Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Cell Cycle ; 22(18): 2088-2096, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37749911

RESUMO

Alcohol contributes to cellular accumulation of acetaldehyde, a primary metabolite of alcohol and a major human carcinogen. Acetaldehyde can form DNA adducts and induce interstrand crosslinks (ICLs) that are repaired by the Fanconi anemia DNA repair pathway (FA pathway). Individuals with deficiency in acetaldehyde detoxification or in the FA pathway have an increased risk of squamous-cell carcinomas (SCCs) including those of the esophagus. In a recent report, we described the molecular basis of acetaldehyde-induced DNA damage in esophageal keratinocytes [1]. We demonstrated that, at physiologically relevant concentrations, acetaldehyde induces DNA damage at the DNA replication fork. This resulted in replication stress, leading to activation of the ATR-Chk1-dependent cell cycle checkpoints. We also reported that the p53 DNA damage response is elevated in response to acetaldehyde and that the FA pathway limits acetaldehyde-induced genomic instability. Here, we highlight these findings and present additional results to discuss the role of the FA pathway and p53 DNA damage response in the protection against genomic instability and esophageal carcinogenesis.


Assuntos
Acetaldeído , Anemia de Fanconi , Humanos , Acetaldeído/toxicidade , Acetaldeído/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Dano ao DNA , Etanol , Instabilidade Genômica , Reparo do DNA , Esôfago/metabolismo , Queratinócitos/metabolismo , Replicação do DNA
2.
Hum Genet ; 141(12): 1811-1836, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35596788

RESUMO

Fanconi anemia is a genetic disorder that is characterized by bone marrow failure, as well as a predisposition to malignancies including leukemia and squamous cell carcinoma (SCC). At least 22 genes are associated with Fanconi anemia, constituting the Fanconi anemia DNA repair pathway. This pathway coordinates multiple processes and proteins to facilitate the repair of DNA adducts including interstrand crosslinks (ICLs) that are generated by environmental carcinogens, chemotherapeutic crosslinkers, and metabolic products of alcohol. ICLs can interfere with DNA transactions, including replication and transcription. If not properly removed and repaired, ICLs cause DNA breaks and lead to genomic instability, a hallmark of cancer. In this review, we will discuss the genetic and phenotypic characteristics of Fanconi anemia, the epidemiology of the disease, and associated cancer risk. The sources of ICLs and the role of ICL-inducing chemotherapeutic agents will also be discussed. Finally, we will review the detailed mechanisms of ICL repair via the Fanconi anemia DNA repair pathway, highlighting critical regulatory processes. Together, the information in this review will underscore important contributions to Fanconi anemia research in the past two decades.


Assuntos
Anemia de Fanconi , Neoplasias , Humanos , Anemia de Fanconi/epidemiologia , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Replicação do DNA , Reparo do DNA/genética , Dano ao DNA , Neoplasias/epidemiologia , Neoplasias/genética
3.
Biomolecules ; 11(10)2021 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-34680112

RESUMO

BACKGROUND: Alcohol (ethanol) consumption is a major risk factor for head and neck and esophageal squamous cell carcinomas (SCCs). However, how ethanol (EtOH) affects SCC homeostasis is incompletely understood. METHODS: We utilized three-dimensional (3D) organoids and xenograft tumor transplantation models to investigate how EtOH exposure influences intratumoral SCC cell populations including putative cancer stem cells defined by high CD44 expression (CD44H cells). RESULTS: Using 3D organoids generated from SCC cell lines, patient-derived xenograft tumors, and patient biopsies, we found that EtOH is metabolized via alcohol dehydrogenases to induce oxidative stress associated with mitochondrial superoxide generation and mitochondrial depolarization, resulting in apoptosis of the majority of SCC cells within organoids. However, CD44H cells underwent autophagy to negate EtOH-induced mitochondrial dysfunction and apoptosis and were subsequently enriched in organoids and xenograft tumors when exposed to EtOH. Importantly, inhibition of autophagy increased EtOH-mediated apoptosis and reduced CD44H cell enrichment, xenograft tumor growth, and organoid formation rate. CONCLUSIONS: This study provides mechanistic insights into how EtOH may influence SCC cells and establishes autophagy as a potential therapeutic target for the treatment of EtOH-associated SCC.


Assuntos
Autofagia , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Etanol/metabolismo , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Estresse Oxidativo , Consumo de Bebidas Alcoólicas/metabolismo , Animais , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Humanos , Receptores de Hialuronatos/metabolismo , Potencial da Membrana Mitocondrial , Camundongos SCID , Mitocôndrias/metabolismo , Organoides/patologia , Oxirredução
4.
Mol Oncol ; 15(11): 3109-3124, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34328261

RESUMO

Individuals with Fanconi anemia (FA), a rare genetic bone marrow failure syndrome, have an increased risk of young-onset head and neck squamous cell carcinomas (SCCs) and esophageal SCC. The FA DNA repair pathway is activated upon DNA damage induced by acetaldehyde, a chief alcohol metabolite and one of the major carcinogens in humans. However, the molecular basis of acetaldehyde-induced genomic instability in SCCs of the head and neck and of the esophagus in FA remains elusive. Here, we report the effects of acetaldehyde on replication stress response in esophageal epithelial cells (keratinocytes). Acetaldehyde-exposed esophageal keratinocytes displayed accumulation of DNA damage foci consisting of 53BP1 and BRCA1. At physiologically relevant concentrations, acetaldehyde activated the ATR-Chk1 pathway, leading to S- and G2/M-phase delay with accumulation of the FA complementation group D2 protein (FANCD2) at the sites of DNA synthesis, suggesting that acetaldehyde impedes replication fork progression. Consistently, depletion of the replication fork protection protein Timeless led to elevated DNA damage upon acetaldehyde exposure. Furthermore, FANCD2 depletion exacerbated replication abnormalities, elevated DNA damage, and led to apoptotic cell death, indicating that FANCD2 prevents acetaldehyde-induced genomic instability in esophageal keratinocytes. These observations contribute to our understanding of the mechanisms that drive genomic instability in FA patients and alcohol-related carcinogenesis, thereby providing a translational implication in the development of more effective therapies for SCCs.


Assuntos
Anemia de Fanconi , Acetaldeído/metabolismo , Acetaldeído/toxicidade , Dano ao DNA , Reparo do DNA/genética , Replicação do DNA/genética , Esôfago/patologia , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Anemia de Fanconi/patologia , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/genética , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/metabolismo , Instabilidade Genômica , Humanos , Queratinócitos/metabolismo
5.
Curr Genet ; 66(3): 593-605, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32034465

RESUMO

Formaldehyde is a common environmental pollutant and is associated with adverse health effects. Formaldehyde is also considered to be a carcinogen because it can form DNA adducts, leading to genomic instability. How these adducts are prevented and removed is not fully understood. In this study, we used the fission yeast Schizosaccharomyces pombe as a model organism to investigate cellular tolerance pathways against formaldehyde exposure. We show that Fmd1 is a major formaldehyde dehydrogenase that functions to detoxify formaldehyde and that Fmd1 is critical to minimize formaldehyde-mediated DNA lesions. Our investigation revealed that nucleotide excision repair and homologous recombination have major roles in cellular tolerance to formaldehyde, while mutations in the Fanconi anemia, translesion synthesis, and base excision repair pathways also render cells sensitive to formaldehyde. We also demonstrate that loss of Wss1 or Wss2, proteases involved in the removal of DNA-protein crosslinks, sensitizes cells to formaldehyde and leads to replication defects. These results suggest that formaldehyde generates a variety of DNA lesions, including interstrand crosslinks, DNA-protein crosslinks, and base adducts. Thus, our genetic studies provide a framework for future investigation regarding health effects resulting from formaldehyde exposure.


Assuntos
Dano ao DNA , Reparo do DNA , Replicação do DNA , Proteínas de Grupos de Complementação da Anemia de Fanconi/metabolismo , Formaldeído/efeitos adversos , Recombinação Homóloga , Schizosaccharomyces/genética , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Formaldeído/toxicidade , Hipersensibilidade Respiratória , Schizosaccharomyces/efeitos dos fármacos , Schizosaccharomyces/metabolismo
6.
Genes (Basel) ; 8(3)2017 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-28272375

RESUMO

All living organisms need to duplicate their genetic information while protecting it from unwanted mutations, which can lead to genetic disorders and cancer development. Inaccuracies during DNA replication are the major cause of genomic instability, as replication forks are prone to stalling and collapse, resulting in DNA damage. The presence of exogenous DNA damaging agents as well as endogenous difficult-to-replicate DNA regions containing DNA-protein complexes, repetitive DNA, secondary DNA structures, or transcribing RNA polymerases, increases the risk of genomic instability and thus threatens cell survival. Therefore, understanding the cellular mechanisms required to preserve the genetic information during S phase is of paramount importance. In this review, we will discuss our current understanding of how cells cope with these natural impediments in order to prevent DNA damage and genomic instability during DNA replication.

7.
Cell Cycle ; 16(1): 45-58, 2017 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-27687866

RESUMO

Acetaldehyde, a primary metabolite of alcohol, forms DNA adducts and disrupts the DNA replication process, causing genomic instability, a hallmark of cancer. Indeed, chronic alcohol consumption accounts for approximately 3.6% of all cancers worldwide. However, how the adducts are prevented and repaired after acetaldehyde exposure is not well understood. In this report, we used the fission yeast Schizosaccharomyces pombe as a model organism to comprehensively understand the genetic controls of DNA damage avoidance in response to acetaldehyde. We demonstrate that Atd1 functions as a major acetaldehyde detoxification enzyme that prevents accumulation of Rad52-DNA repair foci, while Atd2 and Atd3 have minor roles in acetaldehyde detoxification. We found that acetaldehyde causes DNA damage at the replication fork and activates the cell cycle checkpoint to coordinate cell cycle arrest with DNA repair. Our investigation suggests that acetaldehyde-mediated DNA adducts include interstrand-crosslinks and DNA-protein crosslinks. We also demonstrate that acetaldehyde activates multiple DNA repair pathways. Nucleotide excision repair and homologous recombination, which are both epistatically linked to the Fanconi anemia pathway, have major roles in acetaldehyde tolerance, while base excision repair and translesion synthesis also contribute to the prevention of acetaldehyde-dependent genomic instability. We also show the involvement of Wss1-related metalloproteases, Wss1 and Wss2, in acetaldehyde tolerance. These results indicate that acetaldehyde causes cellular stresses that require cells to coordinate multiple cellular processes in order to prevent genomic instability. Considering that acetaldehyde is a human carcinogen, our genetic studies serve as a guiding investigation into the mechanisms of acetaldehyde-dependent genomic instability and carcinogenesis.


Assuntos
Acetaldeído/toxicidade , Dano ao DNA/genética , Schizosaccharomyces/genética , Adaptação Fisiológica/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Adutos de DNA/metabolismo , Reparo do DNA/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , DNA Fúngico/biossíntese , Epistasia Genética/efeitos dos fármacos , Anemia de Fanconi/genética , Genes Fúngicos , Recombinação Homóloga/efeitos dos fármacos , Inativação Metabólica/efeitos dos fármacos , Modelos Biológicos , Schizosaccharomyces/citologia , Schizosaccharomyces/efeitos dos fármacos , Proteínas de Schizosaccharomyces pombe/genética , Proteínas de Schizosaccharomyces pombe/metabolismo
8.
Am J Cancer Res ; 6(4): 781-96, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27186430

RESUMO

A polymorphic mutation in the acetaldehyde dehydrogenase 2 (ALDH2) gene has been epidemiologically linked to the high susceptibility to esophageal carcinogenesis for individuals with alcohol use disorders. Mice subjected to alcohol drinking show increased oxidative stress and DNA adduct formation in esophageal epithelia where Aldh2 loss augments alcohol-induced genotoxic effects; however, it remains elusive as to how esophageal epithelial cells with dysfunctional Aldh2 cope with oxidative stress related to alcohol metabolism. Here, we investigated the role of autophagy in murine esophageal epithelial cells (keratinocytes) exposed to ethanol and acetaldehyde. We find that ethanol and acetaldehyde trigger oxidative stress via mitochondrial superoxide in esophageal keratinocytes. Aldh2-deficient cells appeared to be highly susceptible to ethanol- or acetaldehyde-mediated toxicity. Alcohol dehydrogenase-mediated acetaldehyde production was implicated in ethanol-induced cell injury in Aldh2 deficient cells as ethanol-induced oxidative stress and cell death was partially inhibited by 4-methylpyrazole. Acetaldehyde activated autophagy flux in esophageal keratinocytes where Aldh2 deficiency increased dependence on autophagy to cope with ethanol-induced acetaldehyde-mediated oxidative stress. Pharmacological inhibition of autophagy flux by chloroquine stabilized p62/SQSTM1, and increased basal and acetaldehyde-mediate oxidative stress in Aldh2 deficient cells as documented in monolayer culture as well as single-cell derived three-dimensional esophageal organoids, recapitulating a physiological esophageal epithelial proliferation-differentiation gradient. Our innovative approach indicates, for the first time, that autophagy may provide cytoprotection to esophageal epithelial cells responding to oxidative stress that is induced by ethanol and its major metabolite acetaldehyde. Defining autophagymediated cytoprotection against alcohol-induced genotoxicity in the context of Aldh2 deficiency, our study provides mechanistic insights into the tumor suppressor functions of ALDH2 and autophagy in alcohol-related esophageal carcinogenesis.

9.
Curr Genet ; 62(4): 725-730, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27068713

RESUMO

The DNA replication machinery encounters problems at numerous genomic regions that are inherently difficult to replicate. These genomic regions include telomeres, which contain repetitive DNA and telomere-binding proteins. If not properly regulated, replication of such genomic regions can result in DNA damage, leading to genomic instability. Studies implicated a role of Timeless-related proteins at difficult-to-replicate genomic regions, including telomeres. However, how these proteins maintain telomeres was elusive. In a recent report, we described the role of Swi1, a Timeless-related protein, in telomere maintenance in fission yeast. We demonstrated that Swi1 is required for proper replication of repeat DNA sequences at telomeres. We also showed that Swi1-deficient cells utilize recombination-based ALT (alternative lengthening of telomeres)-like mechanisms to maintain telomeres in the absence of telomerase. Here, we highlight these findings and present additional data to discuss the role of Swi1Timeless in telomere protection and ALT prevention.


Assuntos
Telômero/genética , Telômero/metabolismo , Proteínas de Transporte , Proteínas Cromossômicas não Histona/metabolismo , Dano ao DNA , Replicação do DNA , DNA Polimerase Dirigida por DNA/metabolismo , Genoma , Genômica , Ligação Proteica , Recombinases/antagonistas & inibidores , Recombinases/metabolismo , Recombinação Genética , Sequências Repetitivas de Ácido Nucleico , Telomerase/metabolismo , Homeostase do Telômero
10.
PLoS Genet ; 12(3): e1005943, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26990647

RESUMO

Genomic instability associated with DNA replication stress is linked to cancer and genetic pathologies in humans. If not properly regulated, replication stress, such as fork stalling and collapse, can be induced at natural replication impediments present throughout the genome. The fork protection complex (FPC) is thought to play a critical role in stabilizing stalled replication forks at several known replication barriers including eukaryotic rDNA genes and the fission yeast mating-type locus. However, little is known about the role of the FPC at other natural impediments including telomeres. Telomeres are considered to be difficult to replicate due to the presence of repetitive GT-rich sequences and telomere-binding proteins. However, the regulatory mechanism that ensures telomere replication is not fully understood. Here, we report the role of the fission yeast Swi1(Timeless), a subunit of the FPC, in telomere replication. Loss of Swi1 causes telomere shortening in a telomerase-independent manner. Our epistasis analyses suggest that heterochromatin and telomere-binding proteins are not major impediments for telomere replication in the absence of Swi1. Instead, repetitive DNA sequences impair telomere integrity in swi1Δ mutant cells, leading to the loss of repeat DNA. In the absence of Swi1, telomere shortening is accompanied with an increased recruitment of Rad52 recombinase and more frequent amplification of telomere/subtelomeres, reminiscent of tumor cells that utilize the alternative lengthening of telomeres pathway (ALT) to maintain telomeres. These results suggest that Swi1 ensures telomere replication by suppressing recombination and repeat instability at telomeres. Our studies may also be relevant in understanding the potential role of Swi1(Timeless) in regulation of telomere stability in cancer cells.


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas de Ligação a DNA/genética , Instabilidade de Microssatélites , Sequências Repetitivas de Ácido Nucleico/genética , Proteínas de Schizosaccharomyces pombe/genética , Proteínas de Ligação a Telômeros/genética , Replicação do DNA/genética , Instabilidade Genômica , Heterocromatina/genética , Humanos , Proteína Rad52 de Recombinação e Reparo de DNA/genética , Schizosaccharomyces/genética , Telômero/genética , Homeostase do Telômero , Encurtamento do Telômero/genética
11.
Methods Mol Biol ; 1170: 539-47, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24906335

RESUMO

A variety of DNA-binding proteins regulate DNA transactions including DNA replication and DNA damage response. To initiate DNA replication in S phase of the cell cycle, numerous replication proteins must be recruited to the replication origin in order to unwind and synthesize DNA. Some replication factors stay at the origin, while replisome components move with the replication fork. When the replisome encounters DNA damage or other issues during DNA replication, the replication fork stalls and accumulates single-stranded DNA that triggers the ATR-dependent replication checkpoint, in order to slow down S phase and arrest the cell cycle at the G2-M transition. It is also possible that replication forks collapse, leading to double-strand breaks that recruit various DNA damage response proteins to activate cell cycle checkpoints and DNA repair pathways. Therefore, defining the localization of DNA transaction factors during the cell cycle should provide important insights into mechanistic understanding of DNA replication and its related processes. In this chapter, we describe a chromatin immunoprecipitation method to locate replisome components at replication origins in human cells.


Assuntos
Imunoprecipitação da Cromatina/métodos , Genes myc , Origem de Replicação , DNA/genética , DNA/isolamento & purificação , Células HeLa , Humanos , Reação em Cadeia da Polimerase/métodos
13.
Mol Cell ; 48(4): 532-46, 2012 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-23084836

RESUMO

Complex genome organizations participate in various nuclear processes including transcription, DNA replication, and repair. However, the mechanisms that generate and regulate these functional genome structures remain largely unknown. Here, we describe how the Ku heterodimer complex, which functions in nonhomologous end joining, mediates clustering of long terminal repeat retrotransposons at centromeres in fission yeast. We demonstrate that the CENP-B subunit, Abp1, functions as a recruiter of the Ku complex, which in turn loads the genome-organizing machinery condensin to retrotransposons. Intriguingly, histone H3 lysine 56 (H3K56) acetylation, which functions in DNA replication and repair, interferes with Ku localization at retrotransposons without disrupting Abp1 localization and, as a consequence, dissociates condensin from retrotransposons. This dissociation releases condensin-mediated genomic associations during S phase and upon DNA damage. ATR (ATM- and Rad3-related) kinase mediates the DNA damage response of condensin-mediated genome organization. Our study describes a function of H3K56 acetylation that neutralizes condensin-mediated genome organization.


Assuntos
Adenosina Trifosfatases/metabolismo , Ciclo Celular , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Epigênese Genética , Genoma , Histonas/química , Histonas/metabolismo , Lisina/metabolismo , Complexos Multiproteicos/metabolismo , Acetilação , Adenosina Trifosfatases/genética , DNA Fúngico/genética , DNA Fúngico/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas dos Microfilamentos/metabolismo , Complexos Multiproteicos/genética , Proteínas Serina-Treonina Quinases/metabolismo , Fase S , Saccharomyces cerevisiae/citologia , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo
14.
Cell Cycle ; 11(12): 2337-47, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22672906

RESUMO

A variety of telomere protection programs are utilized to preserve telomere structure. However, the complex nature of telomere maintenance remains elusive. The Timeless protein associates with the replication fork and is thought to support efficient progression of the replication fork through natural impediments, including replication fork block sites. However, the mechanism by which Timeless regulates such genomic regions is not understood. Here, we report the role of Timeless in telomere length maintenance. We demonstrate that Timeless depletion leads to telomere shortening in human cells. This length maintenance is independent of telomerase, and Timeless depletion causes increased levels of DNA damage, leading to telomere aberrations. We also show that Timeless is associated with Shelterin components TRF1 and TRF2. Timeless depletion slows telomere replication in vitro, and Timeless-depleted cells fail to maintain TRF1-mediated accumulation of replisome components at telomeric regions. Furthermore, telomere replication undergoes a dramatic delay in Timeless-depleted cells. These results suggest that Timeless functions together with TRF1 to prevent fork collapse at telomere repeat DNA and ensure stable maintenance of telomere length and integrity.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Replicação do DNA , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Telômero/metabolismo , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Dano ao DNA , Células HEK293 , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/genética , Ligação Proteica , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Homeostase do Telômero , Proteína 1 de Ligação a Repetições Teloméricas/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo
15.
Biotechnol J ; 3(9-10): 1280-5, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18729046

RESUMO

The fission yeast Schizosaccharomyces pombe is a popular model organism to study various cellular processes, although research tools available for S. pombe are relatively inadequate. To facilitate genetic and biochemical investigation in S. pombe, we report here a system of vectors for genomic FLAG epitope-tagging. These vectors enable us to amplify gene-targeting fragments for integration into specific loci of the S. pombe genome. All vectors in this report were designed to express FLAG epitope-tagged proteins from their endogenous genomic loci. Vectors for N-terminal FLAG epitope-tagging allow us to control protein expression levels using the wild-type nmt1 promoter, its weaker derivatives, and the urg1 promoter. These vectors are available with various antibiotic markers including kanMX6, hphMX6, natMX6 and bleMX6, and the his3(+) marker. Vectors for C-terminal FLAG epitope-tagging were designed to express FLAG-fusion proteins under the control of their native promoters at their own genomic loci, allowing us to characterize protein functions under physiological conditions. These vectors are available with kanMX6, hphMX6, nat-MX6 and bleMX6 markers. The series of vectors described in this report should prove useful for protein studies in fission yeast.


Assuntos
Vetores Genéticos/genética , Peptídeos/genética , Schizosaccharomyces/genética , Epitopos/genética , Genoma Fúngico/genética , Oligopeptídeos , Proteínas de Schizosaccharomyces pombe/genética
16.
J Biochem ; 134(1): 71-82, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12944373

RESUMO

ts ET24 cells are a novel temperature-sensitive (ts) mutant for cell proliferation of hamster BHK21 cells. The human genomic DNA which rescued the temperature-sensitive lethality of ts ET24 cells was isolated and screened for an open reading frame in the deposited human genomic library. X chromosomal DBX gene encoding the RNA helicase, DEAD-BOX X isoform, which is homologous to yeast Ded1p, was found to be defective in this mutant. The single point mutation (P267S) was localized between the Motifs I and Ia of the hamster DBX of ts ET24 cells. At the nonpermissive temperature of 39.5 degrees C, ts ET24 cells were arrested in the G1-phase and survived for more than 3 days. In ts ET24 cells, total protein synthesis was not reduced at 39.5 degrees C for 24 h, while mRNA accumulated in the nucleus after incubation at 39.5 degrees C for 17 h. The amount of cyclin A mRNA decreased in ts ET24 cells within 4 h after the temperature shift to 39.5 degrees C, consistent with the fact that the entry into the S-phase was delayed by the temperature shift.


Assuntos
Fase G1/genética , RNA Helicases/genética , RNA Helicases/fisiologia , Fase S/genética , Sequência de Aminoácidos , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Cricetinae , Ciclina A/biossíntese , Ciclina B/biossíntese , DNA/química , DNA/genética , DNA Complementar/genética , Fator de Iniciação 4E em Eucariotos/biossíntese , Fase G1/fisiologia , Teste de Complementação Genética , Genoma Humano , Células HeLa , Humanos , Isoenzimas , Dados de Sequência Molecular , Mutação Puntual , RNA Mensageiro/biossíntese , Fase S/fisiologia , Análise de Sequência de Proteína , Temperatura , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA