Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 353
Filtrar
1.
Redox Biol ; 52: 102316, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35489241

RESUMO

Mycobacterium tuberculosis (Mtb) senses and responds to host-derived gasotransmitters NO and CO via heme-containing sensor kinases DosS and DosT and the response regulator DosR. Hydrogen sulfide (H2S) is an important signaling molecule in mammals, but its role in Mtb physiology is unclear. We have previously shown that exogenous H2S can modulate expression of genes in the Dos dormancy regulon via an unknown mechanism(s). Here, we test the hypothesis that Mtb senses and responds to H2S via the DosS/T/R system. Using UV-Vis and EPR spectroscopy, we show that H2S binds directly to the ferric (Fe3+) heme of DosS (KDapp = 5.30 µM) but not the ferrous (Fe2+) form. No interaction with DosT(Fe2+-O2) was detected. We found that the binding of sulfide can slowly reduce the DosS heme iron to the ferrous form. Steered Molecular Dynamics simulations show that H2S, and not the charged HS- species, can enter the DosS heme pocket. We also show that H2S increases DosS autokinase activity and subsequent phosphorylation of DosR, and H2S-mediated increases in Dos regulon gene expression is lost in Mtb lacking DosS. Finally, we demonstrate that physiological levels of H2S in macrophages can induce DosR regulon genes via DosS. Overall, these data reveal a novel mechanism whereby Mtb senses and responds to a third host gasotransmitter, H2S, via DosS(Fe3+). These findings highlight the remarkable plasticity of DosS and establish a new paradigm for how bacteria can sense multiple gasotransmitters through a single heme sensor kinase.


Assuntos
Gasotransmissores , Mycobacterium tuberculosis , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Ácido Dioctil Sulfossuccínico/metabolismo , Gasotransmissores/metabolismo , Regulação Bacteriana da Expressão Gênica , Heme/metabolismo , Ferro/metabolismo , Mamíferos/genética , Mamíferos/metabolismo , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/metabolismo , Protamina Quinase/química , Protamina Quinase/genética , Protamina Quinase/metabolismo , Regulon
2.
Commun Biol ; 2: 349, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31552302

RESUMO

Dormancy is a key characteristic of the intracellular life-cycle of Mtb. The importance of sensor kinase DosS in mycobacteria are attributed in part to our current findings that DosS is required for both persistence and full virulence of Mtb. Here we show that DosS is also required for optimal replication in macrophages and involved in the suppression of TNF-α and autophagy pathways. Silencing of these pathways during the infection process restored full virulence in MtbΔdosS mutant. Notably, a mutant of the response regulator DosR did not exhibit the attenuation in macrophages, suggesting that DosS can function independently of DosR. We identified four DosS targets in Mtb genome; Rv0440, Rv2859c, Rv0994, and Rv0260c. These genes encode functions related to hypoxia adaptation, which are not directly controlled by DosR, e.g., protein recycling and chaperoning, biosynthesis of molybdenum cofactor and nitrogen metabolism. Our results strongly suggest a DosR-independent role for DosS in Mtb.


Assuntos
Autofagossomos/metabolismo , Autofagossomos/microbiologia , Proteínas de Bactérias/metabolismo , Mycobacterium tuberculosis/fisiologia , Protamina Quinase/metabolismo , Proteínas Quinases/metabolismo , Tuberculose/metabolismo , Tuberculose/microbiologia , Autofagossomos/imunologia , Autofagia , Proteínas de Bactérias/genética , Proteínas de Ligação a DNA , Perfilação da Expressão Gênica , Inativação Gênica , Interações Hospedeiro-Patógeno/imunologia , Humanos , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Mutação , Mycobacterium tuberculosis/enzimologia , Fagócitos/imunologia , Fagócitos/metabolismo , Fagócitos/microbiologia , Fosforilação , Protamina Quinase/genética , Proteínas Quinases/genética , Tuberculose/imunologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Virulência
3.
Mol Cell ; 64(1): 176-188, 2016 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-27716482

RESUMO

How deregulation of chromatin modifiers causes malignancies is of general interest. Here, we show that histone H2A T120 is phosphorylated in human cancer cell lines and demonstrate that this phosphorylation is catalyzed by hVRK1. Cyclin D1 was one of ten genes downregulated upon VRK1 knockdown in two different cell lines and showed loss of H2A T120 phosphorylation and increased H2A K119 ubiquitylation of its promoter region, resulting in impaired cell growth. In vitro, H2A T120 phosphorylation and H2A K119 ubiquitylation are mutually inhibitory, suggesting that histone phosphorylation indirectly activates chromatin. Furthermore, expression of a phosphomimetic H2A T120D increased H3 K4 methylation. Finally, both VRK1 and the H2A T120D mutant histone transformed NIH/3T3 cells. These results suggest that histone H2A T120 phosphorylation by hVRK1 causes inappropriate gene expression, including upregulated cyclin D1, which promotes oncogenic transformation.


Assuntos
Transformação Celular Neoplásica/genética , Ciclina D1/genética , Regulação Neoplásica da Expressão Gênica , Histonas/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Processamento de Proteína Pós-Traducional , Proteínas Serina-Treonina Quinases/genética , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Cromatina/química , Cromatina/metabolismo , Ciclina D1/metabolismo , Proteínas de Drosophila , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Células HeLa , Histonas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Metilação , Camundongos , Oligopeptídeos/genética , Oligopeptídeos/metabolismo , Fosforilação , Protamina Quinase/genética , Protamina Quinase/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Treonina/metabolismo , Ubiquitinação
4.
Expert Rev Mol Diagn ; 16(3): 297-306, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26750583

RESUMO

Histones are the chief components of chromatin. When being catalyzed by a series of histone modifying enzymes, histones may undergo various post-translational modifications such as acetylation, methylation, phosphorylation, ubiquitylation and SUMOylation. The dysregulation of histone modifying enzymes will alter the histone post-modification patterns and cause diverse diseases including cancers. Consequently, the histone modifying enzymes have emerged as the promising biomarkers for disease diagnosis and prognosis. In this review, we summarize the recent researches about the histone modifying enzymes as the disease biomarkers, and highlight the development of methods for histone modifying enzyme assays.


Assuntos
Biomarcadores Tumorais/metabolismo , Histona Acetiltransferases/metabolismo , Histona Desacetilases/metabolismo , Histona Desmetilases/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Neoplasias/diagnóstico , Protamina Quinase/metabolismo , Animais , Ensaios Enzimáticos/métodos , Histona Metiltransferases , Humanos , Técnicas de Diagnóstico Molecular/métodos
5.
Am J Respir Cell Mol Biol ; 52(6): 708-16, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25322074

RESUMO

Mycobacterium tuberculosis (Mtb) must counter hypoxia within granulomas to persist. DosR, in concert with sensor kinases DosS and DosT, regulates the response to hypoxia. Yet Mtb lacking functional DosR colonize the lungs of C57Bl/6 mice, presumably owing to the lack of organized lesions with sufficient hypoxia in that model. We compared the phenotype of the Δ-dosR, Δ-dosS, and Δ-dosT mutants to Mtb using C3HeB/FeJ mice, an alternate mouse model where lesions develop hypoxia. C3HeB/FeJ mice were infected via aerosol. The progression of infection was analyzed by tissue bacterial burden and histopathology. A measure of the comparative global immune responses was also analyzed. Although Δ-dosR and Δ-dosT grew comparably to wild-type Mtb, Δ-dosS exhibited a significant defect in bacterial burden and pathology in vivo, accompanied by ablated proinflammatory response. Δ-dosS retained the ability to induce DosR. The Δ-dosS mutant was also attenuated in murine macrophages ex vivo, with evidence of reduced expression of the proinflammatory signature. Our results show that DosS, but not DosR and DosT, is required by Mtb to survive in C3HeB/FeJ mice. The attenuation of Δ-dosS is not due to its inability to induce the DosR regulon, nor is it a result of the accumulation of hypoxia. That the in vivo growth restriction of Δ-dosS could be mimicked ex vivo suggested sensitivity to macrophage oxidative burst. Anoxic caseous centers within tuberculosis lesions eventually progress to cavities. Our results provide greater insight into the molecular mechanisms of Mtb persistence within host lungs.


Assuntos
Proteínas de Bactérias/genética , Granuloma do Sistema Respiratório/microbiologia , Mycobacterium tuberculosis/patogenicidade , Protamina Quinase/genética , Tuberculose Pulmonar/microbiologia , Animais , Proteínas de Bactérias/metabolismo , Hipóxia Celular , Células Cultivadas , Regulação Bacteriana da Expressão Gênica , Macrófagos/microbiologia , Masculino , Camundongos Endogâmicos C3H , Viabilidade Microbiana , Mycobacterium tuberculosis/genética , Protamina Quinase/metabolismo , Regulon , Virulência
6.
PLoS One ; 9(8): e103993, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25093845

RESUMO

OBJECTIVE: Cigarette smoking is a major risk factor for atherosclerotic cardiovascular disease, which is responsible for a significant proportion of smoking-related deaths. However, the precise mechanism whereby smoking induces this pathology has not been fully delineated. Based on observation of DNA double-strand breaks (DSBs), the most harmful type of DNA damage, in atherosclerotic lesions, we hypothesized that there is a direct association between smoking and DSBs. The goal of this study was to investigate whether smoking induces DSBs and smoking cessation reverses DSBs in vivo through examination of peripheral mononuclear cells (MNCs). APPROACH AND RESULTS: Immunoreactivity of oxidative modification of DNA and DSBs were increased in human atherosclerotic lesions but not in the adjacent normal area. DSBs in human MNCs isolated from the blood of volunteers can be detected as cytologically visible "foci" using an antibody against the phosphorylated form of the histone H2AX (γ-H2AX). Young healthy active smokers (n = 15) showed increased γ-H2AX foci number when compared with non-smokers (n = 12) (foci number/cell: median, 0.37/cell; interquartile range [IQR], 0.31-0.58 vs. 4.36/cell; IQR, 3.09-7.39, p<0.0001). Smoking cessation for 1 month reduced the γ-H2AX foci number (median, 4.44/cell; IQR, 4.36-5.24 to 0.28/cell; IQR, 0.12-0.53, p<0.05). A positive correlation was noted between γ-H2AX foci number and exhaled carbon monoxide levels (r = 0.75, p<0.01). CONCLUSIONS: Smoking induces DSBs in human MNCs in vivo, and importantly, smoking cessation for 1 month resulted in a decrease in DSBs to a level comparable to that seen in non-smokers. These data reinforce the notion that the cigarette smoking induces DSBs and highlight the importance of smoking cessation.


Assuntos
Quebras de DNA de Cadeia Dupla , Leucócitos Mononucleares/metabolismo , Abandono do Hábito de Fumar , Adulto , Células Cultivadas , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Histonas/metabolismo , Humanos , Peróxido de Hidrogênio/farmacologia , Leucócitos Mononucleares/efeitos dos fármacos , Masculino , Fosforilação/efeitos dos fármacos , Placa Aterosclerótica/genética , Placa Aterosclerótica/metabolismo , Protamina Quinase/metabolismo , Fumaça/efeitos adversos , Nicotiana/efeitos adversos , Adulto Jovem
7.
Exp Cell Res ; 324(1): 75-83, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24704462

RESUMO

Hyperglycemia is a critical risk factor for development and progression of breast cancer. We have recently reported that high glucose induces phosphorylation of histone H3 at Ser 10 as well as de-phosphorylation of GSK-3ß at Ser 9 in MDA-MB-231 cells. Here, we elucidate the mechanism underlying hyperglycemia-induced proliferation in MDA-MB-231 breast cancer cells. We provide evidence that hyperglycemia led to increased DNA methylation and DNMT1 expression in MDA-MB-231 cells. High glucose condition led to significant increase in the expression of PCNA, cyclin D1 and decrease in the expression of PTPN 12, p21 and PTEN. It also induced hypermethylation of DNA at the promoter region of PTPN 12, whereas hypomethylation at Vimentin and Snail. Silencing of GSK-3ß by siRNA prevented histone H3 phosphorylation and reduced DNMT1 expression. We show that chromatin obtained after immunoprecipitation with phospho-histone H3 was hypermethylated under high glucose condition, which indicates a cross-talk between DNA methylation and histone H3 phosphorylation. ChIP-qPCR analysis revealed up-regulation of DNMT1 and metastatic genes viz. Vimentin, Snail and MMP-7 by phospho-histone H3, which were down-regulated upon GSK-3ß silencing. To the best of our knowledge, this is the first report which shows that interplay between GSK-3ß activation, histone H3 phosphorylation and DNA methylation directs proliferation of breast cancer cells.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Proliferação de Células/efeitos dos fármacos , Epigênese Genética/fisiologia , Quinase 3 da Glicogênio Sintase/fisiologia , Hiperglicemia/metabolismo , Linhagem Celular Tumoral , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/genética , Metilação de DNA/efeitos dos fármacos , Epigênese Genética/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes Supressores de Tumor/efeitos dos fármacos , Glucose/farmacologia , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Histonas/genética , Histonas/metabolismo , Humanos , Hiperglicemia/genética , Protamina Quinase/metabolismo
8.
J Biol Chem ; 288(18): 12437-47, 2013 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-23486471

RESUMO

The sensor histidine kinases of Mycobacterium tuberculosis, DosS and DosT, are responsible for sensing hypoxic conditions and consist of sensor and kinase cores responsible for accepting signals and phosphorylation activity, respectively. The kinase core contains a dimerization and histidine phosphate-accepting (DHp) domain and an ATP binding domain (ABD). The 13 histidine kinase genes of M. tuberculosis can be grouped based on the presence or absence of the ATP lid motif and F box (elements known to play roles in ATP binding) in their ABDs; DosS and DosT have ABDs lacking both these elements, and the crystal structures of their ABDs indicated that they were unsuitable for ATP binding, as a short loop covers the putative ATP binding site. Although the ABD alone cannot bind ATP, the kinase core is functional in autophosphorylation. Appropriate spatial arrangement of the ABD and DHp domain within the kinase core is required for both autophosphorylation and ATP binding. An ionic interaction between Arg(440) in the DHp domain and Glu(537) in the short loop of the ABD is available and may open the ATP binding site, by repositioning the short loop away from the site. Mutations at Arg(440) and Glu(537) reduce autophosphorylation activity. Unlike other histidine kinases containing an ATP lid, which protects bound ATP, DosS is unable to accept ATP until the ABD is properly positioned relative to the histidine; this may prevent unexpected ATP reactions. ATP binding can, therefore, function as a control mechanism for histidine kinase activity.


Assuntos
Trifosfato de Adenosina/química , Proteínas de Bactérias/química , Mycobacterium tuberculosis/enzimologia , Protamina Quinase/química , Trifosfato de Adenosina/metabolismo , Motivos de Aminoácidos , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Ativação Enzimática/fisiologia , Fosforilação/fisiologia , Protamina Quinase/metabolismo
9.
FEBS Lett ; 587(1): 17-22, 2013 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-23178685

RESUMO

ERα, a critical transcriptional factor for breast cancer proliferation, is regulated by a complex binding repertoire that includes coactivators and corepressors. Here, we identified a novel class of ERα coregulator called CAC1. The CoRNR box of CAC1 was required for the binding to and inactivation of ERα. CAC1 also associated with histone demethylase LSD1 and suppressed LSD1-enhanced ERα activity. CAC1 impaired recruitment of ERα and LSD1 to the ERα-responsive promoter, leading to greater H3K9me3 accumulation. This effect was reversed by CAC1 depletion. Finally, CAC1 increased paclitaxel-induced cell death in ERα-positive MCF7 cells, which are paclitaxel-resistant. Overall, our study provides the first evidence that CAC1, associated with LSD1, functions as an ERα corepressor, implicating a potential antitumor target in ERα-positive breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Proteínas Correpressoras/metabolismo , Proteínas Culina/metabolismo , Receptor alfa de Estrogênio/metabolismo , Histona Desmetilases/metabolismo , Proteínas de Neoplasias/metabolismo , Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/enzimologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proteínas Correpressoras/antagonistas & inibidores , Proteínas Correpressoras/genética , Proteínas Culina/antagonistas & inibidores , Proteínas Culina/genética , Resistencia a Medicamentos Antineoplásicos , Receptor alfa de Estrogênio/genética , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes Reporter/efeitos dos fármacos , Histona Desmetilases/genética , Histonas/metabolismo , Humanos , Proteínas Mutantes/antagonistas & inibidores , Proteínas Mutantes/metabolismo , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Paclitaxel/farmacologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Protamina Quinase/metabolismo , Interferência de RNA , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/metabolismo , Fator Trefoil-1 , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
11.
J Bacteriol ; 193(19): 5105-18, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21821774

RESUMO

Mycobacterium tuberculosis, the etiological agent of tuberculosis, remains a significant cause of morbidity and mortality throughout the world despite a vaccine and cost-effective antibiotics. The success of this organism can be attributed, in part, to its ability to adapt to potentially harmful stress within the host and establish, maintain, and reactivate from long-term persistent infection within granulomatous structures. The DosRS-DosT/DevRS-Rv2027c, and MprAB two-component signal transduction systems have previously been implicated in aspects of persistent infection by M. tuberculosis and are known to be responsive to conditions likely to be found within the granuloma. Here, we describe initial characterization of a locus (Rv0081-Rv0088) encoding components of a predicted formate hydrogenylase enzyme complex that is directly regulated by DosR/DevR and MprA, and the product of the first gene in this operon, Rv0081. In particular, we demonstrate that Rv0081 negatively regulates its own expression and that of downstream genes by binding an inverted repeat element in its upstream region. In contrast, DosR/DevR and MprA positively regulate Rv0081 expression by binding to recognition sequences that either partially or completely overlap that recognized by Rv0081, respectively. Expression of Rv0081 initiates from two promoter elements; one promoter located downstream of the DosR/DevR binding site but overlapping the sequence recognized by both Rv0081 and MprA and another promoter downstream of the DosR/DevR, Rv0081, and MprA binding sites. Interestingly, Rv0081 represses Rv0081 and downstream determinants following activation of DosRS-DosT/DevRS-Rv2027c by nitric oxide, suggesting that expression of this locus is complex and subject to multiple levels of regulation. Based on this and other published information, a model is proposed detailing Rv0081-Rv0088 expression by these transcription factors within particular growth environments.


Assuntos
Proteínas de Bactérias/metabolismo , Formiato Desidrogenases/metabolismo , Complexos Multienzimáticos/metabolismo , Mycobacterium tuberculosis/metabolismo , Protamina Quinase/metabolismo , Proteínas Quinases/metabolismo , Proteínas de Bactérias/genética , Proteínas de Ligação a DNA , Ensaio de Desvio de Mobilidade Eletroforética , Formiato Desidrogenases/genética , Regulação Bacteriana da Expressão Gênica/genética , Regulação Bacteriana da Expressão Gênica/fisiologia , Complexos Multienzimáticos/genética , Mutação , Mycobacterium tuberculosis/genética , Protamina Quinase/genética , Proteínas Quinases/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
BMC Cancer ; 11: 58, 2011 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-21299907

RESUMO

BACKGROUND: Histone modifications in tumorigenesis are increasingly recognized as important epigenetic factors leading to cancer. Increased phosphorylation levels of histone H3 as a result of aurora B and pMSK1 overexpression were observed in various tumors. We selected aurora B and MSK1 as representatives for testing various compounds and drugs, and found that squamocin, a bis-tetrahydrofuran annonaceous acetogenin, exerted a potent effect on histone H3 phosphorylation. METHODS: GBM8401, Huh-7, and SW620 cells were incubated with 15, 30, and 60 µM squamocin for 24 h. The expressions of mRNA and proteins were analyzed by qRT-PCR and Western blotting, respectively. The cell viability was determined by an MTT assay. Cell cycle distribution and apoptotic cells were analyzed by flow cytometry. RESULTS: Our results showed that squamocin inhibited the proliferation of GBM8401, Huh-7, and SW620 cells, arrested the cell cycle at the G1 phase, and activated both intrinsic and extrinsic pathways to apoptosis. In addition, we demonstrated that squamocin had the ability to modulate the phosphorylation levels of H3S10 (H3S10p) and H3S28 (H3S28p) in association with the downregulation of aurora B and pMSK1 expressions. CONCLUSIONS: This study is the first to show that squamocin affects epigenetic alterations by modulating histone H3 phosphorylation at S10 and S28, providing a novel view of the antitumor mechanism of squamocin.


Assuntos
Apoptose/efeitos dos fármacos , Furanos/farmacologia , Fase G1/efeitos dos fármacos , Histonas/metabolismo , Lactonas/farmacologia , Neoplasias/patologia , Protamina Quinase/metabolismo , Antineoplásicos Fitogênicos/farmacologia , Apoptose/fisiologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Citotoxinas/farmacologia , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Humanos , Modelos Biológicos , Neoplasias/metabolismo , Fosforilação/efeitos dos fármacos
13.
Int J Radiat Biol ; 86(4): 334-43, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20353343

RESUMO

PURPOSE: To investigate the effect of 2.45 GHz microwave radiation on rat brain of male wistar strain. MATERIAL AND METHODS: Male rats of wistar strain (35 days old with 130 +/- 10 g body weight) were selected for this study. Animals were divided into two groups: Sham exposed and experimental. Animals were exposed for 2 h a day for 35 days to 2.45 GHz frequency at 0.34 mW/cm(2) power density. The whole body specific absorption rate (SAR) was estimated to be 0.11 W/Kg. Exposure took place in a ventilated Plexiglas cage and kept in anechoic chamber in a far field configuration from the horn antenna. After the completion of exposure period, rats were sacrificed and the whole brain tissue was dissected and used for study of double strand DNA (Deoxyribonucleic acid) breaks by micro gel electrophoresis and the statistical analysis was carried out using comet assay (IV-2 version software). Thereafter, antioxidant enzymes and histone kinase estimation was also performed. RESULTS: A significant increase was observed in comet head (P < 0.002), tail length (P < 0.0002) and in tail movement (P < 0.0001) in exposed brain cells. An analysis of antioxidant enzymes glutathione peroxidase (P < 0.005), and superoxide dismutase (P < 0.006) showed a decrease while an increase in catalase (P < 0.006) was observed. A significant decrease (P < 0.023) in histone kinase was also recorded in the exposed group as compared to the control (sham-exposed) ones. One-way analysis of variance (ANOVA) method was adopted for statistical analysis. CONCLUSION: The study concludes that the chronic exposure to these radiations may cause significant damage to brain, which may be an indication of possible tumour promotion (Behari and Paulraj 2007).


Assuntos
Antioxidantes/efeitos da radiação , Encéfalo/efeitos da radiação , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Micro-Ondas , Animais , Antioxidantes/metabolismo , Encéfalo/enzimologia , Encéfalo/metabolismo , Encéfalo/patologia , Catalase/metabolismo , Catalase/efeitos da radiação , Ensaio Cometa , Glutationa Peroxidase/metabolismo , Glutationa Peroxidase/efeitos da radiação , Masculino , Protamina Quinase/metabolismo , Protamina Quinase/efeitos da radiação , Ratos , Ratos Wistar , Superóxido Dismutase/metabolismo , Superóxido Dismutase/efeitos da radiação , Fatores de Tempo
14.
J Vet Med Sci ; 72(7): 887-92, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20215718

RESUMO

This study was designed to evaluate the nuclear maturation and maturation promoting factor (MPF) level at different maturation times, and the effect of parthenogenetic activation on nuclear maturation in canine oocytes. Cumulus-oocyte complexes (COCs) were matured in TCM-199 supplemented with 10% fetal bovine serum, hormones, 0.57 mM cysteine, and 10 ng/ml epidermal growth factor for 72 hr at 38.5 degrees C. In Experiment 1, COCs at 0, 24, 48 and 72 hr of culture were assessed for nuclear maturation and MPF levels using histone H1 kinase activity assay. A significantly higher rate of oocytes at 72 hr than 0, 24 and 48 hr of culture developed to metaphase I-anaphase I and metaphase II. Relative abundance of histone H1 kinase activity of oocytes matured for 48 hr increased to ~1.5 x, with a marked increase to approximately 2.5 x for 72 hr, significantly higher than others. In Experiment 2, oocytes matured for 48 hr were parthenogenetically activated with 5 microm ionomycin for 5 min (Group 1) and followed by 10 microg/ml cycloheximide for 3 hr (Group 2), or no treatment (Control). Oocytes were then cultured for 24 hr and assessed for nuclear maturation. A significantly higher rate of oocytes in Group 1 developed to metaphase II than in Group 2 and the control. These results indicated that ionomycin treatment at 48 hr of in vitro maturation had a positive influence on oocyte progression to the metaphase II stage.


Assuntos
Oócitos/fisiologia , Animais , Bovinos , Técnicas de Cultura de Células/métodos , Núcleo Celular/fisiologia , Cicloeximida/farmacologia , Cães , Feminino , Humanos , Ionomicina/farmacologia , Metáfase , Oócitos/citologia , Oócitos/efeitos dos fármacos , Oócitos/enzimologia , Partenogênese/efeitos dos fármacos , Partenogênese/fisiologia , Protamina Quinase/metabolismo , Maturidade Sexual/fisiologia
15.
Oncogene ; 28(35): 3167-76, 2009 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-19561641

RESUMO

Our laboratory has previously described the presence of five tumor-specific low molecular weight isoforms of cyclin E in both tumor cell lines and breast cancer patient biopsies. We have also shown that one of these low forms arises from an alternate start site, whereas the other four appear as two sets of doublets following cleavage through an elastase-like enzyme. However, the origin of both sets of doublets was unknown. Here, we demonstrate that the larger isoform of each doublet is the result of phosphorylation at a key degradation site. Through site-directed mutagenesis of different phosphorylation sites within the cyclin E protein, we discovered that phosphorylation of threonine 395 is responsible for generating the larger isoform of each doublet. Because phosphorylation of threonine 395 has been linked to the proteasome-mediated degradation of full length cyclin E, we examined the stability of T395A phospho-mutants in both non-tumorigenic mammary epithelial cells and tumor cells. The results revealed that the low molecular weight isoforms appear to be stable in both a tumor cell line and a non-tumor forming cell line regardless of the presence of this critical phosphorylation site. The stability of low molecular weight cyclin E may have implications for both tumorigenesis and treatment of tumors expressing them.


Assuntos
Ciclina E/química , Ciclina E/metabolismo , Processamento de Proteína Pós-Traducional , Alanina/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Neoplasias da Mama/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Ciclina E/genética , Quinase 2 Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Humanos , Rim/citologia , Modelos Biológicos , Dados de Sequência Molecular , Peso Molecular , Fosforilação , Protamina Quinase/análise , Protamina Quinase/metabolismo , Ligação Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transfecção , Quinases Ativadas por p21/metabolismo
16.
Biochemistry ; 48(25): 5839-48, 2009 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-19463006

RESUMO

DevS is one of the two sensing kinases responsible for DevR activation and the subsequent entry of Mycobacterium tuberculosis into dormancy. Full-length wild-type DevS forms a stable oxy-ferrous complex. The DevS autoxidation rates are extremely low (half-lives of >24 h) in the presence of cations such as K(+), Na(+), Mg(2+), and Ca(2+). At relatively high concentrations (100 mM), Cu(2+) accelerates autoxidation more than 1500-fold. Contrary to expectations, removal of the key hydrogen bond between the iron-coordinated oxygen and Tyr171 in the Y171F mutant provides a protein of comparable stability to autoxidation and similar oxygen dissociation rate. This correlates with our earlier finding that the Y171F mutant and wild-type kinase activities are similarly regulated by the binding of oxygen: namely, the ferrous five-coordinate complex is active, whereas the oxy-ferrous six-coordinate species is inactive. Our results indicate that DevS is a gas sensor in vivo rather than a redox sensor and that the stability of its ferrous-oxy complex is enhanced by interdomain interactions.


Assuntos
Proteínas de Bactérias/metabolismo , Hemeproteínas/metabolismo , Mycobacterium tuberculosis/química , Mycobacterium tuberculosis/metabolismo , Oxigênio/metabolismo , Protamina Quinase/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Sítios de Ligação/genética , Compostos Ferrosos/química , Compostos Ferrosos/metabolismo , Hemeproteínas/química , Hemeproteínas/genética , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/crescimento & desenvolvimento , Oxirredução , Oxigênio/química , Protamina Quinase/química , Protamina Quinase/genética , Estabilidade Proteica , Estrutura Terciária de Proteína/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
17.
Endocrinology ; 150(5): 2334-41, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19116339

RESUMO

We have shown previously that Ser10 phosphorylation of histone H3 occurs in rat pinealocytes after stimulation with norepinephrine (NE) and that histone modifications such as acetylation appear to play an important role in pineal gene transcription. Here we report the nocturnal phosphorylation of a Ser10 histone H3 kinase, Aurora C, in the rat pineal gland. The time profile of this phosphorylation parallels the increase in the level of phospho-Ser10 histone H3. Studies with cultured pinealocytes indicate that Aurora C phosphorylation is induced by NE and this induction can be blocked by cotreatment with propranolol or KT5720, a protein kinase A inhibitor. Moreover, only treatment with dibutyryl cAMP, but not other kinase activators, mimics the effect of NE on Aurora C phosphorylation. These results indicate that Aurora C is phosphorylated primarily by a beta-adrenergic/protein kinase A-mediated mechanism. Treatment with an Aurora C inhibitor reduces the NE-induced histone H3 phosphorylation and suppresses the NE-stimulated induction of arylalkylamine N-acetyltransferase (AA-NAT), the rhythm-controlling enzyme of melatonin synthesis, and melatonin production. The effects of Aurora C inhibitors on adrenergic-induced genes in rat pinealocytes are gene specific: inhibitory for Aa-nat and inducible cAMP repressor but stimulatory for c-fos. Together our results support a role for the NE-stimulated phosphorylation of Aurora C and the subsequent remodeling of chromatin in NE-stimulated Aa-nat transcription. This phenomenon suggests that activation of this mitotic kinase can be induced by extracellular signals to participate in the transcriptional induction of a subset of genes in the rat pineal gland.


Assuntos
Ritmo Circadiano/fisiologia , Histonas/genética , Histonas/metabolismo , Norepinefrina/farmacologia , Glândula Pineal/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Animais , Aurora Quinases , Células Cultivadas , Ritmo Circadiano/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Especificidade de Órgãos/efeitos dos fármacos , Especificidade de Órgãos/genética , Fosforilação/efeitos dos fármacos , Glândula Pineal/efeitos dos fármacos , Protamina Quinase/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Ratos , Ratos Sprague-Dawley
18.
Biol Reprod ; 80(4): 813-22, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19109225

RESUMO

Abnormal regulation of placental apoptosis and proliferation has been implicated in placental disorders. Recently, several DNA-damaging agents were reported to induce excessive apoptosis and reduce cell proliferation in the placenta; however, the molecular pathways of these toxic effects on the placenta are unclear. The aim of the present study was to determine the involvement of TRP53, a tumor suppressor that mediates cellular responses to DNA damage, in the induction of apoptosis and cell cycle arrest in the developing placenta. For this purpose, we treated pregnant mice on Day 12 of gestation with 10 mg/kg of etoposide and 5-Gy gamma irradiation, potent inducers of DNA damage. We found an increase in the number of trophoblastic apoptoses 8 and 24 h after etoposide injection and 6 and 24 h after irradiation in the placental labyrinth zone. The number of mitoses and DNA syntheses in trophoblasts decreased after treatment. The accumulation and phosphorylation of TRP53 protein were detected 8 and 6 h after etoposide injection and irradiation, respectively. In Trp53-deficient placentas, the induction of etoposide-induced trophoblastic apoptosis is abrogated, while the reduction of proliferation occurred similarly as in wild-type placentas. CDC2A, a regulator of G2/M progression, was inactivated by phosphorylation after etoposide injection and irradiation, suggesting that the cell cycle was arrested at the G2/M border by treatment. Our study demonstrated that etoposide injection induced TRP53-dependent apoptosis and TRP53-independent cell cycle arrest in labyrinthine trophoblasts, providing insights into the molecular pathway of placental disorders.


Assuntos
Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Etoposídeo/farmacologia , Trofoblastos/efeitos dos fármacos , Proteína Supressora de Tumor p53/fisiologia , Animais , Apoptose/genética , Apoptose/efeitos da radiação , Ciclo Celular/genética , Proliferação de Células/efeitos dos fármacos , Feminino , Raios gama , Histonas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fosforilação/efeitos dos fármacos , Placenta/efeitos dos fármacos , Placenta/metabolismo , Placenta/efeitos da radiação , Placentação , Gravidez , Protamina Quinase/metabolismo , Trofoblastos/metabolismo , Trofoblastos/efeitos da radiação , Proteína Supressora de Tumor p53/genética
19.
J Bacteriol ; 190(20): 6795-804, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18708494

RESUMO

The DevS histidine kinase of Mycobacterium smegmatis contains tandem GAF domains (GAF-A and GAF-B) in its N-terminal sensory domain. The heme iron of DevS is in the ferrous state when purified and is resistant to autooxidation from a ferrous to a ferric state in the presence of O(2). The redox property of the heme and the results of sequence comparison analysis indicate that DevS of M. smegmatis is more closely related to DosT of Mycobacterium tuberculosis than DevS of M. tuberculosis. The binding of O(2) to the deoxyferrous heme led to a decrease in the autokinase activity of DevS, whereas NO binding did not. The regulation of DevS autokinase activity in response to O(2) and NO was not observed in the DevS derivatives lacking its heme, indicating that the ligand-binding state of the heme plays an important role in the regulation of DevS kinase activity. The redox state of the quinone/quinol pool of the respiratory electron transport chain appears not to be implicated in the regulation of DevS activity. Neither cyclic GMP (cGMP) nor cAMP affected DevS autokinase activity, excluding the possibility that the cyclic nucleotides serve as the effector molecules to modulate DevS kinase activity. The three-dimensional structure of the putative GAF-B domain revealed that it has a GAF folding structure without cyclic nucleotide binding capacity.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Mycobacterium smegmatis/fisiologia , Óxido Nítrico/metabolismo , Oxigênio/metabolismo , Protamina Quinase/química , Protamina Quinase/metabolismo , Transdução de Sinais , Sequência de Aminoácidos , Cristalografia por Raios X , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Compostos Ferrosos/metabolismo , Heme/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Fosforilação , Estrutura Terciária de Proteína , Alinhamento de Sequência , Ubiquinona/metabolismo , Vitamina K 2/metabolismo
20.
Biochem Biophys Res Commun ; 372(1): 137-41, 2008 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-18477470

RESUMO

NF-kappaB plays an important role in oncogenesis. Recently, we have demonstrated that loss of p53 function enhances DNA binding and transcriptional activities of NF-kappaB via IKKalpha and IKKbeta, and that glycolysis, activated by NF-kappaB, has an integral role in oncogene-induced cell transformation. Here, we show that ectopically expressed p53 induces acetylation and phosphorylation at Ser 536 of p65, an NF-kappaB component, and enhances DNA-binding activity of NF-kappaB. However, activated p53 suppresses transcriptional activity of NF-kappaB. Under non-stimulating conditions, p65 formed a complex with IKKalpha and IKKbeta. Activated p53 bound to p65 on DNA and disrupted binding of p65 to IKKbeta. Moreover, histone H3 kinase activity, which requires transcriptional activation of NF-kappaB, was diminished by p53. Thus, activated p53 may suppress transcriptional activity of NF-kappaB through inhibition of IKK and histone H3 kinase on DNA, suggesting a novel p53-mediated suppression system for tumorigenesis.


Assuntos
DNA/metabolismo , Fator de Transcrição RelA/antagonistas & inibidores , Fator de Transcrição RelA/metabolismo , Ativação Transcricional , Proteína Supressora de Tumor p53/metabolismo , Acetilação , Animais , Humanos , Quinase I-kappa B/antagonistas & inibidores , Quinase I-kappa B/metabolismo , Camundongos , Fosforilação , Protamina Quinase/antagonistas & inibidores , Protamina Quinase/metabolismo , Ligação Proteica , Serina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA