Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Oncol ; 13: 1334112, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38304034

RESUMEN

Background: Bladder cancer (BLCA) is a common and deadly disease that results in a reduced quality of life for the patients and a significant economic burden on society. A better understanding of tumorigenesis is needed to improve clinical outcomes. Recent evidence places the RNA modification m1A and its regulatory proteins TRMT6/TRMT61A and ALKBH3 in BLCA pathogenesis. Methods: TRMT6/TRMT61A, ALKBH1, and ALKBH3 expression was examined in human BLCA cell lines and a normal urinary tract epithelium cell line through qRT-PCR and western blot analysis. Prestoblue Cell Viability Reagent, wound-healing assay, and live-cell imaging-based cell displacement analysis, were conducted to assess proliferation, migration, and displacement of this BLCA cell line panel. Cell survival was assessed after inducing cellular stress and activating the unfolded protein response (UPR) with tunicamycin. Moreover, siRNA-mediated gene silencing in two BLCA cell lines (5637 and HT1197) was conducted to investigate the biological roles of TRMT6/TRMT61A. Results: Heterogeneous morphology, proliferation, displacement, tunicamycin sensitivity, and expression levels of m1A regulators were observed among the panel of cell lines examined. In general, TRMT61A expression was increased in BLCA cell lines when compared to SV-HUC-1. Depletion of TRMT6/TRMT61A reduced proliferation capacity in both 5637 and HT1197 cell lines. The average cell displacement of 5637 was also reduced upon TRMT6/TRMT61A depletion. Interestingly, TRMT6/TRMT61A depletion decreased mRNA expression of targets associated with the ATF6-branch of the UPR in 5637 but not in HT1197. Moreover, cell survival after induction of cellular stress was compromised after TRMT6/TRMT61A knockdown in 5637 but not in HT1197 cells. Conclusion: The findings suggest that TRMT6/TRMT61A plays an oncogenic role in BLCA and is involved in desensitizing BLCA cells against cellular stress. Further investigation into the regulation of TRMT6/TRMT61A expression and its impact on cellular stress tolerance may provide insights for future BLCA treatment.

2.
Front Mol Biosci ; 9: 887686, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35923465

RESUMEN

Background: Bladder cancer (BLCA) is one of the most common cancer types worldwide. The disease is responsible for about 200,000 deaths annually, thus improved diagnostics and therapy is needed. A large body of evidence reveal that small RNAs of less than 40 nucleotides may act as tumor suppressors, oncogenes, and disease biomarkers, with a major focus on microRNAs. However, the role of other families of small RNAs is not yet deciphered. Recent results suggest that small RNAs and their modification status, play a role in BLCA development and are promising biomarkers due to their high abundance in the exomes and body fluids (including urine). Moreover, free modified nucleosides have been detected at elevated levels from the urine of BLCA patients. A genome-wide view of small RNAs, and their modifications, will help pinpoint the molecules that could be used as biomarker or has important biology in BLCA development. Methods: BLCA tumor tissue specimens were obtained from 12 patients undergoing transurethral resection of non-muscle invasive papillary urothelial carcinomas. Genome-wide profiling of small RNAs less than 40 bases long was performed by a modified protocol with TGIRT (thermostable group II reverse transcriptase) to identify novel small RNAs and their modification status. Results: Comprehensive analysis identified not only microRNAs. Intriguingly, 57 ± 15% (mean ± S.D.) of sequencing reads mapped to non-microRNA-small RNAs including tRNA-derived fragments (tRFs), ribosomal RNA-derived fragments (rRFs) and YRNA-derived fragments (YRFs). Misincorporation (mismatch) sites identified potential base modification positions on the small RNAs, especially on tRFs, corresponding to m1A (N1-methyladenosine), m1G (N1-methylguanosine) and m2 2G (N2, N2-dimethylguanosine). We also detected mismatch sites on rRFs corresponding to known modifications on 28 and 18S rRNA. Conclusion: We found abundant non-microRNA-small RNAs in BLCA tumor samples. Small RNAs, especially tRFs and rRFs, contain modifications that can be captured as mismatch by TGIRT sequencing. Both the modifications and the non-microRNA-small RNAs should be explored as a biomarker for BLCA detection or follow-up.

3.
Nat Commun ; 13(1): 2165, 2022 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-35444240

RESUMEN

RNA modifications are important regulatory elements of RNA functions. However, most genome-wide mapping of RNA modifications has focused on messenger RNAs and transfer RNAs, but such datasets have been lacking for small RNAs. Here we mapped N1-methyladenosine (m1A) in the cellular small RNA space. Benchmarked with synthetic m1A RNAs, our workflow identified specific groups of m1A-containing small RNAs, which are otherwise disproportionally under-represented. In particular, 22-nucleotides long 3' tRNA-fragments are highly enriched for TRMT6/61A-dependent m1A located within the seed region. TRMT6/61A-dependent m1A negatively affects gene silencing by tRF-3s. In urothelial carcinoma of the bladder, where TRMT6/61A is over-expressed, higher m1A modification on tRFs is detected, correlated with a dysregulation of tRF targetome. Lastly, TRMT6/61A regulates tRF-3 targets involved in unfolded protein response. Together, our results reveal a mechanism of regulating gene expression via base modification of small RNA.


Asunto(s)
Carcinoma de Células Transicionales , Neoplasias de la Vejiga Urinaria , Carcinoma de Células Transicionales/genética , Femenino , Silenciador del Gen , Humanos , Masculino , Metilación , ARN/metabolismo , ARN de Transferencia/genética , ARN de Transferencia/metabolismo , Respuesta de Proteína Desplegada/genética , Neoplasias de la Vejiga Urinaria/genética
4.
Int J Colorectal Dis ; 37(3): 597-605, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34882271

RESUMEN

PURPOSE: Children with constipation and suspected Hirschsprung's disease are referred for rectal biopsy. Since this is an invasive procedure, appropriate indications should be applied to minimize the number of "unnecessary" biopsies. METHODS: We reviewed all constipated children who underwent a rectal biopsy to diagnose a possible Hirschsprung's disease at a tertiary referral hospital over a 6-year period (2013-2018). We registered clinical and demographic factors in these children and conducted correlation and multivariate regression analysis to evaluate the relation between these factors and a diagnosis of Hirschsprung's disease. RESULTS: We identified 225 children, aged 0-17 years. In total, Hirschsprung's disease was diagnosed in only 49/225 (22%). Among the 49 children with Hirschsprung's disease, 29 (59%) were diagnosed in the neonatal period. Among girls, HD was confirmed in only 10/101 (10%) children, and only 1 of these 10 girls was older than 6 months at the time of the biopsy. The following factors correlated significantly with Hirschsprung's disease diagnosis in children older than 1 month: "male sex", "failure to thrive", "gross abdominal distention plus vomiting" and "fulfils the Rome 4 criteria for functional constipation". CONCLUSION: In children referred for rectal biopsy, the factors most indicative of Hirschsprung's disease were "male sex", "failure to thrive", "gross abdominal distention plus vomiting" and "fulfils the Rome 4 criteria for functional constipation". Notably, the prevalence of Hirschsprung's disease decreased with the increasing age of the children. Girls referred for a biopsy rarely had Hirschsprung's disease, especially those older than 1 month.


Asunto(s)
Enfermedad de Hirschsprung , Recto , Adolescente , Biopsia , Niño , Preescolar , Estreñimiento/diagnóstico , Estreñimiento/etiología , Femenino , Enfermedad de Hirschsprung/complicaciones , Enfermedad de Hirschsprung/diagnóstico , Humanos , Lactante , Recién Nacido , Masculino , Recto/patología , Vómitos
5.
Clin Epigenetics ; 11(1): 135, 2019 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-31519213

RESUMEN

Hirschsprung disease is a neurocristopathy, characterized by aganglionosis in the distal bowel. It is caused by failure of the enteric nervous system progenitors to migrate, proliferate, and differentiate in the gut. Development of an enteric nervous system is a tightly regulated process. Both the neural crest cells and the surrounding environment are regulated by different genes, signaling pathways, and morphogens. For this process to be successful, the timing of gene expression is crucial. Hence, alterations in expression of genes specific for the enteric nervous system may contribute to the pathogenesis of Hirschsprung's disease. Several epigenetic mechanisms contribute to regulate gene expression, such as modifications of DNA and RNA, histone modifications, and microRNAs. Here, we review the current knowledge of epigenetic and epitranscriptomic regulation in the development of the enteric nervous system and its potential significance for the pathogenesis of Hirschsprung's disease. We also discuss possible future therapies and how targeting epigenetic and epitranscriptomic mechanisms may open new avenues for novel treatment.


Asunto(s)
Epigénesis Genética , Enfermedad de Hirschsprung/genética , Metilación de ADN , Regulación de la Expresión Génica , Enfermedad de Hirschsprung/terapia , Humanos , MicroARNs/genética
6.
Cell Physiol Biochem ; 38(1): 173-84, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26765775

RESUMEN

BACKGROUND/AIMS: ALKBH1, an AlkB homologue in the 2-oxoglutarate and Fe2+ dependent hydroxylase family, is a histone dioxygenase that removes methyl groups from histone H2A. Studies of transgenic mice lacking Alkbh1 reveal that most Alkbh1-/- embryos die during embryonic development. Embryonic stem cells (ESCs) derived from these mice have prolonged expression of pluripotency markers and delayed induction of genes involved in neural differentiation, indicating that ALKBH1 is involved in regulation of pluripotency and differentiation. The aim of this study was to further investigate the role ALKBH1 in early development. METHODS: Double-filter methods for nitrocellulose-filter binding, dot blot, enzyme-linked immunosorbent assay (ELISA), immonocytochemistry, cell culture and differentiation of mouse ESCs, Co-IP and miRNA analysis. RESULTS: We found that SOX2 and NANOG bind the ALKBH1 promoter, and we identified protein-protein interactions between ALKBH1 and these core transcription factors of the pluripotency network. Furthermore, lack of ALKBH1 affected the expression of developmentally important miRNAs, which are involved in the regulation of NANOG, SOX2 and neural differentiation. CONCLUSION: Our results suggest that ALKBH1 interacts with the core transcriptional pluripotency network of ESCs and is involved in regulation of pluripotency and differentiation.


Asunto(s)
ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Redes Reguladoras de Genes , Factores de Transcripción/metabolismo , Regiones no Traducidas 5' , Histona H2a Dioxigenasa, Homólogo 1 de AlkB , Animales , Secuencia de Bases , Sitios de Unión , Diferenciación Celular , Línea Celular , ADN-(Sitio Apurínico o Apirimidínico) Liasa/deficiencia , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Técnicas de Inactivación de Genes , Proteínas de Homeodominio/química , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , MicroARNs/metabolismo , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/química , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Regiones Promotoras Genéticas , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Factores de Transcripción SOXB1/química , Factores de Transcripción SOXB1/metabolismo , Factores de Transcripción/química
7.
J Mol Cell Biol ; 7(6): 494-504, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26003568

RESUMEN

The DNA repair enzyme AlkB was identified in E. coli more than three decades ago. Since then, nine mammalian homologs, all members of the superfamily of alpha-ketoglutarate and Fe(II)-dependent dioxygenases, have been identified (designated ALKBH1-8 and FTO). While E. coli AlkB serves as a DNA repair enzyme, only two mammalian homologs have been confirmed to repair DNA in vivo. The other mammalian homologs have remarkably diverse substrate specificities and biological functions. Substrates recognized by the different AlkB homologs comprise erroneous methyl- and etheno adducts in DNA, unique wobble uridine modifications in certain tRNAs, methylated adenines in mRNA, and methylated lysines on proteins. The phenotypes of organisms lacking or overexpressing individual AlkB homologs include obesity, severe sensitivity to inflammation, infertility, growth retardation, and multiple malformations. Here we review the present knowledge of the mammalian AlkB homologs and their implications for human disease and development.


Asunto(s)
Dioxigenasas/química , Proteínas de Escherichia coli/química , Ácidos Cetoglutáricos/química , Oxigenasas de Función Mixta/química , ARN de Transferencia/metabolismo , Animales , Biología Computacional , Metilación de ADN , Reparación del ADN/fisiología , Dioxigenasas/clasificación , Dioxigenasas/metabolismo , Proteínas de Escherichia coli/clasificación , Proteínas de Escherichia coli/metabolismo , Humanos , Ratones , Oxigenasas de Función Mixta/clasificación , Oxigenasas de Función Mixta/metabolismo , Filogenia , Procesamiento Proteico-Postraduccional , Procesamiento Postranscripcional del ARN , Homología de Secuencia de Aminoácido , Especificidad por Sustrato
8.
Stem Cell Res ; 11(3): 1264-71, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24041806

RESUMEN

Huntington's disease (HD) is a neurodegenerative disorder caused by an expanded trinucleotide CAG repeat in the Huntingtin (Htt) gene. The molecular basis for the development and progression of HD is currently poorly understood. However, different DNA repair pathways have been implicated in both somatic expansion and disease progression. Embryonic stem cells provide a remarkable in vitro system to study HD and could have implications for understanding disease development and for therapeutic treatment. Here, we derive pluripotent stem cells from the mouse R6/1 HD model and demonstrate that repeated exposure to genotoxic agents inducing oxidative DNA damage gave a significant and dose dependent increase in somatic triplet expansion. Further investigation into specific steps of DNA repair revealed impaired double stranded break repair in exposed R6/1 cells, accompanied by the induction of apoptosis. We also found that differentiation status, and consequently DNA repair efficiency influenced somatic expansion. Our data underscore the importance of DNA damage and repair for the stability of the HD triplet in pluripotent stem cells.


Asunto(s)
ADN/metabolismo , Células Madre Embrionarias/citología , Estrés Oxidativo , Animales , Apoptosis/efectos de los fármacos , Diferenciación Celular , Reparación del ADN , Modelos Animales de Enfermedad , Células Madre Embrionarias/metabolismo , Genotipo , Proteína Huntingtina , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Peróxido de Hidrógeno/toxicidad , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Estrés Oxidativo/efectos de los fármacos , Expansión de Repetición de Trinucleótido/efectos de los fármacos
9.
Mol Neurobiol ; 47(3): 1093-102, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23361256

RESUMEN

The human genome is under continuous attack by a plethora of harmful agents. Without the development of several dedicated DNA repair pathways, the genome would have been destroyed and cell death, inevitable. However, while DNA repair enzymes generally maintain the integrity of the whole genome by properly repairing mutagenic and cytotoxic intermediates, there are cases in which the DNA repair machinery is implicated in causing disease rather than protecting against it. One case is the instability of gene-specific trinucleotides, the causative mutations of numerous disorders including Huntington's disease. The DNA repair proteins induce mutations that are different from the genome-wide mutations that arise in the absence of repair enzymes; they occur at definite loci, they occur in specific tissues during development, and they are age-dependent. These latter characteristics make pluripotent stem cells a suitable model system for triplet repeat expansion disorders. Pluripotent stem cells can be kept in culture for a prolonged period of time and can easily be differentiated into any tissue, e.g., cells along the neural lineage. Here, we review the role of DNA repair proteins in the process of triplet repeat instability in Huntington's disease and also the potential use of pluripotent stem cells to investigate neurodegenerative disorders.


Asunto(s)
Reparación del ADN/genética , Enfermedad de Huntington/genética , Animales , Daño del ADN , Humanos , Mamíferos/metabolismo , Células Madre Pluripotentes/citología , Expansión de Repetición de Trinucleótido/genética
10.
Stem Cells ; 30(12): 2672-82, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22961808

RESUMEN

AlkB homolog 1 (ALKBH1) is one of nine members of the family of mammalian AlkB homologs. Most Alkbh1(-/-) mice die during embryonic development, and survivors are characterized by defects in tissues originating from the ectodermal lineage. In this study, we show that deletion of Alkbh1 prolonged the expression of pluripotency markers in embryonic stem cells and delayed the induction of genes involved in early differentiation. In vitro differentiation to neural progenitor cells (NPCs) displayed an increased rate of apoptosis in the Alkbh1(-/-) NPCs when compared with wild-type cells. Whole-genome expression analysis and chromatin immunoprecipitation revealed that ALKBH1 regulates both directly and indirectly, a subset of genes required for neural development. Furthermore, our in vitro enzyme activity assays demonstrate that ALKBH1 is a histone dioxygenase that acts specifically on histone H2A. Mass spectrometric analysis demonstrated that histone H2A from Alkbh1(-/-) mice are improperly methylated. Our results suggest that ALKBH1 is involved in neural development by modifying the methylation status of histone H2A.


Asunto(s)
ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/enzimología , Histonas/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/enzimología , Histona H2a Dioxigenasa, Homólogo 1 de AlkB , Animales , Apoptosis/genética , Apoptosis/fisiología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Núcleo Celular/enzimología , Metilación de ADN , ADN-(Sitio Apurínico o Apirimidínico) Liasa/deficiencia , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Epigenómica , Histonas/genética , Ratones , Análisis por Micromatrices , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/enzimología , Transfección
11.
Nat Protoc ; 7(2): 340-50, 2012 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-22281869

RESUMEN

We describe a method for the efficient and selective identification of DNA containing the 5-hydroxymethylcytosine (5-hmC) modification. This protocol takes advantage of two proteins: T4 ß-glucosyltransferase (ß-gt), which converts 5-hmC to ß-glucosyl-5-hmC (ß-glu-5-hmC), and J-binding protein 1 (JBP1), which specifically recognizes and binds to ß-glu-5-hmC. We describe the steps necessary to purify JBP1 and modify this protein such that it can be fixed to magnetic beads. Thereafter, we detail how to use the JBP1 magnetic beads to obtain DNA that is enriched with 5-hmC. This method is likely to produce results similar to those of other 5-hmC pull-down assays; however, all necessary components for the completion of this protocol are readily available or can be easily and rapidly synthesized using basic molecular biology techniques. This protocol can be completed in less than 2 weeks and allows the user to isolate 5-hmC-containing genomic DNA that is suitable for analysis by quantitative PCR (qPCR), sequencing, microarray and other molecular biology assays.


Asunto(s)
Citosina/análogos & derivados , Proteínas de Unión al ADN/química , ADN/metabolismo , Proteínas de Escherichia coli/química , 5-Metilcitosina/análogos & derivados , Citosina/análisis , ADN/química , Proteínas de Unión al ADN/aislamiento & purificación , Escherichia coli/genética , Proteínas de Escherichia coli/aislamiento & purificación , Glucosiltransferasas/química , Fenómenos Magnéticos , Microesferas
12.
PLoS One ; 5(11): e13827, 2010 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-21072209

RESUMEN

BACKGROUND: Escherichia coli AlkB is a 2-oxoglutarate- and iron-dependent dioxygenase that reverses alkylated DNA damage by oxidative demethylation. Mouse AlkB homolog 1 (Alkbh1) is one of eight members of the newly discovered family of mammalian dioxygenases. METHODS AND FINDINGS: In the present study we show non-Mendelian inheritance of the Alkbh1 targeted allele in mice. Both Alkbh1(-/-) and heterozygous Alkbh1(+/-) offspring are born at a greatly reduced frequency. Additionally, the sex-ratio is considerably skewed against female offspring, with one female born for every three to four males. Most mechanisms that cause segregation distortion, act in the male gametes and affect male fertility. The skewing of the sexes appears to be of paternal origin, and might be set in the pachythene stage of meiosis during spermatogenesis, in which Alkbh1 is upregulated more than 10-fold. In testes, apoptotic spermatids were revealed in 5-10% of the tubules in Alkbh1(-/-) adults. The deficiency of Alkbh1 also causes misexpression of Bmp2, 4 and 7 at E11.5 during embryonic development. This is consistent with the incompletely penetrant phenotypes observed, particularly recurrent unilateral eye defects and craniofacial malformations. CONCLUSIONS: Genetic and phenotypic assessment suggests that Alkbh1 mediates gene regulation in spermatogenesis, and that Alkbh1 is essential for normal sex-ratio distribution and embryonic development in mice.


Asunto(s)
Dioxigenasas/genética , Embrión no Mamífero/metabolismo , Anomalías del Ojo/genética , Razón de Masculinidad , Factores de Transcripción/genética , Histona H2a Dioxigenasa, Homólogo 1 de AlkB , Animales , Animales Recién Nacidos , Anoftalmos/genética , Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 7/genética , ADN-(Sitio Apurínico o Apirimidínico) Liasa , Dioxigenasas/deficiencia , Embrión no Mamífero/anomalías , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Microftalmía/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espermatogénesis/genética , Testículo/embriología , Testículo/crecimiento & desarrollo , Testículo/metabolismo , Factores de Transcripción/deficiencia
13.
Mol Cell ; 16(1): 107-16, 2004 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-15469826

RESUMEN

Deleterious 1-methyladenine (1-meA) and 3-methylcytosine (3-meC) lesions are introduced into nucleic acids by methylating agents. It was recently demonstrated that the E. coli AlkB protein and a human homolog, hABH3, can demethylate these lesions both in DNA and RNA. To elucidate the biological significance of the RNA repair, we have tested whether such repair can rescue the function of chemically methylated RNA. We demonstrate that a methylation-induced block in translation of an mRNA can be readily relieved by treatment with AlkB and hABH3 prior to translation. Furthermore, we show that chemical methylation of tRNAPhe inhibits aminoacylation and translation, but that the inhibition can be reversed by AlkB and hABH3. AlkB-mediated repair of 1-meA in tRNA was also observed in E. coli in vivo. Our data demonstrate that AlkB proteins can mediate functional recovery of RNA exposed to methylation damage, supporting the notion that RNA repair is important.


Asunto(s)
Proteínas de Escherichia coli/metabolismo , Oxigenasas de Función Mixta/metabolismo , ARN Mensajero/metabolismo , ARN de Transferencia/metabolismo , Histona H2a Dioxigenasa, Homólogo 1 de AlkB , Dioxigenasa Dependiente de Alfa-Cetoglutarato, Homólogo 3 de AlkB , Enzimas Reparadoras del ADN , Proteínas de Unión al ADN/metabolismo , Dioxigenasas , Humanos , Metilación , Análisis de Secuencia de ARN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...