Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Front Immunol ; 12: 782852, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34925363

RESUMO

In recent years, it became apparent that cancers either associated with viral infections or aberrantly expressing endogenous retroviral elements (EREs) are more immunogenic, exhibiting an intense intra-tumor immune cell infiltration characterized by a robust cytolytic apparatus. On the other hand, epigenetic regulation of EREs is crucial to maintain steady-state conditions and cell homeostasis. In line with this, epigenetic disruptions within steady-state cells can lead to cancer development and trigger the release of EREs into the cytoplasmic compartment. As such, detection of viral molecules by intracellular innate immune sensors leads to the production of type I and type III interferons that act to induce an antiviral state, thus restraining viral replication. This knowledge has recently gained momentum due to the possibility of triggering intratumoral activation of interferon responses, which could be used as an adjuvant to elicit strong anti-tumor immune responses that ultimately lead to a cascade of cytokine production. Accordingly, several therapeutic approaches are currently being tested using this rationale to improve responses to cancer immunotherapies. In this review, we discuss the immune mechanisms operating in viral infections, show evidence that exogenous viruses and endogenous retroviruses in cancer may enhance tumor immunogenicity, dissect the epigenetic control of EREs, and point to interferon pathway activation in the tumor milieu as a promising molecular predictive marker and immunotherapy target. Finally, we briefly discuss current strategies to modulate these responses within tumor tissues, including the clinical use of innate immune receptor agonists and DNA demethylating agents.


Assuntos
Epigênese Genética/imunologia , Imunoterapia/métodos , Interferon Tipo I/metabolismo , Interferons/metabolismo , Neoplasias/terapia , Antimetabólitos Antineoplásicos/farmacologia , Antimetabólitos Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Ensaios Clínicos como Assunto , Desmetilação do DNA/efeitos dos fármacos , Retrovirus Endógenos/genética , Retrovirus Endógenos/imunologia , Epigênese Genética/efeitos dos fármacos , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Imunidade Inata/genética , Neoplasias/genética , Neoplasias/imunologia , Vírus Oncolíticos/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Receptor 3 Toll-Like/agonistas , Receptor 3 Toll-Like/metabolismo , Receptor Toll-Like 9/agonistas , Receptor Toll-Like 9/metabolismo , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia , Interferon lambda
2.
Cells ; 10(11)2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34831209

RESUMO

Nutritional intake impacts the human epigenome by directing epigenetic pathways in normal cell development via as yet unknown molecular mechanisms. Consequently, imbalance in the nutritional intake is able to dysregulate the epigenetic profile and drive cells towards malignant transformation. Here we present a novel epigenetic effect of the essential nutrient, NAD. We demonstrate that impairment of DNMT1 enzymatic activity by NAD-promoted ADP-ribosylation leads to demethylation and transcriptional activation of the CEBPA gene, suggesting the existence of an unknown NAD-controlled region within the locus. In addition to the molecular events, NAD- treated cells exhibit significant morphological and phenotypical changes that correspond to myeloid differentiation. Collectively, these results delineate a novel role for NAD in cell differentiation, and indicate novel nutri-epigenetic strategies to regulate and control gene expression in human cells.


Assuntos
Diferenciação Celular , Metilação de DNA/genética , NAD/farmacologia , Proteínas Estimuladoras de Ligação a CCAAT/genética , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Desmetilação do DNA/efeitos dos fármacos , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Células Mieloides/citologia , Células Mieloides/efeitos dos fármacos , Neoplasias/genética , Neoplasias/patologia , Fosforilação Oxidativa/efeitos dos fármacos , Poli Adenosina Difosfato Ribose/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcrição Gênica/efeitos dos fármacos
3.
Nat Commun ; 12(1): 6831, 2021 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-34819502

RESUMO

Exhausted CD8+ T cells are key targets of immune checkpoint blockade therapy and their ineffective reinvigoration limits the durable benefit in some cancer patients. Here, we demonstrate that histone demethylase LSD1 acts to enforce an epigenetic program in progenitor exhausted CD8+ T cells to antagonize the TCF1-mediated progenitor maintenance and to promote terminal differentiation. Consequently, genetic perturbation or small molecules targeting LSD1 increases the persistence of the progenitor exhausted CD8+ T cells, which provide a sustained source for the proliferative conversion to numerically larger terminally exhausted T cells with tumor-killing cytotoxicity, thereby leading to effective and durable responses to anti-PD1 therapy. Collectively, our findings provide important insights into epigenetic mechanisms that regulate T cell exhaustion and have important implications for durable immunotherapy.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Regulação Neoplásica da Expressão Gênica/imunologia , Histona Desmetilases/metabolismo , Inibidores de Checkpoint Imunológico/farmacologia , Neoplasias/imunologia , Animais , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Tumoral/transplante , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Desmetilação do DNA/efeitos dos fármacos , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos/genética , Epigênese Genética/imunologia , Feminino , Células HEK293 , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Histona Desmetilases/antagonistas & inibidores , Histona Desmetilases/genética , Humanos , Inibidores de Checkpoint Imunológico/uso terapêutico , Linfócitos do Interstício Tumoral , Masculino , Camundongos , Camundongos Transgênicos , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Cultura Primária de Células , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/metabolismo , Proteínas Recombinantes
4.
Curr Med Sci ; 41(5): 869-879, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34669117

RESUMO

OBJECTIVE: To investigate the effects and mechanisms of genistein on the gene expression in the Wnt pathway in acute leukemia (AL) cells. METHODS: The expression of Wnt pathway genes and cell cycle-related genes were analyzed in two AL cell lines. Pyrophosphate sequencing was performed to determine the methylation degree. Then, the enrichment of H4K20me1 and H3K9ac was determined using ChIP-qPCR. Flow cytometry was used to analyze the cell cycle. RESULTS: The IC50 of genistein in the two AL cell lines was lower than that for the bone marrow mesenchymal stem cell line. Genistein upregulated H4K20me1, KMT5A and Wnt suppressor genes, including Wnt5a, and downregulated the downstream target genes of Wnt, such as c-myc and ß-catenin. The methylation degree and H3K9ac enrichment in the Wnt5a promoter region remained unchanged. However, the enrichment of H4K20me1 in the Wnt5a promoter and coding regions increased. In addition, genistein upregulated Phospho-cdc2, Myt1, Cyclin A, Cyclin E2, p21 and Phospho-histone H3, but downregulated Phospho-wee1. Cell cycle arrest was induced in the G2/M phase. CONCLUSION: Genistein inhibits the activation of the Wnt pathway by promoting the expression of Wnt5a through the activation of KMT5A and enrichment of H4K20me1 in the Wnt5a gene promoter and coding regions, rather than demethylation. Genistein also blocks the cell cycle in the G2/M phase. Therefore, genistein is a potential anti-leukemia drug.


Assuntos
Anticarcinógenos/farmacologia , Genisteína/farmacologia , Histonas/metabolismo , Leucemia Mieloide Aguda/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Desmetilação do DNA/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Metilação/efeitos dos fármacos , Fosforilação , Análise de Sequência de DNA , Regulação para Cima
5.
Molecules ; 26(13)2021 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-34201712

RESUMO

Alnus sibirica (AS) is distributed in Korea, Japan, China, and Russia and has reported anti-oxidant, anti-inflammatory, and reducing activities on atopic dermatitis-like skin lesions, along with other beneficial health properties. In the present study, we tried to prove the cancer-preventive activity against prostate cancer. The extracted and isolated compounds, oregonin (1), hirsutenone (2), and hirsutanonol (3), which were isolated from AS, were tested for anti-proliferative activity. To do this, we used the MTT assay; NF-κB inhibitory activity, using Western blotting; apoptosis-inducing activity using flow cytometry; DNA methylation activity, using methylation-specific polymerase chain reaction in androgen-dependent (LNCaP) and androgen-independent (PC-3) prostate cancer cell lines. The compounds (1-3) showed potent anti-proliferative activity against both prostate cancer cell lines. Hirsutenone (2) exhibited the strongest NF-κB inhibitory and apoptosis-inducing activities compared with oregonin (1) and hirsutanonol (3). DNA methylation activity, which was assessed for hirsutenone (2), revealed a concentration-dependent enhancement of the unmethylated DNA content and a reduction in the methylated DNA content in both PC-3 and LNCaP cells. Overall, these findings suggest that hirsutenone (2), when isolated from AS, may be a potential agent for preventing the development or progression of prostate cancer.


Assuntos
Alnus/química , Apoptose/efeitos dos fármacos , Catecóis/farmacologia , Proliferação de Células/efeitos dos fármacos , Desmetilação do DNA/efeitos dos fármacos , Diarileptanoides/farmacologia , Glutationa S-Transferase pi/metabolismo , Neoplasias da Próstata/prevenção & controle , Androgênios/farmacologia , Catecóis/química , Catecóis/isolamento & purificação , Linhagem Celular Tumoral , Diarileptanoides/química , Diarileptanoides/isolamento & purificação , Citometria de Fluxo , Humanos , Masculino , NF-kappa B/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/metabolismo
6.
J Immunol Res ; 2021: 5590217, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34239942

RESUMO

Regulatory T (Treg) cells are a subtype of CD4+ T cells that play a significant role in the protection from autoimmunity and the maintenance of immune tolerance via immune regulation. Epigenetic modifications of Treg cells (i.e., cytosine methylation at the promoter region of the transcription factor, Forkhead Box P3) have been found to be closely associated with allergic diseases, including allergic rhinitis, asthma, and food allergies. In this study, we highlighted the recent evidence on the contribution of epigenetic modifications in Treg cells to the pathogenesis of allergic diseases. Moreover, we also discussed directions for future clinical treatment approaches, with a particular emphasis on Treg cell-targeted therapies for allergic disorders.


Assuntos
Metilação de DNA/imunologia , Epigênese Genética/imunologia , Tolerância Imunológica/genética , Hipersensibilidade Respiratória/genética , Linfócitos T Reguladores/imunologia , Animais , DNA (Citosina-5-)-Metiltransferases/antagonistas & inibidores , DNA (Citosina-5-)-Metiltransferases/metabolismo , Desmetilação do DNA/efeitos dos fármacos , Metilação de DNA/efeitos dos fármacos , Modelos Animais de Doenças , Epigênese Genética/efeitos dos fármacos , Humanos , Tolerância Imunológica/efeitos dos fármacos , Regiões Promotoras Genéticas , Hipersensibilidade Respiratória/diagnóstico , Hipersensibilidade Respiratória/tratamento farmacológico , Hipersensibilidade Respiratória/imunologia , Índice de Gravidade de Doença , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/metabolismo , Tretinoína/farmacologia , Tretinoína/uso terapêutico
7.
Proc Natl Acad Sci U S A ; 118(19)2021 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-33941676

RESUMO

Chronic inflammatory diseases like rheumatoid arthritis are characterized by a deficit in fully functional regulatory T cells. DNA-methylation inhibitors have previously been shown to promote regulatory T cell responses and, in the present study, we evaluated their potential to ameliorate chronic and acute animal models of rheumatoid arthritis. Of the drugs tested, decitabine was the most effective, producing a sustained therapeutic effect that was dependent on indoleamine 2,3-dioxygenase (IDO) and was associated with expansion of induced regulatory T cells, particularly at the site of disease activity. Treatment with decitabine also caused apoptosis of Th1 and Th17 cells in active arthritis in a highly selective manner. The molecular basis for this selectivity was shown to be ENT1, a nucleoside transporter, which facilitates intracellular entry of the drug and is up-regulated on effector T cells during active arthritis. It was further shown that short-term treatment with decitabine resulted in the generation of a population of regulatory T cells that were able to suppress arthritis upon adoptive transfer. In summary, a therapeutic approach using an approved drug is described that treats active inflammatory disease effectively and generates robust regulatory T cells with the IDO-dependent capacity to maintain remission.


Assuntos
Artrite Experimental/tratamento farmacológico , Artrite Reumatoide/tratamento farmacológico , Doenças Autoimunes/tratamento farmacológico , Decitabina/farmacologia , Linfócitos T Reguladores/efeitos dos fármacos , Células Th1/efeitos dos fármacos , Células Th17/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Artrite Experimental/imunologia , Artrite Experimental/metabolismo , Artrite Reumatoide/imunologia , Artrite Reumatoide/metabolismo , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Desmetilação do DNA/efeitos dos fármacos , Transportador Equilibrativo 1 de Nucleosídeo/genética , Transportador Equilibrativo 1 de Nucleosídeo/imunologia , Transportador Equilibrativo 1 de Nucleosídeo/metabolismo , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Indolamina-Pirrol 2,3,-Dioxigenase/imunologia , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Knockout , Indução de Remissão , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Células Th1/citologia , Células Th1/imunologia , Células Th17/citologia , Células Th17/imunologia
8.
Clin. transl. oncol. (Print) ; 23(4): 750-756, abr. 2021. graf
Artigo em Inglês | IBECS | ID: ibc-220910

RESUMO

Background Colorectal cancer (CRC) represents an important neoplasm with high mortality. Although PD-L1/PD-1 system-based immunotherapy has benefits for a certain type of CRC, many efforts should be made to enhance the responses to anti-PD-1/PD-L1 drugs. DNA methylation has been critically implicated in the regulation of tumor immunity. Here, we examined the effects of the natural alkaloid oxymatrine on PD-L1 expression in CRC cells and to elucidate the underlying mechanism. Methods Human CRC SW620 and HCT116 cells were treated with interferon γ (IFNγ) and/or oxymatrine. Cell viability was determined using MTT assays. PD-L1 expression was detected by real-time PCR and Western blot analyses. DNA demethylase activity was measured using kits. Results Oxymatrine did not apparently affect the viability of normal human intestinal epithelial cells. IFNγ at 20 ng/ml increased the viability of CRC cells, but oxymatrine concentration-dependently reduced the viability in the absence or presence of IFNγ. IFNγ increased the mRNA and protein expression of PD-L1 in the two cell lines, but oxymatrine significantly abolished IFNγ-elevated PD-L1 levels at both mRNA and protein levels. Furthermore, DNA demethylase activity was remarkably increased in IFNγ-treated CRC cells, which was abolished by oxymatrine concentration-dependently. In addition, DNA methyltransferase inhibitor 5-azacytidine considerably abrogated oxymatrine-induced downregulation of PD-L1 mRNA and protein levels in IFNγ-stimulated CRC cells. Conclusion Oxymatrine suppressed viability and reduced PD-L1 expression in IFNγ-stimulated CRC cells, which was attributed to enhanced DNA demethylation. Our current discoveries suggested oxymatrine as an epigenetic modulatory agent for immunotherapy against CRC via PD-1/PD-L1 blockade (AU)


Assuntos
Humanos , Alcaloides/farmacologia , Antígeno B7-H1/metabolismo , Neoplasias Colorretais/metabolismo , Desmetilação do DNA/efeitos dos fármacos , Quinolizinas/farmacologia , Azacitidina/farmacologia , Antígeno B7-H1/genética , Linhagem Celular Tumoral , RNA Mensageiro/metabolismo
9.
Chem Biol Drug Des ; 97(6): 1170-1184, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33764683

RESUMO

DNA alkylation damage, emanating from the exposure to environmental alkylating agents or produced by certain endogenous metabolic processes, affects cell viability and genomic stability. Fe(II)/2-oxoglutarate-dependent dioxygenase enzymes, such as Escherichia coli AlkB, are involved in protecting DNA from alkylation damage. Inspired by the natural product indenone derivatives reported to inhibit this class of enzymes, and a set of 2-chloro-3-amino indenone derivatives was synthesized and screened for their inhibitory properties against AlkB. The synthesis of 2-chloro-3-amino indenone derivatives was achieved from 2,3-dichloro indenones through addition-elimination method using alkyl/aryl amines under catalyst-free conditions. Using an in vitro reconstituted DNA repair assay, we have identified a 2-chloro-3-amino indenone compound 3o to be an inhibitor of AlkB. We have determined the binding affinity, mode of interaction, and kinetic parameters of inhibition of 3o and tested its ability to sensitize cells to methyl methanesulfonate that mainly produce DNA alkylation damage. This study established the potential of indenone-derived compounds as inhibitors of Fe(II)/2-oxoglutarate-dependent dioxygenase AlkB.


Assuntos
Alquilantes/síntese química , Reparo do DNA , Indenos/química , Alquilantes/farmacologia , Sítios de Ligação , Dano ao DNA , Desmetilação do DNA/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Escherichia coli/enzimologia , Proteínas de Escherichia coli/antagonistas & inibidores , Proteínas de Escherichia coli/metabolismo , Humanos , Indenos/metabolismo , Indenos/farmacologia , Cinética , Oxigenases de Função Mista/antagonistas & inibidores , Oxigenases de Função Mista/metabolismo , Simulação de Acoplamento Molecular , Ligação Proteica
10.
Clin Epigenetics ; 13(1): 33, 2021 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-33573703

RESUMO

BACKGROUND: Although R-CHOP (rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone) remains the standard chemotherapy regimen for diffuse large B cell lymphoma (DLBCL) patients, not all patients are responsive to the scheme, and there is no effective method to predict treatment response. METHODS: We utilized 5hmC-Seal to generate genome-wide 5hmC profiles in plasma cell-free DNA (cfDNA) from 86 DLBCL patients before they received R-CHOP chemotherapy. To investigate the correlation between 5hmC modifications and curative effectiveness, we separated patients into training (n = 56) and validation (n = 30) cohorts and developed a 5hmC-based logistic regression model from the training cohort to predict the treatment response in the validation cohort. RESULTS: In this study, we identified thirteen 5hmC markers associated with treatment response. The prediction performance of the logistic regression model, achieving 0.82 sensitivity and 0.75 specificity (AUC = 0.78), was superior to existing clinical indicators, such as LDH and stage. CONCLUSIONS: Our findings suggest that the 5hmC modifications in cfDNA at the time before R-CHOP treatment are associated with treatment response and that 5hmC-Seal may potentially serve as a clinical-applicable, minimally invasive approach to predict R-CHOP treatment response for DLBCL patients.


Assuntos
5-Metilcitosina/análogos & derivados , Protocolos de Quimioterapia Combinada Antineoplásica/metabolismo , Ácidos Nucleicos Livres/metabolismo , Linfoma Difuso de Grandes Células B/tratamento farmacológico , 5-Metilcitosina/sangue , 5-Metilcitosina/metabolismo , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Biomarcadores Farmacológicos/metabolismo , Estudos de Coortes , Ciclofosfamida/metabolismo , Ciclofosfamida/uso terapêutico , Desmetilação do DNA/efeitos dos fármacos , Doxorrubicina/metabolismo , Doxorrubicina/uso terapêutico , Feminino , Humanos , Modelos Logísticos , Linfoma Difuso de Grandes Células B/diagnóstico , Linfoma Difuso de Grandes Células B/metabolismo , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Prednisona/metabolismo , Prednisona/uso terapêutico , Rituximab/metabolismo , Rituximab/uso terapêutico , Sensibilidade e Especificidade , Vincristina/metabolismo , Vincristina/uso terapêutico
11.
Toxicol In Vitro ; 72: 105075, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33388378

RESUMO

Environmental exposure to arsenic remains a worldwide public health challenge. Oxidative stress and aberrant DNA methylation are both characteristics of arsenic toxicology; however, the relationship between these is not well understood. Ten-eleven translocation (TET1, TET2 and TET3), which is the conversion of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC), plays a central role in the DNA demethylation process. Further, it can prevent cytosine-phosphate-guanine (CpG) islands from developing abnormal hypermethylation under oxidative stress. Here, we observed that NaAsO2 could induce oxidative stress in human bronchial epithelial (HBE) cells. This was accompanied by an inhibition of TET-mediated DNA demethylation. Subsequent results showed that TET1 and TET2 siRNA led to further inhibition of genome 5hmC and a higher level of oxidative stress in NaAsO2-treated HBE cells. Conversely, l-ascorbic acid enhanced TET proteins and effectively upregulated 5hmC, which antagonized the NaAsO2-induced oxidative stress. Additionally, the TETs positively regulated the promoter methylation of the antioxidant genes 8-oxoguanine DNA glycosylase (OGG1) and glutathione S-transferase Pi 1 (GSTP1). Taken together, the results indicate that arsenic induced the inhibition of TET-mediated DNA demethylation, which induced promoter hypermethylation, inhibiting the expression of the OGG1 and GSTP1, and increasing oxidative stress in lung cells in vitro. l-ascorbic acid effectively alleviated arsenic-induced oxidative stress by restoring TET function.


Assuntos
Arsenitos/toxicidade , Desmetilação do DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/genética , Dioxigenases/genética , Oxigenases de Função Mista/genética , Proteínas Proto-Oncogênicas/genética , Compostos de Sódio/toxicidade , Antioxidantes/farmacologia , Ácido Ascórbico/farmacologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , DNA Glicosilases/genética , Metilação de DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Dioxigenases/metabolismo , Glutationa S-Transferase pi/genética , Humanos , Oxigenases de Função Mista/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Regiões Promotoras Genéticas/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , RNA Interferente Pequeno/genética , Espécies Reativas de Oxigênio/metabolismo
12.
Clin Transl Oncol ; 23(4): 750-756, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32737836

RESUMO

BACKGROUND: Colorectal cancer (CRC) represents an important neoplasm with high mortality. Although PD-L1/PD-1 system-based immunotherapy has benefits for a certain type of CRC, many efforts should be made to enhance the responses to anti-PD-1/PD-L1 drugs. DNA methylation has been critically implicated in the regulation of tumor immunity. Here, we examined the effects of the natural alkaloid oxymatrine on PD-L1 expression in CRC cells and to elucidate the underlying mechanism. METHODS: Human CRC SW620 and HCT116 cells were treated with interferon γ (IFNγ) and/or oxymatrine. Cell viability was determined using MTT assays. PD-L1 expression was detected by real-time PCR and Western blot analyses. DNA demethylase activity was measured using kits. RESULTS: Oxymatrine did not apparently affect the viability of normal human intestinal epithelial cells. IFNγ at 20 ng/ml increased the viability of CRC cells, but oxymatrine concentration-dependently reduced the viability in the absence or presence of IFNγ. IFNγ increased the mRNA and protein expression of PD-L1 in the two cell lines, but oxymatrine significantly abolished IFNγ-elevated PD-L1 levels at both mRNA and protein levels. Furthermore, DNA demethylase activity was remarkably increased in IFNγ-treated CRC cells, which was abolished by oxymatrine concentration-dependently. In addition, DNA methyltransferase inhibitor 5-azacytidine considerably abrogated oxymatrine-induced downregulation of PD-L1 mRNA and protein levels in IFNγ-stimulated CRC cells. CONCLUSION: Oxymatrine suppressed viability and reduced PD-L1 expression in IFNγ-stimulated CRC cells, which was attributed to enhanced DNA demethylation. Our current discoveries suggested oxymatrine as an epigenetic modulatory agent for immunotherapy against CRC via PD-1/PD-L1 blockade.


Assuntos
Alcaloides/farmacologia , Antígeno B7-H1/metabolismo , Neoplasias Colorretais/metabolismo , Desmetilação do DNA/efeitos dos fármacos , Quinolizinas/farmacologia , Azacitidina/farmacologia , Antígeno B7-H1/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Células Epiteliais/efeitos dos fármacos , Células HCT116 , Humanos , Inibidores de Checkpoint Imunológico , Interferon gama/antagonistas & inibidores , Interferon gama/farmacologia , RNA Mensageiro/metabolismo
13.
J Neurochem ; 157(6): 1759-1773, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-32219848

RESUMO

Ascorbic acid (vitamin C) is critical for Schwann cells to myelinate peripheral nerve axons during development and remyelination after injury. However, its exact mechanism remains elusive. Vitamin C is a dietary nutrient that was recently discovered to promote active DNA demethylation. Schwann cell myelination is characterized by global DNA demethylation in vivo and may therefore be regulated by vitamin C. We found that vitamin C induces a massive transcriptomic shift (n = 3,848 genes) in primary cultured Schwann cells while simultaneously producing a global increase in genomic 5-hydroxymethylcytosine (5hmC), a DNA demethylation intermediate which regulates transcription. Vitamin C up-regulates 10 pro-myelinating genes which exhibit elevated 5hmC content in both the promoter and gene body regions of these loci following treatment. Using a mouse model of human vitamin C metabolism, we found that maternal dietary vitamin C deficiency causes peripheral nerve hypomyelination throughout early development in resulting offspring. Additionally, dietary vitamin C intake regulates the expression of myelin-related proteins such as periaxin (PRX) and myelin basic protein (MBP) during development and remyelination after injury in mice. Taken together, these results suggest that vitamin C cooperatively promotes myelination through 1) increased DNA demethylation and transcription of pro-myelinating genes, and 2) its known role in stabilizing collagen helices to form the basal lamina that is necessary for myelination.


Assuntos
Ácido Ascórbico/administração & dosagem , Ácido Ascórbico/metabolismo , Desmetilação do DNA/efeitos dos fármacos , Proteínas da Mielina/metabolismo , Bainha de Mielina/metabolismo , Células de Schwann/fisiologia , Animais , Ácido Ascórbico/genética , Deficiência de Ácido Ascórbico/tratamento farmacológico , Deficiência de Ácido Ascórbico/genética , Deficiência de Ácido Ascórbico/metabolismo , Células Cultivadas , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas da Mielina/genética , Bainha de Mielina/efeitos dos fármacos , Bainha de Mielina/genética , Ratos Endogâmicos F344 , Células de Schwann/efeitos dos fármacos , Neuropatia Ciática/tratamento farmacológico , Neuropatia Ciática/genética , Neuropatia Ciática/metabolismo
14.
Cells ; 11(1)2021 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-35011646

RESUMO

Metal ions in the induction medium are essential ingredients allowing green plant regeneration. For instance, Cu(II) and Ag(I) ions may affect the mitochondrial electron transport chain, influencing the Yang cycle and synthesis of S-adenosyl-L-methionine, the prominent donor of the methylation group for all cellular compounds, including cytosines. If the ion concentrations are not balanced, they can interfere with the proper flow of electrons in the respiratory chain and ATP production. Under oxidative stress, methylated cytosines might be subjected to mutations impacting green plant regeneration efficiency. Varying Cu(II) and Ag(I) concentrations in the induction medium and time of anther culture, nine trials of anther culture-derived regenerants of triticale were derived. The methylation-sensitive AFLP approach quantitative characteristics of tissue culture-induced variation, including sequence variation, DNA demethylation, and DNA de novo methylation for all symmetric-CG, CHG, and asymmetric-CHH sequence contexts, were evaluated for all trials. In addition, the implementation of mediation analysis allowed evaluating relationships between factors influencing green plant regeneration efficiency. It was demonstrated that Cu(II) ions mediated relationships between: (1) de novo methylation in the CHH context and sequence variation in the CHH, (2) sequence variation in CHH and green plant regeneration efficiency, (3) de novo methylation in CHH sequences and green plant regeneration, (4) between sequence variation in the CHG context, and green plant regeneration efficiency. Cu(II) ions were not a mediator between de novo methylation in the CG context and green plant regeneration. The latter relationship was mediated by sequence variation in the CG context. On the other hand, we failed to identify any mediating action of Ag(I) ions or the moderating role of time. Furthermore, demethylation in any sequence context seems not to participate in any relationships leading to green plant regeneration, sequence variation, and the involvement of Cu(II) or Ag(I) as mediators.


Assuntos
Cobre/farmacologia , Meios de Cultura/química , Metilação de DNA/genética , Regeneração/genética , Triticale/genética , Triticale/fisiologia , Análise do Polimorfismo de Comprimento de Fragmentos Amplificados , Sequência de Bases , Desmetilação do DNA/efeitos dos fármacos , Metilação de DNA/efeitos dos fármacos , Íons , Regeneração/efeitos dos fármacos , Triticale/efeitos dos fármacos
15.
Cell Rep ; 33(9): 108452, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33264617

RESUMO

Plasma cells provide high-affinity antibodies against invading pathogens. Although transcriptional and epigenetic mechanisms have been extensively studied for plasma cell differentiation, how these mechanisms respond to environmental cues remains largely unexplored. In this study, we show that ascorbic acid (vitamin C), an essential nutrient, is able to promote plasma cell differentiation and humoral immune response by enhancing TET2/3-mediated DNA demethylation. Ascorbic acid treatment during B cell activation has persistent effects on later plasma cell differentiation by predisposing germinal center B cells toward the plasma cell lineage. Conversely, ascorbic acid deficiency in vivo blocks plasma cell differentiation and attenuates the humoral immune response following antigen immunization. We further demonstrate that such effects of ascorbic acid on plasma cell differentiation require DNA methylcytosine oxidases TET2 and TET3. Our study thus reveals a previously uncharacterized link between essential nutrients and epigenetic regulation of plasma cell differentiation and humoral immune response.


Assuntos
Ácido Ascórbico/uso terapêutico , Desmetilação do DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/efeitos dos fármacos , Dioxigenases/efeitos dos fármacos , Plasma/metabolismo , Animais , Ácido Ascórbico/farmacologia , Humanos , Camundongos
16.
Aging (Albany NY) ; 13(1): 578-597, 2020 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-33257596

RESUMO

Aging impairs the IGF-I signaling of bone marrow mesenchymal stem cells (bmMSCs), but the mechanism is unclear. Here, we found that the ability to auto-phosphorylate IGF-I receptor (IGF-IR) in response to IGF-I was decreased in the bmMSCs of aged donors. Conversely, data showed that decorin (DCN) expression was prominently increased in aged bmMSCs, and that under IGF-I treatment, DCN knockdown in serum-starved aged bmMSCs potentiated their mitogenic activity and IGF-IR auto-phosphorylation, whereas DCN overexpression in serum-starved adult bmMSCs decreased both activities. Co-immunoprecipitation assays suggested that IGF-I and DCN bound to IGF-IR in a competitive manner. Online MethPrimer predicted 4 CpG islands (CGIs) in the introns of DCN gene. RT-qPCR and bisulfite sequencing showed that dimethyloxalylglycine, an inhibitor of DNA demethylation, increased DCN mRNA expression and CGI-I methylation in adult bmMSCs, whereas 5-aza-2'-deoxycytidine, a DNA methylation inhibitor, decreased DCN mRNA expression and CGI-I methylation in aged bmMSCs, and ultimately enhanced the proliferation of serum-starved aged bmMSCs under IGF-I stimulation. Thus, IGF-IR could be the prime target of aging in down-regulating the IGF-I signaling of bmMSCs, where DCN could be a critical mediator.


Assuntos
Envelhecimento/genética , Células da Medula Óssea/metabolismo , Decorina/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Fator de Crescimento Insulin-Like I/metabolismo , Células-Tronco Mesenquimais/metabolismo , Receptor IGF Tipo 1/metabolismo , Adulto , Idoso , Aminoácidos Dicarboxílicos/farmacologia , Células da Medula Óssea/efeitos dos fármacos , Desmetilação do DNA/efeitos dos fármacos , Metilação de DNA/efeitos dos fármacos , Decitabina/farmacologia , Decorina/efeitos dos fármacos , Decorina/metabolismo , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Pessoa de Meia-Idade , Fosforilação , RNA Mensageiro/metabolismo , Transdução de Sinais
17.
Endocrinology ; 161(11)2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32865566

RESUMO

Thyroid hormone (T3) plays pivotal roles in vertebrate development, acting via nuclear T3 receptors (TRs) that regulate gene transcription by promoting post-translational modifications to histones. Methylation of cytosine residues in deoxyribonucleic acid (DNA) also modulates gene transcription, and our recent finding of predominant DNA demethylation in the brain of Xenopus tadpoles at metamorphosis, a T3-dependent developmental process, caused us to hypothesize that T3 induces these changes in vivo. Treatment of premetamorphic tadpoles with T3 for 24 or 48 hours increased immunoreactivity in several brain regions for the DNA demethylation intermediates 5-hydroxymethylcytosine (5-hmC) and 5-carboxylcytosine, and the methylcytosine dioxygenase ten-eleven translocation 3 (TET3). Thyroid hormone treatment induced locus-specific DNA demethylation in proximity to known T3 response elements within the DNA methyltransferase 3a and Krüppel-like factor 9 genes, analyzed by 5-hmC immunoprecipitation and methylation sensitive restriction enzyme digest. Chromatin-immunoprecipitation (ChIP) assay showed that T3 induced TET3 recruitment to these loci. Furthermore, the messenger ribonucleic acid for several genes encoding DNA demethylation enzymes were induced by T3 in a time-dependent manner in tadpole brain. A TR ChIP-sequencing experiment identified putative TR binding sites at several of these genes, and we provide multiple lines of evidence to support that tet2 contains a bona fide T3 response element. Our findings show that T3 can promote DNA demethylation in developing tadpole brain, in part by promoting TET3 recruitment to discrete genomic regions, and by inducing genes that encode DNA demethylation enzymes.


Assuntos
Encéfalo/efeitos dos fármacos , Desmetilação do DNA/efeitos dos fármacos , Hormônios Tireóideos/farmacologia , Xenopus/embriologia , Animais , Animais Geneticamente Modificados , Encéfalo/embriologia , Encéfalo/metabolismo , Ilhas de CpG/efeitos dos fármacos , Ilhas de CpG/genética , Desmetilação/efeitos dos fármacos , Dioxigenases/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Larva/efeitos dos fármacos , Larva/genética , Larva/crescimento & desenvolvimento , Metamorfose Biológica/efeitos dos fármacos , Metamorfose Biológica/genética , Receptores alfa dos Hormônios Tireóideos/genética , Xenopus/genética , Proteínas de Xenopus/metabolismo
18.
FASEB J ; 34(9): 11474-11487, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32729950

RESUMO

Hypoxia is a key pathogenetic characteristic of chronic kidney disease (CKD). Klotho has renoprotective effect and its expression is commonly suppressed in CKD patients. We showed that chronic hypoxia in unilateral ureteral obstruction model mice is associated with renal Klotho promoter methylation and expression silencing. Administration of low-dose of sodium hydrosulfide (NaHS) effectively ameliorated renal tubulointerstitial fibrosis in the mouse model by demethylating Klotho promoter and restoring its expression. Mechanistically, hypoxia microenvironment in CKD reduced cellular oxygen availability and Fe2+ concentration, and led to impaired activity of ten-eleven translocation (TET), which is critical in maintaining Klotho promoter demethylation status. NaHS treatment greatly improved hypoxia condition, restored TET activity, reversed DNA methylation, and thus, increased Klotho expression. Our results strongly suggested that correcting hypoxia condition to restore TET activity could be a promising therapeutic strategy against CKD.


Assuntos
Desmetilação do DNA/efeitos dos fármacos , Proteínas de Ligação a DNA/genética , Glucuronidase/genética , Rim/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas/genética , Sulfetos/farmacologia , Animais , Linhagem Celular , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Fibrose/genética , Fibrose/prevenção & controle , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Rim/metabolismo , Rim/patologia , Proteínas Klotho , Masculino , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas/metabolismo , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/metabolismo
19.
Drug Resist Updat ; 53: 100715, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32679188

RESUMO

It is well established that multifactorial drug resistance hinders successful cancer treatment. Tumor cell interactions with the tumor microenvironment (TME) are crucial in epithelial-mesenchymal transition (EMT) and multidrug resistance (MDR). TME-induced factors secreted by cancer cells and cancer-associated fibroblasts (CAFs) create an inflammatory microenvironment by recruiting immune cells. CD11b+/Gr-1+ myeloid-derived suppressor cells (MDSCs) and inflammatory tumor associated macrophages (TAMs) are main immune cell types which further enhance chronic inflammation. Chronic inflammation nurtures tumor-initiating/cancer stem-like cells (CSCs), induces both EMT and MDR leading to tumor relapses. Pro-thrombotic microenvironment created by inflammatory cytokines and chemokines from TAMs, MDSCs and CAFs is also involved in EMT and MDR. MDSCs are the most common mediators of immunosuppression and are also involved in resistance to targeted therapies, e.g. BRAF inhibitors and oncolytic viruses-based therapies. Expansion of both cancer and stroma cells causes hypoxia by hypoxia-inducible transcription factors (e.g. HIF-1α) resulting in drug resistance. TME factors induce the expression of transcriptional EMT factors, MDR and metabolic adaptation of cancer cells. Promoters of several ATP-binding cassette (ABC) transporter genes contain binding sites for canonical EMT transcription factors, e.g. ZEB, TWIST and SNAIL. Changes in glycolysis, oxidative phosphorylation and autophagy during EMT also promote MDR. Conclusively, EMT signaling simultaneously increases MDR. Owing to the multifactorial nature of MDR, targeting one mechanism seems to be non-sufficient to overcome resistance. Targeting inflammatory processes by immune modulatory compounds such as mTOR inhibitors, demethylating agents, low-dosed histone deacetylase inhibitors may decrease MDR. Targeting EMT and metabolic adaptation by small molecular inhibitors might also reverse MDR. In this review, we summarize evidence for TME components as causative factors of EMT and anticancer drug resistance.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Fibroblastos Associados a Câncer/efeitos dos fármacos , Fibroblastos Associados a Câncer/imunologia , Fibroblastos Associados a Câncer/metabolismo , Linhagem Celular Tumoral , Desmetilação do DNA/efeitos dos fármacos , Modelos Animais de Doenças , Resistência a Múltiplos Medicamentos/imunologia , Resistencia a Medicamentos Antineoplásicos/imunologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Transição Epitelial-Mesenquimal/imunologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Inibidores de Histona Desacetilases/uso terapêutico , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/metabolismo , Neoplasias/imunologia , Neoplasias/patologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia , Macrófagos Associados a Tumor/efeitos dos fármacos , Macrófagos Associados a Tumor/imunologia , Macrófagos Associados a Tumor/metabolismo
20.
Oxid Med Cell Longev ; 2020: 2543504, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32617134

RESUMO

Curcumin, a natural polyphenol antioxidant extracted from the root of turmeric (Curcuma longa), can induce apoptosis and DNA demethylation in several types of cancer cells. However, the mechanism of its anticancer potentials and DNA demethylation effects and the potential relationships between these outcomes have not been clearly elucidated. In the present study, the effects of curcumin on the proliferation, colony formation, and migration of human gastric cancer cells (hGCCs) were explored. Reactive oxygen species (ROS) levels, mitochondrial damage, DNA damage, and apoptosis of curcumin-treated hGCCs were analyzed. Changes in the expression of several genes related to DNA damage repair, the p53 pathway, cell cycle, and DNA methylation following curcumin treatment were also evaluated. We observed that curcumin inhibited the proliferation, colony formation, and migration of hGCCs in a dose- and time-dependent fashion. A high concentration of curcumin elevated ROS levels and triggered mitochondrial damage, DNA damage, and apoptosis of hGCCs. Further, curcumin-induced DNA demethylation of hGCCs was mediated by the damaged DNA repair-p53-p21/GADD45A-cyclin/CDK-Rb/E2F-DNMT1 axis. We propose that the anticancer effect of curcumin could largely be attributed to its prooxidative effect at high concentrations and ROS elevation in cancer cells. Moreover, we present a novel mechanism by which curcumin induces DNA demethylation of hGCCs, suggesting the need to further investigate the demethylation mechanisms of other DNA hypomethylating drugs.


Assuntos
Curcumina/farmacologia , Dano ao DNA , Desmetilação do DNA/efeitos dos fármacos , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Apoptose/efeitos dos fármacos , Sequência de Bases , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Curcumina/química , Reparo do DNA/efeitos dos fármacos , Genoma Humano , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Modelos Biológicos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ensaio Tumoral de Célula-Tronco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...