny of exposed cells can induce DNA damage and homologous recombination in naive cells. Notably, cells exposed to conditioned media also elicit a genome-destabilizing effect on their neighbouring cells, thus demonstrating transmission of genomic instability. Moreover, we show that the underlying basis for the memory of an insult is completely dependent on two of the major DNA cytosine methyltransferases, Dnmt1 and Dnmt3a. Targeted disruption of these genes in exposed cells completely eliminates transmission of genomic instability. Furthermore, transient inactivation of Dnmt1, using a tet-suppressible allele, clears the memory of the insult, thus protecting neighbouring cells from indirect induction of genomic instability. We have thus demonstrated that a single exposure can lead to long-term, genome-destabilizing effects that spread from cell to cell, and we provide a specific molecular mechanism for these persistent bystander effects. Collectively, our results impact the current understanding of risks from toxin exposures and suggest modes of intervention for suppressing genomic instability in people exposed to carcinogenic genotoxins.


Assuntos
Efeito Espectador/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Dano ao DNA , Células-Tronco Embrionárias/enzimologia , Células-Tronco Embrionárias/efeitos da radiação , Instabilidade Genômica , Animais , Células Cultivadas , Meios de Cultivo Condicionados/química , Meios de Cultivo Condicionados/toxicidade , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/genética , DNA Metiltransferase 3A , Células-Tronco Embrionárias/efeitos dos fármacos , Raios gama , Camundongos
2.
Oncogene ; 25(31): 4267-75, 2006 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-16532033

RESUMO

Irradiated cells induce chromosomal instability in unirradiated bystander cells in vitro. Although bystander effects are thought to be linked to radiation-induced secondary cancers, almost no studies have evaluated bystander effects in vivo. Furthermore, it has been proposed that epigenetic changes mediate bystander effects, but few studies have evaluated epigenetic factors in bystander tissues in vivo. Here, we describe studies in which mice were unilaterally exposed to X-irradiation and the levels of DNA damage, DNA methylation and protein expression were evaluated in irradiated and bystander cutaneous tissue. The data show that X-ray exposure to one side of the animal body induces DNA strand breaks and causes an increase in the levels of Rad51 in unexposed bystander tissue. In terms of epigenetic changes, unilateral radiation suppresses global methylation in directly irradiated tissue, but not in bystander tissue at given time-points studied. Intriguingly, however, we observed a significant reduction in the levels of the de novo DNA methyltransferases DNMT3a and 3b and a concurrent increase in the levels of the maintenance DNA methyltransferase DNMT1 in bystander tissues. Furthermore, the levels of two methyl-binding proteins known to be involved in transcriptional silencing, MeCP2 and MBD2, were also increased in bystander tissue. Together, these results show that irradiation induces DNA damage in bystander tissue more than a centimeter away from directly irradiated tissues, and suggests that epigenetic transcriptional regulation may be involved in the etiology of radiation-induced bystander effects.


Assuntos
Efeito Espectador/efeitos da radiação , Dano ao DNA , DNA/efeitos da radiação , Epigênese Genética/efeitos da radiação , Animais , Camundongos , Pele/efeitos da radiação
3.
DNA Repair (Amst) ; 1(8): 645-59, 2002 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-12509287

RESUMO

DNA glycosylases, such as the Mag1 3-methyladenine (3MeA) DNA glycosylase, initiate the base excision repair (BER) pathway by removing damaged bases to create abasic apurinic/apyrimidinic (AP) sites that are subsequently repaired by downstream BER enzymes. Although unrepaired base damage may be mutagenic or recombinogenic, BER intermediates (e.g. AP sites and strand breaks) may also be problematic. To investigate the molecular basis for methylation-induced homologous recombination events in Saccharomyces cerevisiae, spontaneous and methylation-induced recombination were studied in strains with varied MAG1 expression levels. We show that cells lacking Mag1 have increased susceptibility to methylation-induced recombination, and that disruption of nucleotide excision repair (NER; rad4) in mag1 cells increases cellular susceptibility to these events. Furthermore, expression of Escherichia coli Tag 3MeA DNA glycosylase suppresses recombination events, providing strong evidence that unrepaired 3MeA lesions induce recombination. Disruption of REV3 (required for polymerase zeta (Pol zeta)) in mag1 rad4 cells causes increased susceptibility to methylation-induced toxicity and recombination, suggesting that Pol zeta can replicate past 3MeAs. However, at subtoxic levels of methylation damage, disruption of REV3 suppresses methylation-induced recombination, indicating that the effects of Pol zeta on recombination are highly dose-dependent. We also show that overproduction of Mag1 can increase the levels of spontaneous recombination, presumably due to increased levels of BER intermediates. However, additional APN1 endonuclease expression or disruption of REV3 does not affect MAG1-induced recombination, suggesting that downstream BER intermediates (e.g. single strand breaks) are responsible for MAG1-induced recombination, rather than uncleaved AP sites. Thus, too little Mag1 sensitizes cells to methylation-induced recombination, while too much Mag1 can put cells at risk of recombination induced by single strand breaks formed during BER.


Assuntos
DNA Glicosilases , Metilação de DNA , Reparo do DNA/genética , DNA Polimerase Dirigida por DNA/farmacologia , N-Glicosil Hidrolases/fisiologia , Recombinação Genética , Saccharomyces cerevisiae/enzimologia , Ácido Apurínico/química , Ácido Apurínico/metabolismo , Sobrevivência Celular/genética , Dano ao DNA , DNA Fúngico/genética , DNA Polimerase Dirigida por DNA/genética , DNA Polimerase Dirigida por DNA/metabolismo , Escherichia coli/enzimologia , Pirimidinas/química , Pirimidinas/metabolismo , Proteínas de Saccharomyces cerevisiae/genética
4.
J Bacteriol ; 183(1): 131-8, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11114909

RESUMO

Nitric oxide (NO(.)) is critical to numerous biological processes, including signal transduction and macrophage-mediated immunity. In this study, we have explored the biological effects of NO(.)-induced DNA damage on Escherichia coli. The relative importance of base excision repair, nucleotide excision repair (NER), and recombinational repair in preventing NO(.)-induced toxicity was determined. E. coli strains lacking either NER or DNA glycosylases (including those that repair alkylation damage [alkA tag strain], oxidative damage [fpg nei nth strain], and deaminated cytosine [ung strain]) showed essentially wild-type levels of NO(.) resistance. However, apyrimidinic/apurinic (AP) endonuclease-deficient cells (xth nfo strain) were very sensitive to killing by NO(.), which indicates that normal processing of abasic sites is critical for defense against NO(.). In addition, recA mutant cells were exquisitely sensitive to NO(.)-induced killing. Both SOS-deficient (lexA3) and Holliday junction resolvase-deficient (ruvC) cells were very sensitive to NO(.), indicating that both SOS and recombinational repair play important roles in defense against NO(.). Furthermore, strains specifically lacking double-strand end repair (recBCD strains) were very sensitive to NO(.), which suggests that NO(.) exposure leads to the formation of double-strand ends. One consequence of these double-strand ends is that NO(.) induces homologous recombination at a genetically engineered substrate. Taken together, it is now clear that, in addition to the known point mutagenic effects of NO(.), it is also important to consider recombination events among the spectrum of genetic changes that NO(. ) can induce. Furthermore, the importance of recombinational repair for cellular survival of NO(.) exposure reveals a potential susceptibility factor for invading microbes.


Assuntos
Reparo do DNA/genética , Proteínas de Escherichia coli , Escherichia coli/efeitos dos fármacos , Escherichia coli/crescimento & desenvolvimento , Óxido Nítrico/farmacologia , Recombinação Genética , Carbono-Oxigênio Liases/genética , Carbono-Oxigênio Liases/metabolismo , Dano ao DNA , DNA Glicosilases , DNA Bacteriano/genética , DNA Bacteriano/metabolismo , DNA Liase (Sítios Apurínicos ou Apirimidínicos) , Desoxirribonuclease IV (Fago T4-Induzido) , Escherichia coli/genética , Mutação , N-Glicosil Hidrolases/genética , N-Glicosil Hidrolases/metabolismo , Raios Ultravioleta
5.
Nucleic Acids Res ; 28(17): 3294-300, 2000 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-10954597

RESUMO

3-Methyladenine (3MeA) DNA glycosylases initiate base excision repair by removing 3MeA. These glycosylases also remove a broad spectrum of spontaneous and environmentally induced base lesions in vitro. Mouse cells lacking the Aag 3MeA DNA glycosylase (also known as the Mpg, APNG or ANPG DNA glycosylase) are susceptible to 3MeA-induced S phase arrest, chromosome aberrations and apoptosis, but it is not known if Aag is solely responsible for repair of 3MeA in vivo. Here we show that in AAG:(-/-) cells, 3MeA lesions disappear from the genome slightly faster than would be expected by spontaneous depurination alone, suggesting that there may be residual repair of 3MeA. However, repair of 3MeA is at least 10 times slower in AAG:(-/-) cells than in AAG:(+/+) cells. Consequently, 24 h after exposure to [(3)H]MNU, 30% of the original 3MeA burden is intact in AAG:(-/-) cells, while 3MeA is undetectable in AAG:(+/+) cells. Thus, Aag is the major DNA glycosylase for 3MeA repair. We also investigated the in vivo repair kinetics of another Aag substrate, 7-methylguanine. Surprisingly, 7-methylguanine is removed equally efficiently in AAG:(+/+) and AAG:(-/-) cells, suggesting that another DNA glycosylase acts on lesions previously thought to be repaired by Aag.


Assuntos
Adenina/análogos & derivados , Adutos de DNA/metabolismo , Dano ao DNA/genética , DNA Glicosilases , Metilação de DNA , Reparo do DNA/genética , Guanina/análogos & derivados , Guanosina/análogos & derivados , N-Glicosil Hidrolases/deficiência , N-Glicosil Hidrolases/metabolismo , Adenina/química , Adenina/metabolismo , Animais , Linhagem Celular , Adutos de DNA/química , Adutos de DNA/efeitos dos fármacos , Adutos de DNA/genética , Dano ao DNA/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , Deleção de Genes , Genótipo , Guanina/metabolismo , Guanosina/genética , Guanosina/metabolismo , Metilnitrosoureia/toxicidade , Camundongos , Camundongos Knockout , N-Glicosil Hidrolases/genética
6.
Mutagenesis ; 14(3): 339-47, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10375003

RESUMO

We have generated mice deficient in O6-methylguanine DNA methyltransferase activity encoded by the murine Mgmt gene using homologous recombination to delete the region encoding the Mgmt active site cysteine. Tissues from Mgmt null mice displayed very low O6-methylguanine DNA methyltransferase activity, suggesting that Mgmt constitutes the major, if not the only, O6-methylguanine DNA methyltransferase. Primary mouse embryo fibroblasts and bone marrow cells from Mgmt -/- mice were significantly more sensitive to the toxic effects of the chemotherapeutic alkylating agents 1,3-bis(2-chloroethyl)-1-nitrosourea, streptozotocin and temozolomide than those from Mgmt wild-type mice. As expected, Mgmt-deficient fibroblasts and bone marrow cells were not sensitive to UV light or to the crosslinking agent mitomycin C. In addition, the 50% lethal doses for Mgmt -/- mice were 2- to 10-fold lower than those for Mgmt +/+ mice for 1,3-bis(2chloroethyl)-1-nitrosourea, N-methyl-N-nitrosourea and streptozotocin; similar 50% lethal doses were observed for mitomycin C. Necropsies of both wild-type and Mgmt -/mice following drug treatment revealed histological evidence of significant ablation of hematopoietic tissues, but such ablation occurred at much lower doses for the Mgmt -/- mice. These results demonstrate the critical importance of O6-methylguanine DNA methyltransferase in protecting cells and animals against the toxic effects of alkylating agents used for cancer chemotherapy.


Assuntos
Alquilantes/toxicidade , O(6)-Metilguanina-DNA Metiltransferase/genética , Animais , Antibióticos Antineoplásicos/farmacologia , Antineoplásicos Alquilantes/farmacologia , Carcinógenos/farmacologia , Carmustina/farmacologia , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Genótipo , Sistema Hematopoético/anatomia & histologia , Fígado/enzimologia , Metilnitronitrosoguanidina/farmacologia , Camundongos , Camundongos Knockout , Mitomicina/farmacologia , Modelos Biológicos , Estreptozocina/farmacologia , Temozolomida
7.
Cancer Res ; 58(17): 3965-73, 1998 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-9731510

RESUMO

DNA repair status is recognized as an important determinant of the clinical efficacy of cancer chemotherapy. To assess the role that a mammalian DNA glycosylase plays in modulating the toxicity and clastogenicity of the chemotherapeutic DNA cross-linking alkylating agents, we compared the sensitivity of wild-type murine cells to that of isogenic cells bearing homozygous null mutations in the 3-methyladenine DNA glycosylase gene (Aag). We show that Aag protects against the toxic and clastogenic effects of 1,3-bis(2-chloroethyl)-1-nitrosourea and mitomycin C (MMC), as measured by cell killing, sister chromatid exchange, and chromosome aberrations. This protection is accompanied by suppression of apoptosis and a slightly reduced p53 response. Our results identify 3-methyladenine DNA glycosylase-initiated base excision repair as a potentially important determinant of the clinical efficacy and, possibly, the carcinogenicity of these widely used chemotherapeutic agents. However, Aag does not contribute significantly to protection against the toxic and clastogenic effects of several chemotherapeutic nitrogen mustards (namely, mechlorethamine, melphalan, and chlorambucil), at least in the mouse embryonic stem cells used here. We also compare the Aag null phenotype with the Fanconi anemia phenotype, a human disorder characterized by cellular hypersensitivity to DNA cross-linking agents, including MMC. Although Aag null cells are sensitive to MMC-induced growth delay and cell cycle arrest, their sensitivity is modest compared to that of Fanconi anemia cells.


Assuntos
Antineoplásicos Alquilantes/toxicidade , Carmustina/toxicidade , Reparo do DNA , Guanina/análogos & derivados , Mitomicina/toxicidade , Mutagênicos/toxicidade , N-Glicosil Hidrolases/farmacologia , Animais , Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , DNA Glicosilases , Fase G2/efeitos dos fármacos , Guanina/metabolismo , Humanos , Camundongos , Mitose/efeitos dos fármacos
8.
J Exp Med ; 187(11): 1735-43, 1998 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-9607915

RESUMO

To investigate the possible involvement of DNA repair in the process of somatic hypermutation of rearranged immunoglobulin variable (V) region genes, we have analyzed the occurrence, frequency, distribution, and pattern of mutations in rearranged Vlambda1 light chain genes from naive and memory B cells in DNA repair-deficient mutant mouse strains. Hypermutation was found unaffected in mice carrying mutations in either of the following DNA repair genes: xeroderma pigmentosum complementation group (XP)A and XPD, Cockayne syndrome complementation group B (CSB), mutS homologue 2 (MSH2), radiation sensitivity 54 (RAD54), poly (ADP-ribose) polymerase (PARP), and 3-alkyladenine DNA-glycosylase (AAG). These results indicate that both subpathways of nucleotide excision repair, global genome repair, and transcription-coupled repair are not required for somatic hypermutation. This appears also to be true for mismatch repair, RAD54-dependent double-strand-break repair, and AAG-mediated base excision repair.


Assuntos
Linfócitos B/imunologia , Reparo do DNA/fisiologia , Rearranjo Gênico do Linfócito B , Genes de Imunoglobulinas , Memória Imunológica/imunologia , Mutação , Animais , Região Variável de Imunoglobulina/genética , Cadeias lambda de Imunoglobulina/genética , Camundongos , Camundongos Mutantes , Reação em Cadeia da Polimerase
9.
J Biol Chem ; 273(9): 5412-8, 1998 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-9479003

RESUMO

DNA-damaging agents produce a plethora of cellular responses that include p53 induction, cell cycle arrest, and apoptosis. It is generally assumed that it is the DNA damage produced by these agents that triggers such responses, but there is limited direct evidence to support this assumption. Here, we used DNA alkylation repair proficient and deficient isogenic mouse cell lines to demonstrate that the signal to trigger p53 induction, cell cycle arrest, and apoptosis in response to alkylating agents does emanate from DNA damage. Moreover, we established that 3-methyladenine, a relatively minor DNA lesion produced by most methylating agents (which form mainly 7-methylguanine), can specifically induce sister chromatid exchange, chromatid and chromosome gaps and breaks, S phase arrest, the accumulation of p53, and apoptosis. This study was made possible by the generation of 3-methyladenine DNA glycosylase null mutant cells by targeted homologous recombination and by the chemical synthesis of a methylating agent that almost exclusively produces 3-methyladenine DNA lesions. The combined use of these two experimental tools has defined the biological consequences of 3-methyladenine, a DNA lesion produced by endogenous cellular metabolites, environmental carcinogens, and chemotherapeutic alkylating agents.


Assuntos
Adenina/análogos & derivados , Dano ao DNA , DNA Glicosilases , Reparo do DNA , N-Glicosil Hidrolases/deficiência , Adenina/metabolismo , Alquilantes , Animais , Apoptose , Linhagem Celular , Aberrações Cromossômicas , Adutos de DNA/metabolismo , Metilação de DNA , Camundongos , Netropsina/análogos & derivados , Netropsina/farmacologia , Troca de Cromátide Irmã , Proteína Supressora de Tumor p53/biossíntese
10.
Proc Natl Acad Sci U S A ; 94(24): 13087-92, 1997 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-9371804

RESUMO

3-methyladenine (3MeA) DNA glycosylases remove 3MeAs from alkylated DNA to initiate the base excision repair pathway. Here we report the generation of mice deficient in the 3MeA DNA glycosylase encoded by the Aag (Mpg) gene. Alkyladenine DNA glycosylase turns out to be the major DNA glycosylase not only for the cytotoxic 3MeA DNA lesion, but also for the mutagenic 1,N6-ethenoadenine (epsilonA) and hypoxanthine lesions. Aag appears to be the only 3MeA and hypoxanthine DNA glycosylase in liver, testes, kidney, and lung, and the only epsilonA DNA glycosylase in liver, testes, and kidney; another epsilonA DNA glycosylase may be expressed in lung. Although alkyladenine DNA glycosylase has the capacity to remove 8-oxoguanine DNA lesions, it does not appear to be the major glycosylase for 8-oxoguanine repair. Fibroblasts derived from Aag -/- mice are alkylation sensitive, indicating that Aag -/- mice may be similarly sensitive.


Assuntos
DNA Glicosilases , Reparo do DNA/genética , N-Glicosil Hidrolases/metabolismo , Alquilantes/farmacologia , Animais , Cruzamentos Genéticos , DNA Complementar , Feminino , Genótipo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , N-Glicosil Hidrolases/genética
11.
EMBO J ; 15(4): 945-52, 1996 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-8631315

RESUMO

In Escherichia coli, the repair of 3-methyladenine (3MeA) DNA lesions prevents alkylation-induced cell death because unrepaired 3MeA blocks DNA replication. Whether this lesion is cytotoxic to mammalian cells has been difficult to establish in the absence of 3MeA repair-deficient cell lines. We previously isolated and characterized a mouse 3MeA DNA glycosylase cDNA (Aag) that provides resistance to killing by alkylating agents in E. coli. To determine the in vivo role of Aag, we cloned a large fragment of the Aag gene and used it to create Aag-deficient mouse cells by targeted homologous recombination. Aag null cells have no detectable Aag transcripts or 3MeA DNA glycosylase activity. The loss of Aag renders cells significantly more sensitive to methyl methanesulfonate-induced chromosome damage, and to cell killing induced by two methylating agents, one of which produces almost exclusively 3MeAs. Aag null embryonic stem cells become sensitive to two cancer chemotherapeutic alkylating agents, namely 1,3-bis(2-chloroethyl)-1-nitrosourea and mitomycin C, indicating that Aag status is an important determinant of cellular resistance to these agents. We conclude that this mammalian 3MeA DNA glycosylase plays a pivotal role in preventing alkylation-induced chromosome damage and cytotoxicity.


Assuntos
Dano ao DNA , DNA Glicosilases , N-Glicosil Hidrolases/metabolismo , Alquilantes/toxicidade , Animais , Divisão Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Reparo do DNA , Camundongos , Camundongos Knockout , Mutagênese , Troca de Cromátide Irmã , Raios Ultravioleta
12.
Carcinogenesis ; 14(2): 175-81, 1993 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8435858

RESUMO

In Escherichia coli, the repair of 3-methyladenine (3MeA) DNA lesions by DNA glycosylases prevents alkylation induced cell death. We described previously the isolation of a human 3MeA DNA glycosylase (AAG) cDNA that maps to chromosome 16 and hybridizes to specific genomic DNA fragments from a number of mammals, including mouse. As a first step in the generation of a 3MeA DNA glycosylase deficient mouse by homologous replacement in embryonic stem cells, we have cloned the mouse 3MeA DNA glycosylase cDNA. The cloned 1095 base pair cDNA contains a complete 333 amino acid open reading frame that predicts a 36.5 kDa protein and hybridizes to a 1.5 kb mRNA transcript. Mouse 3MeA DNA glycosylase (Aag) transcript levels vary by up to 21 fold among tissues, being highest in the testes and lowest in the heart. The Aag cDNA encodes a glycosylase able to release 3MeA, 7-methylguanine (7MeG) and 3-methylguanine (3MeG) from alkylated DNA. The expression of Aag in E. coli provides substantial resistance against killing by methylating agents, but, unlike its E. coli counterparts, the Aag glycosylase fails to protect against killing by ethylating and propylating agents. A 232 amino acid stretch of the predicted mouse protein shares extensive amino acid identity with rat (93%) and human (83%) 3MeA DNA glycosylases and we observe that all three mammalian glycosylases have a bipartite nuclear localization signal. The Aag gene maps to mouse chromosome 11, suggesting a segment of conserved synteny between mouse chromosome 11 and human chromosome 16, which bears the human 3MeA DNA glycosylase gene. Cloning the mouse 3MeA DNA glycosylase cDNA is a step toward understanding the role of this DNA repair enzyme in mammals.


Assuntos
Adenina/análogos & derivados , Cromossomos/fisiologia , DNA Glicosilases , Reparo do DNA/fisiologia , DNA/isolamento & purificação , Guanina/análogos & derivados , N-Glicosil Hidrolases/genética , Adenina/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Southern Blotting , Núcleo Celular/fisiologia , Mapeamento Cromossômico , Clonagem Molecular , Escherichia coli/enzimologia , Escherichia coli/genética , Expressão Gênica , Guanina/metabolismo , Humanos , Camundongos , Dados de Sequência Molecular , N-Glicosil Hidrolases/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...