Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
Semin Cancer Biol ; 51: 59-67, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29102482

RESUMEN

Over the past few years it has become clear that vitamin C, as a provider of reduced iron, is an essential factor for the function of epigenetic regulators that initiate the demethylation of DNA and histones. Vitamin C deficiency is rare in the general population, but is frequently observed in patients with cancer. Genes encoding epigenetic regulators are often mutated in cancer, underscoring their central roles in carcinogenesis. In hematological cancers, such as acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS), drugs that reverse epigenetic aberrations are now the standard of care. Recent in vitro studies suggest that vitamin C at physiological concentrations, combined with hypomethylating agents may act synergistically to cause DNA demethylation through active and passive mechanisms, respectively. Additionally, several recent studies have renewed interest in the use of pharmacological doses of vitamin C injected intravenously to selectively kill tumor cells. This review will focus on the potential of vitamin C to optimize the outcome of epigenetic therapy in cancer patients and alternatively to act as a therapeutic at high doses.


Asunto(s)
Antioxidantes/uso terapéutico , Ácido Ascórbico/uso terapéutico , Metilación de ADN , Epigénesis Genética , Epigenómica/métodos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias/genética , Animales , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/patología
2.
Clin Epigenetics ; 15(1): 91, 2023 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-37237325

RESUMEN

BACKGROUND: Idiopathic non-clonal cytopenia (ICUS) and clonal cytopenia (CCUS) are common in the elderly population. While these entities have similar clinical presentations with peripheral blood cytopenia and less than 10% bone marrow dysplasia, their malignant potential is different and the biological relationship between these disorders and myeloid neoplasms such as myelodysplastic syndrome (MDS) is not fully understood. Aberrant DNA methylation has previously been described to play a vital role in MDS and acute myeloid leukemia (AML) pathogenesis. In addition, obesity confers a poorer prognosis in MDS with inferior overall survival and a higher rate of AML transformation. In this study, we measured DNA methylation of the promoter for the obesity-regulated gene LEP, encoding leptin, in hematopoietic cells from ICUS, CCUS and MDS patients and healthy controls. We investigated whether LEP promoter methylation is an early event in the development of myeloid neoplasms and whether it is associated with clinical outcome. RESULTS: We found that blood cells of patients with ICUS, CCUS and MDS all have a significantly hypermethylated LEP promoter compared to healthy controls and that LEP hypermethylation is associated with anemia, increased bone marrow blast percentage, and lower plasma leptin levels. MDS patients with a high LEP promoter methylation have a higher risk of progression, shorter progression-free survival, and inferior overall survival. Furthermore, LEP promoter methylation was an independent risk factor for the progression of MDS in a multivariate Cox regression analysis. CONCLUSION: In conclusion, hypermethylation of the LEP promoter is an early and frequent event in myeloid neoplasms and is associated with a worse prognosis.


Asunto(s)
Anemia , Leptina , Leucemia Mieloide Aguda , Síndromes Mielodisplásicos , Anciano , Humanos , Anemia/genética , Hematopoyesis Clonal , Metilación de ADN , Leptina/genética , Leucemia Mieloide Aguda/genética , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/patología , Obesidad/genética
3.
Cancer Res ; 80(12): 2441-2450, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32245794

RESUMEN

The DNA methyltransferase inhibitors (DNMTi) 5-azacytidine and 5-aza-2-deoxycytidine have been approved for the treatment of different types of hematologic malignancies. However, only about 50% of patients respond to treatment. Therefore, a more comprehensive understanding of the molecular changes in patients treated with DNMTi is needed. Here, we examined gene expression profiles in a total of 150 RNA samples from two adult cohorts and one pediatric cohort with hematologic cancers taken before, during, and after treatment with 5-azacytidine (40 patients; 15 nonresponders, 25 responders). Using each patient as their own control, malignant cells showed preferential activation of a subset of evolutionarily young transposable elements (TE), including endogenous retroviral long terminal repeats (LTR), short and long interspersed nuclear elements (SINE and LINE), and the type I IFN pathway in responders, all independent of disease classification. Transfection of eight upregulated LTRs into recipient human cells in culture showed robust and heterogenous activation of six genes in the type I IFN pathway. These results, obtained in diverse hematologic disease entities, show that common targets (TE) activated by the same drug (5-azacytidine) elicit an immune response, which may be important for patient's responses to DNMTi. SIGNIFICANCE: Activation of specific classes of evolutionarily young transposable elements can lead to activation of the innate immune system.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Azacitidina/farmacología , Elementos Transponibles de ADN/efectos de los fármacos , Neoplasias Hematológicas/tratamiento farmacológico , Inmunidad Innata/genética , Anciano , Anciano de 80 o más Años , Antimetabolitos Antineoplásicos/uso terapéutico , Azacitidina/uso terapéutico , Estudios de Cohortes , Elementos Transponibles de ADN/genética , Resistencia a Antineoplásicos/genética , Resistencia a Antineoplásicos/inmunología , Retrovirus Endógenos/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/inmunología , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/inmunología , Humanos , Inmunidad Innata/efectos de los fármacos , Interferón Tipo I/metabolismo , Masculino , Persona de Mediana Edad , Imitación Molecular/inmunología , RNA-Seq , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/inmunología , Regulación hacia Arriba/efectos de los fármacos
4.
Clin Epigenetics ; 11(1): 143, 2019 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-31623675

RESUMEN

BACKGROUND: Patients with haematological malignancies are often vitamin C deficient, and vitamin C is essential for the TET-induced conversion of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC), the first step in active DNA demethylation. Here, we investigate whether oral vitamin C supplementation can correct vitamin C deficiency and affect the 5hmC/5mC ratio in patients with myeloid cancers treated with DNA methyltransferase inhibitors (DNMTis). RESULTS: We conducted a randomized, double-blinded, placebo-controlled pilot trial (NCT02877277) in Danish patients with myeloid cancers performed during 3 cycles of DNMTi-treatment (5-azacytidine, 100 mg/m2/d for 5 days in 28-day cycles) supplemented by oral dose of 500 mg vitamin C (n = 10) or placebo (n = 10) daily during the last 2 cycles. Fourteen patients (70%) were deficient in plasma vitamin C (< 23 µM) and four of the remaining six patients were taking vitamin supplements at inclusion. Global DNA methylation was significantly higher in patients with severe vitamin C deficiency (< 11.4 µM; 4.997 vs 4.656% 5mC relative to deoxyguanosine, 95% CI [0.126, 0.556], P = 0.004). Oral supplementation restored plasma vitamin C levels to the normal range in all patients in the vitamin C arm (mean increase 34.85 ± 7.94 µM, P = 0.0004). We show for the first time that global 5hmC/5mC levels were significantly increased in mononuclear myeloid cells from patients receiving oral vitamin C compared to placebo (0.037% vs - 0.029%, 95% CI [- 0.129, - 0.003], P = 0.041). CONCLUSIONS: Normalization of plasma vitamin C by oral supplementation leads to an increase in the 5hmC/5mC ratio compared to placebo-treated patients and may enhance the biological effects of DNMTis. The clinical efficacy of oral vitamin C supplementation to DNMTis should be investigated in a large randomized, placebo-controlled clinical trial. TRIAL REGISTRATION: ClinicalTrials.gov, NCT02877277 . Registered on 9 August 2016, retrospectively registered.


Asunto(s)
Ácido Ascórbico/administración & dosificación , Azacitidina/administración & dosificación , Metilación de ADN/efectos de los fármacos , Leucemia Mieloide/terapia , Síndromes Mielodisplásicos/terapia , Administración Oral , Anciano , Anciano de 80 o más Años , Ácido Ascórbico/sangre , Ácido Ascórbico/farmacología , Azacitidina/farmacología , Islas de CpG/efectos de los fármacos , Dinamarca , Método Doble Ciego , Epigénesis Genética/efectos de los fármacos , Femenino , Humanos , Leucemia Mieloide/sangre , Leucemia Mieloide/genética , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/sangre , Síndromes Mielodisplásicos/genética , Proyectos Piloto
5.
Oncotarget ; 6(11): 9612-26, 2015 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-25823822

RESUMEN

The hypomethylating agents (HMAs) are standard therapy for patients with higher-risk myelodysplastic syndrome (MDS); however, the majority of the patients will lose their response to HMAs over time due to unknown mechanisms. It has recently been shown that T cell expression of the immunoinhibitory receptor PD-1 is regulated by DNA methylation. In 12 of 27 patients (44%) PD-1 promoter demethylation was observed in sorted peripheral blood T cells isolated over consecutive cycles of treatment with 5-azacytidine (5-aza). The PD-1 promoter demethylation correlated with an increase in PD-1 expression. Moreover, demethylation of the PD-1 promoter correlated with a significantly worse overall response rate (8% vs. 60%, p = 0.014), and a trend towards a shorter overall survival (p = 0.11) was observed. A significantly higher baseline methylation level of the PD-1 promoter was observed in T cells of non-responding patients compared to healthy controls (p = 0.023). Accordingly, in addition to their beneficial function, HMAs induce PD-1 expression on T cells in the MDS microenvironment, thereby likely hampering the immune response against the MDS blasts. Thus, we suggest that activation of the PD-1 checkpoint during HMA treatment can be a possible resistance mechanism, which may be overcome by combination therapy with a PD-1 pathway inhibitor.


Asunto(s)
Antimetabolitos/farmacología , Azacitidina/farmacología , Metilación de ADN/efectos de los fármacos , Síndromes Mielodisplásicos/tratamiento farmacológico , Receptor de Muerte Celular Programada 1/genética , Regiones Promotoras Genéticas/genética , Subgrupos de Linfocitos T/metabolismo , Anciano , Anciano de 80 o más Años , Células Sanguíneas/efectos de los fármacos , Células Sanguíneas/metabolismo , Resistencia a Medicamentos , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mielomonocítica Crónica/tratamiento farmacológico , Leucemia Mielomonocítica Crónica/genética , Leucemia Mielomonocítica Crónica/metabolismo , Activación de Linfocitos , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/metabolismo , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Receptor de Muerte Celular Programada 1/biosíntesis , Receptor de Muerte Celular Programada 1/fisiología , Regiones Promotoras Genéticas/efectos de los fármacos , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , Subgrupos de Linfocitos T/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA