Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Nursing ; 53(4): 54-61, 2023 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-36946641

RESUMO

PURPOSE: To evaluate the severity and longitudinal trends of depression in critical care nurses caring for patients with COVID-19 in the US during a global pandemic. METHODS: The study employed longitudinal mixed methods. Using the Patient Health Questionnaire (PHQ-9), nurses were sent electronic surveys at baseline, 1 month, and between 3 and 6 months to measure the severity and trends of depression during the prevaccination stage of the COVID-19 pandemic. One-on-one interviews were conducted with critical care nurses to evaluate their depressive symptoms. RESULTS: Forty-eight nurses completed the questionnaire at baseline, 40 completed 1-month surveys, and 20 completed the 3 to 6 month surveys. The mean PHQ-9 score was 5.85, 6.20, and 8.30 at baseline, at 1 month, and at 3 to 6 months, respectively. PHQ-9 scores increased significantly over time (estimate = 1.120, P = .037). The probability of participants being moderately to severely depressed was 0.980 (P = .049) at baseline, 0.990 (P = .013) at 1 month, and 1.0 (P = .002) at 3 to 6 months. Fourteen nurses were included in a single, one-on-one interview. Eight major themes were found in qualitative analyses. For example, nurses expressed fear of spreading COVID-19 to their loved one and community. Common themes identified within the interviews included uncertainty, limited human interaction, fluctuations in mood, life is in my hands, a threat to others, positive and negative coping, nurses as scapegoats, and emerging vulnerability to COVID-19 exposure. All 14 nurses who were interviewed denied accessing any mental health services. CONCLUSIONS: More research is needed to evaluate critical care nurses who care for patients with COVID-19 and their levels of depression to improve practice at the bedside further and develop policies to promote their well-being.


Assuntos
COVID-19 , Enfermeiras e Enfermeiros , Humanos , COVID-19/epidemiologia , Depressão/epidemiologia , Depressão/diagnóstico , Pandemias , Inquéritos e Questionários
2.
J Mol Cell Cardiol ; 88: 101-10, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26388265

RESUMO

The transcriptional regulation of pathological cardiac hypertrophy involves the interplay of transcription factors and chromatin remodeling enzymes. The Microphthalmia-Associated Transcription Factor (MITF) is highly expressed in cardiomyocytes and is required for cardiac hypertrophy. However, the transcriptional mechanisms by which MITF promotes cardiac hypertrophy have not been elucidated. In this study, we tested the hypothesis that MITF promotes cardiac hypertrophy by activating transcription of pro-hypertrophy genes through interactions with the SWI/SNF chromatin remodeling complex. In an in vivo model of cardiac hypertrophy, expression of MITF and the BRG1 subunit of the SWI/SNF complex increased coordinately in response to pressure overload. Expression of MITF and BRG1 also increased in vitro when cardiomyocytes were stimulated with angiotensin II or a ß-adrenergic agonist. Both MITF and BRG1 were required to increase cardiomyocyte size and activate expression of hypertrophy markers in response to ß-adrenergic stimulation. We detected physical interactions between MITF and BRG1 in cardiomyocytes and found that they cooperate to regulate expression of a pro-hypertrophic transcription factor, GATA4. Our data show that MITF binds to the E box element in the GATA4 promoter and facilitates recruitment of BRG1. This is associated with enhanced expression of the GATA4 gene as evidenced by increased Histone3 lysine4 tri-methylation (H3K4me3) on the GATA4 promoter. Thus, in hypertrophic cardiomyoctes, MITF is a key transcriptional activator of a pro-hypertrophic gene, GATA4, and this regulation is dependent upon the BRG1 component of the SWI/SNF complex.


Assuntos
Cardiomegalia/genética , DNA Helicases/genética , Fator de Transcrição GATA4/genética , Fator de Transcrição Associado à Microftalmia/genética , Miócitos Cardíacos/metabolismo , Proteínas Nucleares/genética , Fatores de Transcrição/genética , Angiotensina II/farmacologia , Animais , Aorta/cirurgia , Sequência de Bases , Sítios de Ligação , Cardiomegalia/etiologia , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Linhagem Celular , Constrição Patológica/complicações , Constrição Patológica/cirurgia , DNA Helicases/metabolismo , Fator de Transcrição GATA4/metabolismo , Regulação da Expressão Gênica , Isoproterenol/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fator de Transcrição Associado à Microftalmia/metabolismo , Dados de Sequência Molecular , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Proteínas Nucleares/metabolismo , Cultura Primária de Células , Ligação Proteica , Ratos , Transdução de Sinais , Fatores de Transcrição/metabolismo , Transcrição Gênica
3.
Mutat Res ; 743-744: 53-66, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23391514

RESUMO

The field of DNA mismatch repair (MMR) has rapidly expanded after the discovery of the MutHLS repair system in bacteria. By the mid 1990s yeast and human homologues to bacterial MutL and MutS had been identified and their contribution to hereditary non-polyposis colorectal cancer (HNPCC; Lynch syndrome) was under intense investigation. The human MutS homologue 6 protein (hMSH6), was first reported in 1995 as a G:T binding partner (GTBP) of hMSH2, forming the hMutSα mismatch-binding complex. Signal transduction from each DNA-bound hMutSα complex is accomplished by the hMutLα heterodimer (hMLH1 and hPMS2). Molecular mechanisms and cellular regulation of individual MMR proteins are now areas of intensive research. This review will focus on molecular mechanisms associated with mismatch binding, as well as emerging evidence that MutSα, and in particular, MSH6, is a key protein in MMR-dependent DNA damage response and communication with other DNA repair pathways within the cell. MSH6 is unstable in the absence of MSH2, however it is the DNA lesion-binding partner of this heterodimer. MSH6, but not MSH2, has a conserved Phe-X-Glu motif that recognizes and binds several different DNA structural distortions, initiating different cellular responses. hMSH6 also contains the nuclear localization sequences required to shuttle hMutSα into the nucleus. For example, upon binding to O(6)meG:T, MSH6 triggers a DNA damage response that involves altered phosphorylation within the N-terminal disordered domain of this unique protein. While many investigations have focused on MMR as a post-replication DNA repair mechanism, MMR proteins are expressed and active in all phases of the cell cycle. There is much more to be discovered about regulatory cellular roles that require the presence of MutSα and, in particular, MSH6.


Assuntos
Dano ao DNA , Reparo de Erro de Pareamento de DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteína 2 Homóloga a MutS/genética , Proteína 2 Homóloga a MutS/metabolismo , Animais , Humanos , Transdução de Sinais
4.
Mutat Res ; 706(1-2): 36-45, 2011 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-21035467

RESUMO

DNA mismatch repair (MMR) maintains genomic integrity by correction of mispaired bases and insertion-deletion loops. The MMR pathway can also trigger a DNA damage response upon binding of MutSα to specific DNA lesions such as O(6)methylguanine (O(6)meG). Limited information is available regarding cellular regulation of these two different pathways. Within this report, we demonstrate that phosphorylated hMSH6 increases in concentration in the presence of a G:T mismatch, as compared to an O(6)meG:T lesion. TPA, a kinase activator, enhances the phosphorylation of hMSH6 and binding of hMutSα to a G:T mismatch, though not to O(6)meG:T. UCN-01, a kinase inhibitor, decreases both phosphorylation of hMSH6 and binding of hMutSα to G:T and O(6)meG:T. HeLa MR cells, pretreated with UCN-01 and exposed to MNNG, undergo activation of Cdk1 and mitosis despite phosphorylation of Chk1 and inactivating phosphorylation of Cdc25c. These results indicate that UCN-01 may inhibit an alternative cell cycle arrest pathway associated with the MMR pathway that does not involve Cdc25c. In addition, recombinant hMutSα containing hMSH6 mutated at an N-terminal cluster of four phosphoserines exhibits decreased phosphorylation and decreased binding of hMutSα to G:T and O(6)meG:T. Taken together, these results suggest a model in which the amount of phosphorylated hMSH6 bound to DNA is dependent on the presence of either a DNA mismatch or DNA alkylation damage. We hypothesize that both phosphorylation of hMSH6 and total concentration of bound hMutSα are involved in cellular signaling of either DNA mismatch repair or MMR-dependent damage recognition activities.


Assuntos
Pareamento Incorreto de Bases , Dano ao DNA , Reparo de Erro de Pareamento de DNA/fisiologia , Proteínas de Ligação a DNA/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Linhagem Celular Tumoral , DNA/genética , DNA/metabolismo , Reparo de Erro de Pareamento de DNA/efeitos dos fármacos , Reparo de Erro de Pareamento de DNA/genética , Proteínas de Ligação a DNA/genética , Ensaio de Desvio de Mobilidade Eletroforética , Células HeLa , Humanos , Immunoblotting , Camundongos , Dados de Sequência Molecular , Mutação , Células NIH 3T3 , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Serina/genética , Serina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Spodoptera , Estaurosporina/análogos & derivados , Estaurosporina/farmacologia
5.
Virol J ; 7: 359, 2010 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-21129204

RESUMO

The bacteriophage T4 encodes 10 proteins, known collectively as the replisome, that are responsible for the replication of the phage genome. The replisomal proteins can be subdivided into three activities; the replicase, responsible for duplicating DNA, the primosomal proteins, responsible for unwinding and Okazaki fragment initiation, and the Okazaki repair proteins. The replicase includes the gp43 DNA polymerase, the gp45 processivity clamp, the gp44/62 clamp loader complex, and the gp32 single-stranded DNA binding protein. The primosomal proteins include the gp41 hexameric helicase, the gp61 primase, and the gp59 helicase loading protein. The RNaseH, a 5' to 3' exonuclease and T4 DNA ligase comprise the activities necessary for Okazaki repair. The T4 provides a model system for DNA replication. As a consequence, significant effort has been put forth to solve the crystallographic structures of these replisomal proteins. In this review, we discuss the structures that are available and provide comparison to related proteins when the T4 structures are unavailable. Three of the ten full-length T4 replisomal proteins have been determined; the gp59 helicase loading protein, the RNase H, and the gp45 processivity clamp. The core of T4 gp32 and two proteins from the T4 related phage RB69, the gp43 polymerase and the gp45 clamp are also solved. The T4 gp44/62 clamp loader has not been crystallized but a comparison to the E. coli gamma complex is provided. The structures of T4 gp41 helicase, gp61 primase, and T4 DNA ligase are unknown, structures from bacteriophage T7 proteins are discussed instead. To better understand the functionality of T4 DNA replication, in depth structural analysis will require complexes between proteins and DNA substrates. A DNA primer template bound by gp43 polymerase, a fork DNA substrate bound by RNase H, gp43 polymerase bound to gp32 protein, and RNase H bound to gp32 have been crystallographically determined. The preparation and crystallization of complexes is a significant challenge. We discuss alternate approaches, such as small angle X-ray and neutron scattering to generate molecular envelopes for modeling macromolecular assemblies.


Assuntos
Bacteriófago T4/química , Bacteriófago T4/fisiologia , Replicação do DNA , Proteínas Virais/química , Proteínas Virais/metabolismo , Bacteriófago T7/química , Cristalografia por Raios X , Substâncias Macromoleculares/química , Modelos Biológicos , Modelos Moleculares , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Espalhamento a Baixo Ângulo
6.
DNA Repair (Amst) ; 7(6): 951-69, 2008 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-18468964

RESUMO

Treatment with low concentrations of monofunctional alkylating agents induces a G2 arrest only after the second round of DNA synthesis in mammalian cells and requires a proficient mismatch repair (MMR) pathway. Here, we have investigated rapid alkylation-induced recruitment of DNA repair proteins to chromosomal DNA within synchronized populations of MMR proficient cells (HeLa MR) after N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) treatment. Within the first hour, the concentrations of MutS alpha and PCNA increase well beyond their constitutive chromosomally bound levels and MutL alpha is newly recruited to the chromatin-bound MutS alpha. Remarkably, immunoprecipitation experiments demonstrate rapid association of these proteins on the alkylation-damaged chromatin, even when DNA replication is completely blocked. The extent of association of PCNA and MMR proteins on the chromatin is dependent upon the concentration of MNNG and on the specific type of replication block. A subpopulation of the MutS alpha-associated PCNA also becomes monoubiquitinated, a known requirement for PCNA to interact with translesion synthesis (TLS) polymerases. In addition, chromatin-bound SMC1 and NBS1 proteins, associated with DNA double-strand-breaks (DSBs), become phosphorylated within 1-2h of exposure to MNNG. However, these activated proteins are not co-localized on the chromatin with MutS alpha in response to MNNG exposure. PCNA, MutS alpha/MutL alpha and activated SMC1/NBS1 remain chromatin-bound for at least 6-8h after alkylation damage. Thus, cells that are exposed to low levels of alkylation treatment undergo rapid recruitment to and/or activation of key proteins already on the chromatin without the requirement for DNA replication, apparently via different DNA-damage signaling pathways.


Assuntos
Pareamento Incorreto de Bases , Cromatina/metabolismo , Reparo do DNA , Metilnitronitrosoguanidina/farmacologia , Western Blotting , Ciclo Celular , Imunoprecipitação da Cromatina , Dano ao DNA , Imunofluorescência , Células HeLa , Humanos
7.
Mutat Res ; 662(1-2): 59-66, 2009 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-19138690

RESUMO

DNA mismatch repair (MMR) within human cells is hypothesized to occur primarily at the replication fork. However, experimental models measuring MMR activity at specific phases of the cell cycle and during genomic DNA synthesis are lacking. We have investigated MMR activity within the nuclear environment of HeLa cells after enriching for G1, S and G2/M phase of the cell cycle by centrifugal elutriation. This approach preserves physiologically normal MMR activity in cell populations subdivided into different phases of the cell cycle. Here we have shown that nuclear protein concentration of hMutSalpha and hMutLalpha increases as cells progress into S phase during routine cell culture. MMR activity, as measured by both in vitro and in vivo approaches, increases during S phase to the highest extent within normally growing cells. Both fidelity and activity of MMR are highest on actively replicating templates within intact cells during S phase. The MMR pathway however, is also active at lower levels at other phases of the cell cycle, and on nonreplicating templates.


Assuntos
Reparo de Erro de Pareamento de DNA , Replicação do DNA , DNA/biossíntese , Núcleo Celular/metabolismo , Sobrevivência Celular , Citometria de Fluxo , Fase G1 , Células HeLa , Humanos , Proteínas Nucleares/metabolismo , Plasmídeos/genética
9.
BMC Mol Biol ; 6: 6, 2005 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-15766387

RESUMO

BACKGROUND: The current investigation was undertaken to determine key steps differentiating G:T and G:A repair at the H-ras oncogenic hot spot within the nuclear environment because of the large difference in repair efficiency of these two mismatches. RESULTS: Electrophoretic mobility shift (gel shift) experiments demonstrate that DNA containing mismatched bases are recognized and bound equally efficiently by hMutSalpha in both MMR proficient and MMR deficient (hMLH1-/-) nuclear extracts. Competition experiments demonstrate that while hMutSalpha predictably binds the G:T mismatch to a much greater extent than G:A, hMutSalpha demonstrates a surprisingly equal ratio of competitive inhibition for both G:T and G:A mismatch binding reactions at the H-ras hot spot of mutation. Further, mismatch repair assays reveal almost 2-fold higher efficiency of overall G:A repair (5'-nick directed correct MMR to G:C and incorrect repair to T:A), as compared to G:T overall repair. Conversely, correct MMR of G:T --> G:C is significantly higher (96%) than that of G:A --> G:C (60%). CONCLUSION: Combined, these results suggest that initiation of correct MMR requires the contribution of two separate steps; initial recognition by hMutSalpha followed by subsequent binding. The 'avidity' of the binding step determines the extent of MMR pathway activation, or the activation of a different cellular pathway. Thus, initial recognition by hMutSalpha in combination with subsequent decreased binding to the G:A mismatch (as compared to G:T) may contribute to the observed increased frequency of incorrect repair of G:A, resulting in the predominant GGC --> GTC (Gly --> Val) ras-activating mutation found in a high percentage of human tumors.


Assuntos
Pareamento Incorreto de Bases/genética , Códon/genética , Reparo do DNA , Proteínas/genética , Proteínas ras/genética , Linhagem Celular Tumoral , DNA/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética/métodos , Células HCT116 , Humanos , Modelos Biológicos , Plasmídeos/genética , Mutação Puntual/genética , Proto-Oncogenes/genética
10.
J Clin Virol ; 28(1): 27-37, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12927748

RESUMO

BACKGROUND: We report an interlaboratory comparison of methods for the determination of hepatitis C virus (HCV) serum load and genotype between a recently, established molecular laboratory at the Alaska Native Medical Center (ANMC) and two independent laboratories using different assays. At ANMC, a Real-time quantitative RT-PCR amplification methodology (QPCR) has been developed in which HCV viral loads are determined by interpolation of QPCR results to those of standards calibrated to the World Health Organization (WHO) First International Standard for HCV. HCV genotype is subsequently determined by direct sequencing of the DNA fragment generated from the QPCR assay. OBJECTIVES AND STUDY DESIGN: The above methods were statistically compared to results obtained for the same patient sera by two independent laboratories using different commercially available viral load assays; Quantiplex HCV RNA (Bayer Diagnostics) and Amplicor HCV Monitor (v 2.0) (Roche Molecular Systems), as well as two different genotyping assays; restriction fragment length polymorphism (RFLP) and INNO-LiPA HCV II (Innogenetics). RESULTS: ANMC's Real-time QPCR HCV viral load results compared moderately well with those obtained by the Quantiplex HCV RNA method (R2=0.3813), and compared quite well with recent lot numbers of Amplicor HCV Monitor in which viral loads are derived in IU/ml (R2=0.6408), but compared poorly with earlier lot numbers of Amplicor HCV Monitor in which viral loads were derived in copies/ml (R2=0.0913). The ANMC direct sequencing method for genotype determination compared moderately to very well with both the RFLP (84-86%) and INNO-LiPA (85-97.5%) methods. CONCLUSIONS: These viral load comparisons highlight the discrepancies that may occur when patient HCV viral loads are monitored using different types of assays. Comparison of HCV genotype by different methods is more reliable statistically and important clinically for predicting probability of response to antiviral therapy. However, viral loads are important for monitoring response once therapy has begun.


Assuntos
Hepacivirus/fisiologia , Hepatite C/virologia , RNA Viral/sangue , Carga Viral , Estudos de Avaliação como Assunto , Genótipo , Hepacivirus/isolamento & purificação , Hepatite C/sangue , Humanos , Técnicas de Amplificação de Ácido Nucleico , Reação em Cadeia da Polimerase , RNA Viral/análise , Kit de Reagentes para Diagnóstico , Sensibilidade e Especificidade
11.
PLoS One ; 8(9): e74071, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24019948

RESUMO

The major dilemma of cancer chemotherapy has always been a double-edged sword, producing resistance in tumor cells and life-threatening destruction of nontumorigenic tissue. Glioblastoma is the most common form of primary brain tumor, with median survival at 14 months after surgery, radiation and temozolomide (monofunctional alkylator) therapy. Treatment failure is most often due to temozolomide-resistant tumor growth. The underlying basis for development of tumor cell resistance to temozolomide instead of death is not understood. Our current results demonstrate that both cervical carcinoma (HeLa MR) and glioblastoma (U251) tumor cells exposed to an equivalent chemotherapeutic concentration of a monofunctional alkylator undergo multiple cell cycles, maintenance of metabolic activity, and a prolonged time to death that involves accumulation of Apoptosis Inducing Factor (AIF) within the nucleus. A minority of the tumor cell population undergoes senescence, with minimal caspase cleavage. Surviving tumor cells are comprised of a very small subpopulation of individual cells that eventually resume proliferation, out of which resistant cells emerge. In contrast, normal human cells (MCF12A) exposed to a monofunctional alkylator undergo an immediate decrease in metabolic activity and subsequent senescence. A minority of the normal cell population undergoes cell death by the caspase cleavage pathway. All cytotoxic events occur within the first cell cycle in nontumorigenic cells. In summation, we have demonstrated that two different highly malignant tumor cell lines slowly undergo very altered cellular and temporal responses to chemotherapeutic monofunctional alkylation, as compared to rapid responses of normal cells. In the clinic, this produces resistance and growth of tumor cells, cytotoxicity of normal cells, and death of the patient.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Linhagem da Célula , Dacarbazina/análogos & derivados , Neoplasias/patologia , Fator de Indução de Apoptose/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Tumoral , Dacarbazina/farmacologia , Ensaio de Desvio de Mobilidade Eletroforética , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Metilnitronitrosoguanidina/farmacologia , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Temozolomida
12.
Gene Regul Syst Bio ; 7: 125-38, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23888109

RESUMO

Inter-individual variation in CCAAT/enhancer binding protein gamma (CEBPG) transcript expression in normal human bronchial epithelial cells (NBEC) is associated with predisposition to lung cancer. We hypothesize that this inter-individual variation is in part explained by cis-acting genetic variation in CEBPG. To test this hypothesis we measured transcript expression derived from each parental copy of CEBPG (ie, allele-specific expression; ASE). There was a significant 2.9-fold higher cell cycle-specific variation in ASE of CEBPG rs2772 A compared to C allele (P < 0.001). In 20% of NBEC samples, CEBPG rs2772 A allele was expressed on average 2.10 fold greater than rs2772 C allele. These data support the hypothesis that genetic variation in linkage disequilibrium with rs2772 influences regulation of CEBPG transcript expression through a trans-effect downstream of RNA polymerase II transcription and confirm that cis-acting genetic variation contributes to inter-individual variation in CEBPG transcript expression in NBEC, which is associated with variation in lung cancer risk.

14.
Cancer Res ; 69(15): 6307-14, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-19638578

RESUMO

Alkylation chemotherapy has been a long-standing treatment protocol for human neoplasia. N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) is a direct-acting monofunctional alkylator. Temozolomide is a clinical chemotherapeutic equivalent requiring metabolic breakdown to the alkylating agent. Both chemicals have similar mechanistic efficacy against DNA mismatch repair-proficient tumor cells that lack expression of methylguanine methyltransferase. Clinically relevant concentrations of both agents affect replicating cells only after the first cell cycle. This phenomenon has been attributed to replication fork arrest at unrepaired O(6)-methyldeoxyguanine lesions mispaired with thymine during the first replication cycle. Here, we show, by several different approaches, that MNNG-treated tumor cells do not arrest within the second cell cycle. Instead, the population slowly traverses through mitosis without cytokinesis into a third cell cycle. The peak of both ssDNA and dsDNA breaks occurs at the height of the long mitotic phase. The majority of the population emerges from mitosis as multinucleated cells that subsequently undergo cell death. However, a very small proportion of cells, <1:45,000, survive to form new colonies. Taken together, these results indicate that multinucleation within the third cell cycle, rather than replication fork arrest within the second cell cycle, is the primary trigger for cell death. Importantly, multinucleation and cell death are consistently avoided by a small percentage of the population that continues to divide. This information should prove clinically relevant for the future design of enhanced cancer chemotherapeutics.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Ciclo Celular/efeitos dos fármacos , Metilnitronitrosoguanidina/farmacologia , Proteína Quinase CDC2 , Ciclo Celular/genética , Ciclina B/metabolismo , Quinases Ciclina-Dependentes , Reparo de Erro de Pareamento de DNA , Metilases de Modificação do DNA/deficiência , Enzimas Reparadoras do DNA/deficiência , Relação Dose-Resposta a Droga , Células HeLa , Histonas/metabolismo , Humanos , Fosforilação , Proteínas Supressoras de Tumor/deficiência
15.
Exp Cell Res ; 313(2): 292-304, 2007 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-17157834

RESUMO

The DNA mismatch repair (MMR) pathway contributes to the fidelity of DNA synthesis and recombination by correcting mispaired nucleotides and insertion/deletion loops (IDLs). We have investigated whether MMR protein expression, activity, and subcellular location are altered during discrete phases of the cell cycle in mammalian cells. Two distinct methods have been used to demonstrate that although physiological MMR protein expression, mismatch binding, and nick-directed MMR activity within the nucleus are at highest levels during S phase, MMR is active throughout the cell cycle. Despite equal MMR nuclear protein concentrations in S and G(2) phases, mismatch binding and repair activities within G(2) are significantly lower, indicating a post-translational decrease in MMR activity specific to G(2). We further demonstrate that typical co-localization of MutSalpha to late S phase replication foci can be disrupted by 2 microM N-methyl-N'-nitro-N-nitrosoguanidine (MNNG). This concentration of MNNG does not decrease ongoing DNA synthesis nor induce cell cycle arrest until the second cell cycle, with long-term colony survival decreased by only 24%. These results suggest that low level alkylation damage can selectively disrupt MMR proofreading activity during DNA synthesis and potentially increase mutation frequency within surviving cells.


Assuntos
Ciclo Celular/genética , Reparo de Erro de Pareamento de DNA , Proteínas de Ligação a DNA/metabolismo , Animais , Núcleo Celular/química , Células Cultivadas , Replicação do DNA , Proteínas de Ligação a DNA/análise , Humanos , Antígeno Nuclear de Célula em Proliferação/análise , Antígeno Nuclear de Célula em Proliferação/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa