Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Exp Cell Res ; 415(1): 113110, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35351403

RESUMO

The Polycomb Repressive Complex (PRC) proteins, EZH2 and EZH1 regulate many biological processes by generating the repressive H3K27me3 modifications in the chromatin. However, the factors that regulate the EZH1/EZH2 functions are poorly studied. We identify that the 3'UTRs of EZH2 and EZH1 mRNAs contain the binding sites for the miRNA, miR-150. MicroRNA-150 (miR-150) controls numerous biological processes including cell proliferation, differentiation and pathogenesis of a variety of diseases including cancer. We find that miR-150 regulates the levels of EZH1 and EZH2 through various experimental investigations. Since EZH2 is known to form a repressive complex with other epigenetic repressors especially DNMT3A and DNMT3B, we investigated whether miR-150 also regulates the DNMT3A and DNMT3B levels. We report that miR-150 regulates DNMT3A and DNMT3B levels through direct and indirect mechanisms respectively. Since these epigenetic repressors promote cell proliferation, we investigated the effect of miR-150 perturbation on HEK293 cell proliferation. We found that miR-150 inhibits cell proliferation and induces S-phase arrest by increasing the levels of tumor suppressors and decreasing the cell cycle regulators. Collectively, our study shows that miR-150 act as a tumor suppressor by down-regulating the oncogenic major epigenetic repressors and controls cell proliferation.


Assuntos
MicroRNAs , Complexo Repressor Polycomb 2 , Linhagem Celular Tumoral , Proliferação de Células/genética , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Epigênese Genética/genética , Células HEK293 , Humanos , MicroRNAs/genética , Complexo Repressor Polycomb 2/genética , Complexo Repressor Polycomb 2/metabolismo , Proteínas do Grupo Polycomb/genética
2.
Mol Cell Biochem ; 439(1-2): 75-85, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28779345

RESUMO

In the cell, misfolded proteins are processed by molecular chaperone-mediated refolding or through ubiquitin-mediated proteosome system. Dysregulation of these mechanisms facilitates the aggregation of misfolded proteins and forms aggresomes in the juxta nuclear position of the cell which are removed by lysosome-mediated autophagy pathway in the subsequent cell division. Accumulation of misfolded proteins in the cell is hallmark of several neurological disorders and other diseases including cancer. However, the exact mechanism of aggresome formation and clearance is not thoroughly understood. Reports have shown that several proteins including p300, p53, TAU, α-synuclein, SOD, etc. contain intrinsically disordered region (IDR) which has the tendency to form aggresome. To study the nature of aggresome formation and stability of the aggresome, we have chosen Twist1 as a model protein since it has IDR regions. Twist1 is a bHLH transcription factor which plays a major role in epithelial mesenchymal transition (EMT) and shown to interact with HAT domain of p300 and p53. In the present study, we generated several deletion mutants of human Twist1 with different fluorescent tags and delineated the regions responsible for aggresome formation. The Twist1 protein contains two NLS motifs at the N-terminal region. We showed that the deletions of regions spanning the amino acids 30-46 (Twist1Δ30-46) which lacks the first NLS motif form larger and intense aggregates while the deletion of residues from 47 to 100 (Twist1Δ47-100) which lacks the second NLS motif generates smaller and less intense aggregates in the juxta nuclear position. This suggests that both the NLS motifs are needed for the proper nuclear localization of Twist1. The aggresome formation of the Twist1 deletion mutants was confirmed by counterstaining with known aggresome markers: Vimentin, HDAC6, and gamma tubulin and further validated by MG-132 treatment. In addition, it was found that the aggresomes generated by the Twist1Δ30-46 construct are more stable than the aggresome produced by the Twist1Δ47-100 construct as well as the wild-type Twist1 protein. Taken together, our data provide an important understanding on the role of IDR regions on the formation and stability of aggresomes.


Assuntos
Sequência de Aminoácidos , Proteínas Nucleares/metabolismo , Agregados Proteicos , Dobramento de Proteína , Deleção de Sequência , Proteína 1 Relacionada a Twist/metabolismo , Células HEK293 , Humanos , Proteínas Nucleares/genética , Proteína 1 Relacionada a Twist/genética
3.
Mol Cell Biochem ; 401(1-2): 219-28, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25542213

RESUMO

Regulated polyubiquitination is a key step for controlling protein degradation and maintaining proper balance between the proliferation of normal and uncontrolled cells. Addition of ubiquitin to the proteins by E3 ubiquitin ligases targets them for degradation by the 26S proteosome machinery. Discrepancies in ubiquitination and/or proteosome degradation might lead to multiple genetic disorders in humans. It is reported that CUL1 and BRCA1 ubiquitin ligases localize on centrosome region and regulate the centrosome duplication cycle for genomic stability. In the current study, we predicted the possible interaction of E3 ubiquitin ligase CUL4A complex with γ-tubulin, a centrosome-specific protein, using bioinformatic protein-protein docking analysis. We also confirmed their interaction by performing co-immunoprecipitation studies using endogenous CUL4A/B and stable cell lines that overexpress Flag-CUL4A or Flag-CUL4B. We additionally noted that the γ-tubulin was polyubiquitinated by CUL4A or 4B immune complex indicating that CUL4A or CUL4B may regulate the stability of γ-tubulin. Furthermore, the inhibition of proteosomal degradation pathway using MG132 or LLNV drugs resulted in accumulation and co-localization of CUL4A with γ-tubulin in the centrosome region. Overall, our observation has identified γ-tubulin as a novel target for E3 ubiquitin ligase CUL4 complex, and might lead to the establishment of a unique mechanism for controlling centrosome stability.


Assuntos
Proteínas Culina/química , Proteínas Culina/metabolismo , Tubulina (Proteína)/metabolismo , Centrossomo/metabolismo , Células HEK293 , Células HeLa , Humanos , Leupeptinas/farmacologia , Modelos Moleculares , Simulação de Acoplamento Molecular , Proteólise , Tubulina (Proteína)/química , Ubiquitinação , Valina/análogos & derivados , Valina/farmacologia
4.
Biol Blood Marrow Transplant ; 20(4): 480-9, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24374212

RESUMO

Epigenetic therapies, including DNA methyltransferase and histone deacetylase (HDAC) inhibitors, are increasingly being considered to treat hematological malignancies, but their effects on normal hematopoietic stem cells (HSCs) remain largely unexplored. We compared the effects of several HDAC inhibitors, including valproic acid (VPA) and trichostatin A (TSA), alone or in combination with 5-aza-2'-deoxycytidine (5azaD) on the expansion of HSCs. VPA induced the highest expansion of CD34+CD90+ cells and progenitor cells compared with other HDAC inhibitors or the sequential addition of 5azaD/TSA in culture. Xenotransplantation studies demonstrated that VPA prevents HSC loss, whereas 5azaD/TSA treatment leads to a net expansion of HSCs that retain serial transplantation ability. 5azaD/TSA-mediated HSC expansion was associated with increased histone acetylation and transient DNA demethylation, which corresponded with higher gene transcript levels. However, some genes with increased transcript levels lacked changes in methylation. Importantly, a global microarray analysis revealed a set of differentially expressed genes in 5azaD/TSA- and VPA-expanded CD34+ cells that might be involved in the expansion and maintenance of transplantable HSCs, respectively. In summary, our data indicate that treatment of HSCs with different chromatin-modifying agents results in either the expansion or maintenance of HSCs, an observation of potential therapeutic importance.


Assuntos
Epigênese Genética , Sangue Fetal/efeitos dos fármacos , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Histonas/genética , Animais , Antígenos CD34/genética , Antígenos CD34/imunologia , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Metilação de DNA , Decitabina , Sangue Fetal/citologia , Sangue Fetal/imunologia , Expressão Gênica , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Histonas/metabolismo , Humanos , Ácidos Hidroxâmicos/farmacologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Antígenos Thy-1/genética , Antígenos Thy-1/imunologia , Transplante Heterólogo , Ácido Valproico/farmacologia
5.
Mol Cell Biochem ; 390(1-2): 93-100, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24452595

RESUMO

Controlled protein ubiquitination through E3 ubiquitin ligases and degradation via 26S proteasome machinery is required for orderly progression through cell cycle, chromatin remodeling, DNA repair, and development. Each cullin-dependent ubiquitin ligase (E3) complex can recruit various substrates for their degradation. Cullin 4A (CUL4A) and Cullin 4B (CUL4B) are members of cullin family proteins that mediate ubiquitin dependent proteolysis. Though, these two cul4 genes are functionally redundant, Cullin 4B is not a substitute for all the Cullin 4A functions. Published report has shown that CUL4A interacts with p53 and induces its decay. Although, CUL4A has been known to control several cellular processes, little is known about CUL4B functions. Therefore, in this study, we analyzed the role of CUL4B on p53 polyubiquitination. Our stable cell line and transient transfection studies show that CUL4B indeed interacts with p53 and induces its polyubiquitination. Importantly, both CUL4A and CUL4B overexpressing cells show almost equal levels of p53 polyubiquitination. Moreover, we observed an increased level of polyubiquitination on p53 in CUL4B overexpressing stable cell line upon treatment with siRNA specific for CUL4A indicating that CUL4B plays a vital role in p53 stability. In addition, we have observed the differential expression of CUL4B in various eukaryotic cell lines and mouse tissues suggesting the important role of CUL4B in various tissues. Together, these observations establish an important negative regulatory role of CUL4B on p53 stability.


Assuntos
Proteínas Culina/genética , Proteína Supressora de Tumor p53/biossíntese , Ubiquitina-Proteína Ligases/genética , Animais , Regulação da Expressão Gênica , Instabilidade Genômica , Células HEK293 , Humanos , Camundongos , Proteólise , Proteína Supressora de Tumor p53/genética , Ubiquitina-Proteína Ligases/biossíntese , Ubiquitinação/genética
6.
J Biol Chem ; 286(34): 29725-33, 2011 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-21705330

RESUMO

With the widespread application of carbon nanotubes (CNTs) in diverse commercial processes, scientists are now concerned about the potential health risk of occupational exposures. In this study, CNT-induced pulmonary toxicity was investigated by exposing BALB/c mice to aerosolized single-wall (SW) CNT and multiwall (MW) CNT (5 µg/g of mice) for 7 consecutive days in a nose-only exposure system. Microscopic studies showed that inhaled CNTs were homogeneously distributed in the mouse lung. The total number of bronchoalveolar lavage polymorphonuclear leukocytes recovered from the mice exposed to SWCNT and MWCNT (1.2 × 10(6) ± 0.52 and 9.87 × 10(5) ± 1.45; respectively) was significantly greater than control mice (5.46 × 10(5) ± 0.78). Rapid development of pulmonary fibrosis in mice that inhaled CNT was also confirmed by significant increases in the collagen level. The lactate dehydrogenase levels were increased nearly 2- and 2.4-fold in mice that inhaled SWCNT and MWCNT, respectively, as compared with control mice. In addition, exposure of CNTs to mice showed a significant (p < 0.05) reduction of antioxidants (glutathione, superoxide dismutase, and catalase) and induction of oxidants (myloperoxidase, oxidative stress, and lipid peroxidation) compared with control. Apoptosis-related proteins such as caspase-3 and -8 activities were also significantly increased in mice that inhaled CNT than in control mice. Together, this study shows that inhaled CNTs induce inflammation, fibrosis, alteration of oxidant and antioxidant levels, and induction of apoptosis-related proteins in the lung tissues to trigger cell death.


Assuntos
Pulmão/metabolismo , Teste de Materiais , Nanotubos de Carbono/efeitos adversos , Pneumonia/induzido quimicamente , Pneumonia/metabolismo , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/metabolismo , Aerossóis , Animais , Antioxidantes/metabolismo , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Caspase 8/metabolismo , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Estresse Oxidativo/efeitos dos fármacos , Oxirredutases/metabolismo , Pneumonia/patologia , Fibrose Pulmonar/patologia
7.
Mol Cell Biochem ; 360(1-2): 189-95, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21948272

RESUMO

Although radiation related research has been conducted extensively, the molecular toxicology and cellular mechanisms affected by proton radiation remain poorly understood. We recently reported that the high energy protons induce cell death through activation of apoptotic signaling genes; caspase 3 and 8 (Baluchamy et al. J Biol Chem 285:24769-24774, 2010). In this study, we investigated the effect of different doses of protons in in vivo mouse system, particularly, brain tissues. A significant dose-dependent induction of reactive oxygen species and lipid peroxidation and reduction of antioxidants; glutathione and superoxide dismutase were observed in proton irradiated mouse brain as compared to control brain. Furthermore, histopathology studies on proton irradiated mouse brain showed significant tissue damage as compared to control brain. Together, our in vitro and in vivo results suggest that proton irradiation alters oxidant and antioxidant levels in the cells to cause proton mediated DNA/tissue damage followed by apoptotic cell death.


Assuntos
Encéfalo/efeitos da radiação , Prótons , Lesões por Radiação/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Antioxidantes/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Glutationa/metabolismo , Peroxidação de Lipídeos/efeitos da radiação , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Prótons/efeitos adversos , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1
8.
J Biol Chem ; 285(32): 24769-74, 2010 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-20538614

RESUMO

Radiation affects several cellular and molecular processes, including double strand breakage and modifications of sugar moieties and bases. In outer space, protons are the primary radiation source that poses a range of potential health risks to astronauts. On the other hand, the use of proton irradiation for tumor radiation therapy is increasing, as it largely spares healthy tissues while killing tumor tissues. Although radiation-related research has been conducted extensively, the molecular toxicology and cellular mechanisms affected by proton irradiation remain poorly understood. Therefore, in this study, we irradiated rat lung epithelial cells with different doses of protons and investigated their effects on cell proliferation and death. Our data show an inhibition of cell proliferation in proton-irradiated cells with a significant dose-dependent activation and repression of reactive oxygen species and antioxidants glutathione and superoxide dismutase, respectively, compared with control cells. In addition, the activities of apoptosis-related genes such as caspase-3 and -8 were induced in a dose-dependent manner with corresponding increased levels of DNA fragmentation in proton-irradiated cells compared with control cells. Together, our results show that proton irradiation alters oxidant and antioxidant levels in cells to activate the apoptotic pathway for cell death.


Assuntos
Antioxidantes/química , Células Epiteliais/citologia , Pulmão/citologia , Oxidantes/química , Animais , Caspase 3/metabolismo , Caspase 8/metabolismo , Morte Celular , Sobrevivência Celular , Fragmentação do DNA , Relação Dose-Resposta a Droga , Glutationa/química , Prótons , Ratos , Espécies Reativas de Oxigênio
9.
J Nanosci Nanotechnol ; 11(8): 6868-74, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22103092

RESUMO

The synthesis of nanoparticles and their functionalization to effectively utilize them in biological applications including drug delivery is currently a challenge. Calcium carbonate among many other inorganic nanosized particles offers promising results for such applications. We have synthesized calcium carbonate nanoparticles using polymer mediated growth technique, where one of the ions bound within polymer matrix and the other diffuses and reacts to form desired compound. The synthesized nanoparticles are characterized using X-ray diffraction, Scanning Electron Microscopy and spectroscopic techniques such as Fourier-Transform Infra-red spectroscopy and UV-Vis spectroscopy. The diameter of the calcium carbonate nanoparticles is estimated to be 39.8 nm and their biocompatibility studies showed no significant induction of oxidative stress or cell death even at higher concentrations (50 microg) upon exposure to HeLa and LE cells. Here, we report that the synthesized calcium carbonate nanosized particles using polymer mediated growth technique are biocompatible and can be safely used for biomedical applications.


Assuntos
Materiais Biocompatíveis , Carbonato de Cálcio/síntese química , Carbonato de Cálcio/química , Células HeLa , Humanos , Microscopia Eletrônica de Varredura , Espectrofotometria Ultravioleta , Espectroscopia de Infravermelho com Transformada de Fourier , Difração de Raios X
10.
J Mater Sci Mater Med ; 22(10): 2301-9, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21823031

RESUMO

Here, we have synthesized Zinc Oxide (ZnO) nanorods at room temperature using zinc acetate and hexamethylenetetramine as precursors followed by characterization using X-ray diffraction (XRD), fourier transform infra red spectroscopy, scanning electron microscopy (SEM) and transmission electron microscopy. The growth of the synthesized ZnO was found to be very close to its hexagonal nature, which is confirmed by XRD. The nanorods were grown perpendicular to the long-axis and grew along the [001] direction, which is the nature of ZnO growth. The morphology of the synthesized ZnO nanorods was also confirmed by SEM. The size of the nanorod was estimated to be around 20-25 nm in diameter and approximately 50-60 nm in length. Our biocompatibility studies using synthesized ZnO showed no significant dose- or time-dependent increase in the formation of free radicals, accumulation of peroxidative products, antioxidant depletion or loss of cell viability on lung epithelial cells.


Assuntos
Nanopartículas/efeitos adversos , Nanopartículas/química , Óxido de Zinco/efeitos adversos , Óxido de Zinco/química , Animais , Antioxidantes/metabolismo , Materiais Biocompatíveis/efeitos adversos , Materiais Biocompatíveis/química , Linhagem Celular , Sobrevivência Celular , Células Epiteliais/efeitos dos fármacos , Glutationa/metabolismo , Microscopia Eletrônica de Varredura , Estresse Oxidativo/efeitos dos fármacos , Ratos
11.
Apoptosis ; 15(12): 1507-16, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20694747

RESUMO

Our previous report on multiwall carbon nanotubes (MWCNT) has demonstrated the generation of reactive radicals and depletion of intracellular antioxidants which in turn cause cell death through activation of caspases. The molecular mechanism of cellular death due to MWCNT is not clear yet. In this study, we investigated the signaling pathways implicated in MWCNT-induced apoptosis in rat lung epithelial cells. First, we assessed the DNA damage in response to MWCNT treatment and showed the significant DNA damage as compared to control. The collapse of the mitochondrial membrane integrity, release of cytochrome c into the cytosol, reduction in cellular ATP content, increased levels of mitochondrial apoptogenic factor and activation and nuclear translocation of NF-κB were observed in MWCNT treated cells. In addition, a time-dependent induction of phosphorylated IκBα and its degradation were detected in cells exposed to MWCNT. Furthermore, MWCNT activated several death related proteins including apoptosis inducing factor, p53, p21 and bax. Together, our results suggest that signaling pathways such as NF-κB and AP-1 are activated upon MWCNT treatment for cellular cytotoxicity.


Assuntos
Trifosfato de Adenosina/análise , Fator de Indução de Apoptose/metabolismo , Apoptose , Citocromos c/análise , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/fisiologia , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , NF-kappa B/metabolismo , Nanotubos de Carbono , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/fisiologia , Fator de Transcrição AP-1/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Linhagem Celular , Pulmão/fisiologia , Potencial da Membrana Mitocondrial/fisiologia , Nanotecnologia , Nanotubos de Carbono/toxicidade , Ratos , Mucosa Respiratória/ultraestrutura , Transdução de Sinais
12.
Mol Cell Biochem ; 341(1-2): 207-15, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20383738

RESUMO

Exposure of living systems to radiation results in a wide assortment of lesions, the most significant of is damage to genomic DNA which alter specific cell functions including cell proliferation. The radiation induced DNA damage investigation is one of the important area in biology, but still the information available regarding the effects of proton is very limited. In this report, we investigated the differential gene expression pattern of DNA damage signaling genes such as damaged DNA binding, repair, cell cycle arrest, checkpoints and apoptosis using quantitative real-time RT-PCR in proton exposed mouse brain tissues. The expression profiles showed significant changes in DNA damage related genes in 2 Gy proton exposed mouse brain tissues as compared to control brain tissues. Furthermore, we also show that significantly increased levels of apoptotic related genes, caspase-3 and 8 activities in these cells, suggesting that in addition to differential expression of DNA damage genes, the alteration of apoptosis related genes may also contribute to the radiation induced DNA damage followed by programmed cell death. In summary, our findings suggest that proton exposed cells undergo severe DNA damage which in turn destabilize the chromatin stability.


Assuntos
Apoptose/genética , Encéfalo/efeitos da radiação , Dano ao DNA/genética , Perfilação da Expressão Gênica , Animais , Apoptose/efeitos da radiação , Dano ao DNA/efeitos da radiação , Fragmentação do DNA , Relação Dose-Resposta à Radiação , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Prótons/efeitos adversos , Transdução de Sinais/genética
13.
Biochimie ; 176: 85-102, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32659446

RESUMO

Epigenetic modifications govern gene expression by guiding the human genome on 'what to express and what not to'. DNA methyltransferases (DNMTs) establish methylation patterns on DNA, particularly in CpG islands, and such patterns play a major role in gene silencing. DNMTs are a family of proteins/enzymes (DNMT1, 2, 3A, 3B, and 3L), among which, DNMT1 (maintenance methyltransferase) and DNMT3 (de novo methyltransferases) that direct mammalian development and genome imprinting are highly investigated. In recent decades, many studies revealed a strong association of DNA methylation patterns with gene expression in various clinical conditions. Differential expression of DNMT3 family proteins and their splice variants result in changes in methylation patterns and such alterations have been associated with the initiation and progression of various diseases, especially cancer. This review will discuss the aberrant modifications generated by DNMT3 proteins under various clinical conditions, suggesting a potential signature for de novo methyltransferases in targeted disease therapy. Further, this review discusses the possibility of using 'CpG island methylation signatures' as promising biomarkers and emphasizes 'targeted hypomethylation' by disrupting the interaction of specific DNMT-protein complexes as the future of cancer therapeutics.


Assuntos
DNA (Citosina-5-)-Metiltransferases , Metilação de DNA , DNA de Neoplasias/metabolismo , Sistemas de Liberação de Medicamentos , Inibidores Enzimáticos/uso terapêutico , Proteínas de Neoplasias , Neoplasias , Animais , DNA (Citosina-5-)-Metiltransferases/antagonistas & inibidores , DNA (Citosina-5-)-Metiltransferases/metabolismo , Humanos , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/patologia
14.
Med Oncol ; 37(8): 68, 2020 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-32710193

RESUMO

CREB signaling is known for several decades, but how it regulates both positive and negative regulators of cell proliferation is not well understood. On the other hand functions of major epigenetic repressors such as DNMT3B, EZH2 and CUL4B for their repressive epigenetic modifications on chromatin have also been well studied. However, there is very limited information available on how these repressors are regulated at their transcriptional level. Here, using computational tools and molecular techniques including site directed mutagenesis, promoter reporter assay, chromatin immunoprecipitation (ChIP), we identified that CREB acts as a common transcription factor for DNMT3B, EZH2, CUL4B and E2F6. ChIP assay revealed that pCREB binds to promoters of these repressors at CREs and induce their transcription. As expected, the expression of these repressors and their associated repressive marks particularly H3K27me3 and H2AK119ub are increased and decreased upon CREB overexpression and knock-down conditions respectively in the cancer cells indicating that CREB regulates the functions of these repressors by activating their transcription. Since CREB and these epigenetic repressors are overexpressed in various cancer types, our findings showed the molecular relationship between them and indicate that CREB is an important therapeutic target for cancer therapy.


Assuntos
Proteínas Culina/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , DNA (Citosina-5-)-Metiltransferases/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Neoplasias/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Biologia Computacional/métodos , Proteínas Culina/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , DNA (Citosina-5-)-Metiltransferases/genética , Fator de Transcrição E2F6/genética , Fator de Transcrição E2F6/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Epigênese Genética , Humanos , Neoplasias/genética , Neoplasias/patologia , Regiões Promotoras Genéticas , Transdução de Sinais , DNA Metiltransferase 3B
15.
Med Oncol ; 36(2): 20, 2019 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-30666499

RESUMO

CUL4A; an E3 ubiquitin ligase is involved in the degradation of negative regulators of cell cycle such as p21, p27, p53, etc., through polyubiquitination-mediated protein degradation. The functional role(s) of CUL4A proteins on their targets are well characterized; however, the transcriptional regulation of CUL4A, particularly at its promoter level is not yet studied. Therefore, in this study, using computational tools, we found cAMP responsive elements (CRE) at the locations of - 926 and - 764 with respect to transcription state site + 1 of CUL4A promoter. Hence, we investigated the role of CREB on the regulation of CUL4A transcription. Our chromatin immunoprecipitation (ChIP) data clearly showed increased levels of promoter occupancy of both CREB and pCREB on both CREs of CUL4A promoter. As expected, the expression of CUL4A increases and decreases upon the overexpression of and knocking down of CREB, respectively. Moreover, the inhibition of ERK pathway by U0126 not only reduces the CREB activation but also the CUL4A levels suggesting that CREB is the upstream activator of CUL4A transcription. The reduction of CUL4A levels upon the knocking down of CREB or by U0126 treatment increases the protein levels of CUL4A substrates such as p21 and p27. It is reported that CUL4A activates the ERK1/2 transcription and ERK1/2 pathway activates the CREB by phosphorylation. Based on our data and earlier findings, we report that CREB regulates the CUL4A levels positively which in turn activates the CREB through ERK1/2 pathway in the form of auto-regulatory looped mechanism.This suggests that CUL4A might be involved in proliferation of cancer cells by regulating the ERK1/2 and CREB signaling.


Assuntos
Proteínas Culina/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , MAP Quinases Reguladas por Sinal Extracelular/genética , Retroalimentação Fisiológica , Regulação da Expressão Gênica/genética , Proteínas Culina/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Células HeLa , Humanos , Sistema de Sinalização das MAP Quinases/genética , Regiões Promotoras Genéticas/genética
16.
Med Oncol ; 34(8): 146, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28730334

RESUMO

MiR-181a-2 plays a major role in cell proliferation both positively and negatively depending on tissue type by targeting several regulators 3'UTR regions. We have predicted several targets for miR-181a-2 through computational approaches and characterized one its interesting target, CUL4A, an E3 ubiquitin ligase. CUL4A regulates diverse functions in the cells including DNA repair, DNA replication, cell cycle, genomic stability through polyubiquitination of target proteins. Deregulation of both miR-181a-2 and CUL4A are reported in many cancerous cells, but the functional link between them is unknown. We show that miR-181a-5p binds to 3'UTR of CUL4A and regulates its transcripts levels in HEK293 cells through overexpression studies. In addition, by using MTT and Neutral red assays, we showed that miR-181a-2 overexpression increased the proliferation in HEK293 cells. Moreover, cell cycle analysis using flow cytometer revealed that an increase in S-phase cells upon the overexpression of miR-181a-2. Though several miRNAs are known to downregulate the CUL4A levels, here we show that miR-181a-2 also participates in the downregulation of CUL4A. Taken together, our data demonstrated that miR-181a-2 increases the cell proliferation in HEK293 cells possibly through the downregulation of CUL4A.


Assuntos
Regiões 3' não Traduzidas , Proliferação de Células , Proteínas Culina/genética , MicroRNAs/genética , Ciclo Celular , Proteínas Culina/metabolismo , Regulação para Baixo , Células HEK293 , Humanos
17.
Med Oncol ; 34(8): 145, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28730333

RESUMO

DNA methyltransferases make use of alternative splicing mechanism to generate various splice variants, but their role(s) in modulating DNA methylation patterns in the cells is unclear. Notably, DNMT3B alone contains nearly 40 different splice variants. In this study, we have identified a novel splice variant of DNMT3B, which lacks exon 7 and 10 from leukemic cell lines which we termed as DNMT3B9. The exon 7 codes for the major part of PWWP domain, and exon 10 inclusion serves as a pluripotent marker. By quantitative RT-PCR using exon-exon junction-specific primers, we showed higher level of DNMT3B9 transcripts in several leukemic cell lines. However, DNMT3B9 expression was less in other tested cancer cell lines indicating that DNMT3B9 might serve as a leukemic-specific biomarker. Surprisingly, endogenous protein for DNMT3B9 was not detected in leukemic cells suggesting the unidentified RNA-related function(s) for DNMT3B9. In addition, we showed that DNMT3B9 protein lacks PWWP domain is less stable compared to other DNMT3B variants which contain PWWP domain through computational predictions and by cycloheximide half-life experiment. Taken together, we demonstrated the existence of novel leukemic-specific splice variant of DNMT3B and provide the evidence for the role of PWWP domain in the stability of DNMT3B. The findings reported here have relevance in epigenetic therapy, which is aimed to target the DNMT3B in cancer cells.


Assuntos
Processamento Alternativo , DNA (Citosina-5-)-Metiltransferases/química , DNA (Citosina-5-)-Metiltransferases/genética , Metilação de DNA , Éxons , Leucemia/genética , Neoplasias/genética , Sequência de Aminoácidos , Sequência de Bases , DNA (Citosina-5-)-Metiltransferases/metabolismo , Humanos , Conformação Proteica , Homologia de Sequência , Células Tumorais Cultivadas , DNA Metiltransferase 3B
18.
Virus Res ; 108(1-2): 69-81, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15681057

RESUMO

We have identified and characterized a cyclin homolog from Bombyx mori nucleopolyhedrovirus (BmNPV), encoding a 34 kDa protein (ORF 120) with 48% homology to the host Bombyx mori cellular cyclin B. The expression of the viral cyclin (v-cyc) was detected from 12 h following virus infection and the maximum transcript levels were seen at 24-36 h. The transcription start site mapping of v-cyc revealed the presence of a transcript initiating from a TAAG motif located 13 nucleotide (nt) upstream of the ORF as well as longer transcripts initiating from farther upstream region and encompassing the preceding ORF 119. The transcription was terminated at 15 nt downstream of the ORF 120. The expression of the host cellular cyclin B declined following virus infection and the transcript disappeared almost completely by 24 h even as the expression of v-cyc reached high levels. The synthesis of the viral cyclin was detected at 36-48 h post-infection. The viral cyclin in association with other host or viral proteins catalysed phosphorylation of histone H1. The host cells were arrested in G2/M phase following virus infection and thus, the virus cyclin in association with other proteins maintains the host cells at the G2/M phase while permitting the virus DNA replication.


Assuntos
Bombyx/virologia , Ciclinas/genética , Ciclinas/fisiologia , Nucleopoliedrovírus/genética , Proteínas Virais/genética , Proteínas Virais/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Northern Blotting , Divisão Celular , Ciclinas/química , DNA Viral/química , DNA Viral/isolamento & purificação , Regulação Viral da Expressão Gênica , Genes Virais , Histonas/metabolismo , Dados de Sequência Molecular , Fases de Leitura Aberta , Fosforilação , RNA Mensageiro/análise , RNA Viral/análise , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Sítio de Iniciação de Transcrição/fisiologia , Proteínas Virais/química
19.
In Vitro Cell Dev Biol Anim ; 46(8): 718-25, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20607620

RESUMO

Radiation is known to potentially interfere with cellular functions at all levels of cell organization. The radiation-induced stress response is very complex and involves altered expression of many genes. Identification of specific genes may allow the determination of pathways important in radiation responses. Although several radiation-related research have been studied extensively, the molecular and cellular processes affected by proton exposure remain poorly understood. Our earlier reports have shown that proton radiation induces reactive oxygen species (ROS) formation and lipid peroxidation and inhibits antioxidants, superoxide dismutase, and glutathione. Therefore, in this present study, we used quantitative real-time reverse transcription polymerase chain reaction approach and showed the modulation of several genes including oxidative stress, antioxidants defense mechanism, ROS metabolism, and oxygen transporters related genes expression in 2-Gy proton-exposed mouse brain. Literature evidences suggest that change in oxidants and antioxidants levels induce DNA damage, followed by cell death. In conclusion, changes in the gene profile of mouse brain after proton irradiation are complex and the exposed cells might undergo programmed cell death through alteration of genes responsible for oxidative stress signaling mechanism.


Assuntos
Encéfalo/efeitos da radiação , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos da radiação , Estresse Oxidativo/genética , Prótons , Animais , Antioxidantes/metabolismo , Apoptose/genética , Encéfalo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Espécies Reativas de Oxigênio/metabolismo
20.
Exp Hematol ; 37(9): 1084-95, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19501128

RESUMO

OBJECTIVE: Augmentation of the number of cord blood (CB) hematopoietic stem cells (HSC) present in a unit is required before it can be considered as an alternative graft for hematopoietic reconstitution for adult patients. In order to further optimize strategies to augment HSC numbers, we examined whether expansion of HSC mediated by epigenetic mechanisms remains permissive to external environmental cues. MATERIALS AND METHODS: The chromatin-modifying agents 5-aza-2'-deoxycytidine (5azaD) and trichostatin A (TSA) were used to ameliorate epigenetic alteration of CB cells during ex vivo culture by adding various cytokines. After culture, CD34(+)CD90(+) cell numbers, their division history, in vitro clonogenic potential, and in vivo hematopoietic reconstitution potential and frequency were determined. RESULTS: 5azaD/TSA-treated, CD34(+)CD90(+) cells were greatly influenced in terms of their degree of expansion, clonogenic potential, cell-division rate, and transplantability by the combination of cytokines used in culture. Furthermore, our current results verify that the sequential addition of 5azaD followed by TSA is crucial for expansion of HSC. We demonstrate that following 5azaD/TSA treatment, the rate of CD34(+)CD90(+) cell division is also dependent on the cytokine cocktail and that this is associated with functional changes, including alteration of in vitro clonogenic potential and in vivo reconstitution potential. CONCLUSIONS: Our studies indicate there are interactions between intrinsic factors influenced by epigenetic mechanisms and external environmental signals in the regulation of HSC expansion. Epigenetic influences on HSC can be accentuated by environmental factors. Regulation of the rate of divisions may be a critical determinant for the maintenance of HSC functional potency during ex vivo expansion.


Assuntos
Antígenos CD34 , Divisão Celular/fisiologia , Epigênese Genética/fisiologia , Sangue Fetal/citologia , Células-Tronco Hematopoéticas/citologia , Antígenos Thy-1 , Adulto , Animais , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Técnicas de Cultura de Células , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Transplante de Células-Tronco de Sangue do Cordão Umbilical/métodos , Citocinas/metabolismo , Decitabina , Inibidores Enzimáticos/farmacologia , Epigênese Genética/efeitos dos fármacos , Humanos , Ácidos Hidroxâmicos/farmacologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Transplante Heterólogo , Transplante Homólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA