Estudio del perfil de metilación de ADN en pacientes con síndrome progeroide neonatal (síndrome de Wiedemann-Rautenstrauch)

dc.contributor.advisorArboleda Bustos, Gonzalo Humbertospa
dc.contributor.authorBarrera-Torres, Herman Fredyspa
dc.contributor.researchgroupMuerte Celularspa
dc.date.accessioned2024-07-17T13:49:11Z
dc.date.available2024-07-17T13:49:11Z
dc.date.issued2024-07-10
dc.descriptionilustraciones (algunas a color), diagramasspa
dc.description.abstractEl síndrome progeroide neonatal de Wiedemann-Rautenstrauch (SWR) se caracteriza por la manifestación de diversos signos de envejecimiento desde el nacimiento, con una esperanza de vida muy reducida, en promedio de 7 meses, lo que lo distingue de otros síndromes progeroides. La etiología del SWR se ha relacionado con mutaciones en el gen de la subunidad A de la ARN polimerasa III (POLR3A), crucial en la regulación de la expresión de ARN de transferencia (tARN), ARN ribosomal 5S (5SrARN), ARN nucleares pequeños (snARN) y otros, lo que resulta en una disminución de la funcionalidad del complejo ARN polimerasa III (POLR3) y alteraciones en la biogénesis ribosomal y la traducción de proteínas, entre otros procesos. La mutación puntual en el gen POLR3A tiene un impacto considerable en el perfil de metilación de ADN de regiones y genes específicos, ocasionando una expresión anómala de genes y cambios en las dinámicas moleculares. En el caso de una paciente de 6 años (POLR3A: c. 3G>T), se observa hipometilación anormal en regiones del cuerpo del gen, mientras que en una paciente de 25 años (POLR3A: c. 3772 3773 del), se observa una tendencia hacia la hipermetilación en las regiones promotoras y del cuerpo del gen. La metilación anormal de genes debido a la mutación de POLR3A incide principalmente en las proteínas de membrana plasmática, alterando procesos celulares cruciales como la transducción de señales y la transcripción de ADN codificante. Los genes significativamente metilados inducen procesos de senescencia celular. La alteración de la metilación normal en dinucleótidos CpG por el SWR provoca una aceleración o desaceleración en la determinación de la edad biológica mediante el uso de relojes epigenéticos, lo cual es característico de un síndrome progeroide. El estudio del SWR y sus implicaciones epigenéticas proporciona una oportunidad singular para comprender los procesos fisiopatológicos del envejecimiento humano. La identificación del gen y la vía metabólica asociada con este síndrome probablemente contribuirá a un nuevo conocimiento sobre la fisiopatología del envejecimiento humano, con potenciales implicaciones significativas en la investigación del envejecimiento en general. (Texto tomado de la fuente)spa
dc.description.abstractThe neonatal progeroid syndrome of Wiedemann-Rautenstrauch (SWR) is characterized by the manifestation of various aging signs from birth, with a greatly reduced life expectancy averaging 7 months, distinguishing it from other progeroid syndromes. The etiology of SWR has been linked to mutations in the gene encoding RNA polymerase III subunit A (POLR3A), crucial in the regulation of transfer RNA (tRNA), 5S ribosomal RNA (5SrRNA), small nuclear RNAs (snRNAs), and others, resulting in decreased functionality of RNA polymerase III (POLR3) complex and disruptions in ribosomal biogenesis and protein translation, among other processes. The single-point mutation in the POLR3A gene has a considerable impact on the DNA methylation profile of specific genes and regions, causing anomalous gene expression and changes in molecular dynamics. In the case of a 6-year-old patient (POLR3A: c. 3G>T), the study observed abnormal hypomethylation in the gene body regions, while in a 25-year-old patient (POLR3A: c. 3772 3773 del), a tendency toward hypermethylation in promoter and gene body regions was observed. Abnormal gene methylation due to POLR3A mutation primarily affects plasma membrane proteins, disrupting crucial cellular processes such as signal transduction and DNA transcription coding. Significantly methylated genes induce cellular senescence processes. The alteration of normal methylation in CpG dinucleotides by SWR causes acceleration or deceleration in the determination of biological age through the use of epigenetic clocks, which is characteristic of a progeroid syndrome. The study of SWR and its epigenetic implications provides a unique opportunity to understand the pathophysiological processes of human aging. The identification of the gene and the associated metabolic pathway with this syndrome will likely contribute to new knowledge of human aging pathophysiology, with potential significant implications for aging research in general. (Texto tomado de la fuente)eng
dc.description.degreelevelMaestríaspa
dc.description.degreenameMagíster en Ciencias-Bioquímicaspa
dc.description.researchareaBiología del envejecimientospa
dc.description.sponsorshipMinisterio de Ciencia, Tecnología e Innovación Convocatoria 844-2019spa
dc.format.extent88 páginasspa
dc.format.mimetypeapplication/pdfspa
dc.identifier.instnameUniversidad Nacional de Colombiaspa
dc.identifier.reponameRepositorio Institucional Universidad Nacional de Colombiaspa
dc.identifier.repourlhttps://repositorio.unal.edu.co/spa
dc.identifier.urihttps://repositorio.unal.edu.co/handle/unal/86512
dc.language.isospaspa
dc.publisherUniversidad Nacional de Colombiaspa
dc.publisher.branchUniversidad Nacional de Colombia - Sede Bogotáspa
dc.publisher.facultyFacultad de Cienciasspa
dc.publisher.placeBogotá, Colombiaspa
dc.publisher.programBogotá - Ciencias - Maestría en Ciencias - Bioquímicaspa
dc.relation.referencesAnton, L., Brown, A. G., Bartolomei, M. S., & Elovitz, M. A. (2014). Differential Methylation of Genes Associated with Cell Adhesion in Preeclamptic Placentas. PLOS ONE, 9(6), 100148. https://doi.org/10.1371/journal.pone.0100148spa
dc.relation.referencesAquino, E. M., Benton, M. C., Haupt, L. M., Sutherland, H. G., riffiths, L. R. G., & Lea, R. A. (2018). Current Understanding of DNA Methylation and Age-related Disease. OBM Genetics, 2(2), 1–1. https://doi.org/10.21926/obm.genet.1802016spa
dc.relation.referencesArboleda, G., Morales, L. C., Quintero, L., & Arboleda, H. (2011). Neonatal progeroid syndrome (Wiedemann-Rautenstrauch syndrome): Report of three affected sibs. American Journal of Medical Genetics, Part A, 155(7), 1712–1715. https://doi.org/10.1002/ajmg.a.34019spa
dc.relation.referencesAtzmon, G. (2015). Longevity genes: a blueprint of ageing. In Advances in Experimental Medicine and Biology0065-2598 (Vol. 847, Issue Chapter 159). http://www.springerlink.com/index/10.1007/978-0-387-73657-0_159%5Cnpapers2://publication/doi/10.1007/978-0-387-73657-0_159spa
dc.relation.referencesBáez-Becerra, C. T., Valencia-Rincón, E., Velásquez-Méndez, K., Ramírez-Suárez, N. J., Guevara, C., Sandoval-Hernandez, A., Arboleda-Bustos, C. E., Olivos-Cisneros, L., Gutiérrez-Ospina, G., Arboleda, H., & Arboleda, G. (2020a). Nucleolar disruption, activation of P53 and premature senescence in POLR3A-mutated Wiedemann-Rautenstrauch syndrome fibroblasts. Mechanisms of Ageing and Development, 192. https://doi.org/10.1016/j.mad.2020.111360spa
dc.relation.referencesBáez-Becerra, C. T., Valencia-Rincón, E., Velásquez-Méndez, K., Ramírez-Suárez, N. J., Guevara, C., Sandoval-Hernandez, A., Arboleda-Bustos, C. E., Olivos-Cisneros, L., Gutiérrez-Ospina, G., Arboleda, H., & Arboleda, G. (2020b). Nucleolar disruption, activation of P53 and premature senescence in POLR3A-mutated Wiedemann-Rautenstrauch syndrome fibroblasts. Mechanisms Http://Www.Bases.Unal.Edu.Co/Subjects/Databases.Php?Letter=Allof Ageing and Development, 192(September). https://doi.org/10.1016/j.mad.2020.111360spa
dc.relation.referencesBergsma, T., & Rogaeva, E. (2020). DNA Methylation Clocks and Their Predictive Capacity for Aging Phenotypes and Healthspan. Neuroscience Insights, 15, 263310552094222. https://doi.org/10.1177/2633105520942221spa
dc.relation.referencesBerridge, M. J. (2012). Calcium signalling remodelling and disease. Biochemical Society Transactions, 40(2), 297–309. https://doi.org/10.1042/BST20110766spa
dc.relation.referencesBezprozvanny, I. (2019). Calcium hypothesis of neurodegeneration – an update. Biochemical and Biophysical Research Communications, 520(4), 667. https://doi.org/10.1016/J.BBRC.2019.10.016spa
dc.relation.referencesBorsig, L., & Läubli, H. (2019). Cell Adhesion During Tumorigenesis and Metastasis. Encyclopedia of Cancer, 307–314. https://doi.org/10.1016/B978-0-12-801238-3.64991-7spa
dc.relation.referencesCalvanese, V., Lara, E., Kahn, A., & Fraga, M. F. (2009). The role of epigenetics in aging and age-related diseases. Ageing Research Reviews, 8(4), 268–276. https://doi.org/10.1016/j.arr.2009.03.004spa
dc.relation.referencesCarvalho, T. S., & Lussi, A. (2017). Age‐related morphological, histological and functional changes in teeth. Journal of Oral Rehabilitation, 44(4), 291–298. https://doi.org/10.1111/joor.12474spa
dc.relation.referencesChen, B. H., Marioni, R. E., Colicino, E., Peters, M. J., Ward-Caviness, C. K., Tsai, P.-C., Roetker, N. S., Just, A. C., Demerath, E. W., Guan, W., Bressler, J., Fornage, M., Studenski, S., Vandiver, A. R., Moore, A. Z., Tanaka, T., Kiel, D. P., Liang, L., Vokonas, P., … Horvath, S. (2016). DNA methylation-based measures of biological age: meta-analysis predicting time to death. Aging, 8(9), 1844–1865. https://doi.org/10.18632/aging.101020spa
dc.relation.referencesChen, M., Fang, Y., Liang, M., Zhang, N., Zhang, X., Xu, L., Ren, X., Zhang, Q., Zhou, Y., Peng, S., Yu, J., Zeng, J., & Li, X. (2023). The activation of mTOR signalling modulates DNA methylation by enhancing DNMT1 translation in hepatocellular carcinoma. Journal of Translational Medicine, 21(1), 1–17. https://doi.org/10.1186/S12967-023-04103-9/FIGURES/8spa
dc.relation.referencesChoukrallah, M. A., & Matthias, P. (2014). The interplay between chromatin and transcription factor networks during B cell development: Who pulls the trigger first? Frontiers in Immunology, 5(APR), 1–11. https://doi.org/10.3389/fimmu.2014.00156spa
dc.relation.referencesDaniel, F. I., Cherubini, K., Yurgel, L. S., De Figueiredo, M. A. Z., & Salum, F. G. (2011). The role of epigenetic transcription repression and DNA methyltransferases in cancer. Cancer, 117(4), 677–687. https://doi.org/10.1002/cncr.25482spa
dc.relation.referencesDutta, S., Goodrich, J. M., Dolinoy, D. C., & Ruden, D. M. (2023). Biological Aging Acceleration Due to Environmental Exposures: An Exciting New Direction in Toxicogenomics Research. Genes 2024, Vol. 15, Page 16, 15(1), 16. https://doi.org/10.3390/GENES15010016spa
dc.relation.referencesFennell, L., Dumenil, T., Wockner, L., Hartel, G., Nones, K., Bond, C., Borowsky, J., Liu, C., McKeone, D., Bowdler, L., Montgomery, G., Klein, K., Hoffmann, I., Patch, A. M., Kazakoff, S., Pearson, J., Waddell, N., Wirapati, P., Lochhead, P., … Whitehall, V. (2019). Integrative Genome-Scale DNA Methylation Analysis of a Large and Unselected Cohort Reveals 5 Distinct Subtypes of Colorectal Adenocarcinomas. Cmgh, 8(2), 269–290. https://doi.org/10.1016/j.jcmgh.2019.04.002spa
dc.relation.referencesFunes, S. C., Fernández-Fierro, A., Rebolledo-Zelada, D., Mackern-Oberti, J. P., & Kalergis, A. M. (2021). Contribution of Dysregulated DNA Methylation to Autoimmunity. International Journal of Molecular Sciences, 22(21), 11892. https://doi.org/10.3390/ijms222111892spa
dc.relation.referencesGilbert, H. T. J., & Swift, J. (2019). The consequences of ageing, progeroid syndromes and cellular senescence on mechanotransduction and the nucleus. Experimental Cell Research, 378(1), 98–103. https://doi.org/10.1016/j.yexcr.2019.03.002spa
dc.relation.referencesGonzalo, S., Kreienkamp, R., & Askjaer, P. (2017). Hutchinson-Gilford Progeria Syndrome: a premature aging disease caused by LMNA gene mutations. Physiology & Behavior, 176(3), 139–148. https://doi.org/10.1016/j.arr.2016.06.007.Hutchinson-Gilfordspa
dc.relation.referencesGuastafierro, T., Bacalini, M. G., Marcoccia, A., Gentilini, D., Pisoni, S., Di Blasio, A. M., Corsi, A., Franceschi, C., Raimondo, D., Spanò, A., Garagnani, P., & Bondanini, F. (2017). Genome-wide DNA methylation analysis in blood cells from patients with Werner syndrome. Clinical Epigenetics, 9(1), 1–10. https://doi.org/10.1186/s13148-017-0389-4spa
dc.relation.referencesHan, Y., Yan, C., Fishbain, S., Ivanov, I., & He, Y. (2018). Structural visualization of RNA polymerase III transcription machineries. Cell Discovery, 4(1), 40. https://doi.org/10.1038/s41421-018-0044-zspa
dc.relation.referencesHannum, G., Guinney, J., Zhao, L., Zhang, L., Hughes, G., Sadda, S., Klotzle, B., Bibikova, M., Fan, J.-B., Gao, Y., Deconde, R., Chen, M., Rajapakse, I., Friend, S., Ideker, T., & Zhang, K. (2013). Genome-wide Methylation Profiles Reveal Quantitative Views of Human Aging Rates. Molecular Cell, 49(2), 359–367. https://doi.org/10.1016/j.molcel.2012.10.016spa
dc.relation.referencesHennekam, R. C. M. (2020). Pathophysiology of premature aging characteristics in Mendelian progeroid disorders. European Journal of Medical Genetics, 63(11), 104028. https://doi.org/10.1016/j.ejmg.2020.104028spa
dc.relation.referencesHiraide, T., Nakashima, M., Ikeda, T., Tanaka, D., Osaka, H., & Saitsu, H. (2020). Identification of a deep intronic POLR3A variant causing inclusion of a pseudoexon derived from an Alu element in Pol III-related leukodystrophy. Journal of Human Genetics, 65(10), 921–925. https://doi.org/10.1038/s10038-020-0786-yspa
dc.relation.referencesHodjat, M., Khan, F., & Saadat, K. A. S. M. (2020). Epigenetic alterations in aging tooth and the reprogramming potential. Ageing Research Reviews, 63(July), 101140. https://doi.org/10.1016/j.arr.2020.101140spa
dc.relation.referencesHorvath, S. (2013). DNA methylation age of human tissues and cell types. Genome Biology, 14(10), R115. https://doi.org/10.1186/gb-2013-14-10-r115spa
dc.relation.referencesHu, S., Wu, J., Chen, L., & Shan, G. (2012). Signals from noncoding RNAs: Unconventional roles for conventional pol III transcripts. International Journal of Biochemistry and Cell Biology, 44(11), 1847–1851. https://doi.org/10.1016/j.biocel.2012.07.013spa
dc.relation.referencesHuidobro, C., Fernandez, A. F., & Fraga, M. F. (2013). Aging epigenetics: Causes and consequences. Molecular Aspects of Medicine, 34(4), 765–781. https://doi.org/10.1016/j.mam.2012.06.006spa
dc.relation.referencesIrizarry, R. A., Ladd-Acosta, C., Wen, B., Wu, Z., Montano, C., Onyango, P., Cui, H., Gabo, K., Rongione, M., & Webster, M. (2009). Genome-wide methylation analysis of human colon cancer reveals similar hypo-and hypermethylation at conserved tissue-specific CpG island shores. Nature Genetics, 41(2), 178. https://doi.org/10.1038/ng.298.Genome-widespa
dc.relation.referencesIllumina. (2021). MethylationEPIC v1.0 LIMS Product Descriptor File (p. 50).spa
dc.relation.referencesIto, S., D’Alessio, A. C., Taranova, O. V., Hong, K., Sowers, L. C., & Zhang, Y. (2010). Role of Tet proteins in 5mC to 5hmC conversion, ES-cell self-renewal and inner cell mass specification. Nature, 466(7310), 1129–1133. https://doi.org/10.1038/nature09303spa
dc.relation.referencesIto, T., Kubiura-Ichimaru, M., Murakami, Y., Bogutz, A. B., Lefebvre, L., Suetake, I., Tajima, S., & Tada, M. (2022). DNMT1 regulates the timing of DNA methylation by DNMT3 in an enzymatic activity-dependent manner in mouse embryonic stem cells. PLOS ONE, 17(1), e0262277. https://doi.org/10.1371/journal.pone.0262277spa
dc.relation.referencesJay, A. M., Conway, R. L., Thiffault, I., Saunders, C., Farrow, E., Adams, J., & Toriello, H. V. (2016). Neonatal progeriod syndrome associated with biallelic truncating variants in POLR3A. American Journal of Medical Genetics, Part A, 170(12), 3343–3346. https://doi.org/10.1002/ajmg.a.37960spa
dc.relation.referencesJeltsch, A., Ehrenhofer-Murray, A., Jurkowski, T. P., Lyko, F., Reuter, G., Ankri, S., Nellen, W., Schaefer, M., & Helm, M. (2017). Mechanism and biological role of Dnmt2 in Nucleic Acid Methylation. RNA Biology, 14(9), 1108–1123. https://doi.org/10.1080/15476286.2016.1191737spa
dc.relation.referencesJi, Y., Xie, Y., Zhang, M., Zhou, J., Peng, L., Zheng, Y., & Shu, S. (2023). Role of SATB2 5’ Untranslated Region Promoter Methylation in Formation of Non-syndromic Cleft Palate Only. Eurasian Journal of Medicine and Oncology, 7(2), 165–173. https://doi.org/10.14744/ejmo.2023.41377spa
dc.relation.referencesJin, B., & Robertson, K. D. (2013). DNA methyltransferases, DNA damage repair, and cancer. Advances in Experimental Medicine and Biology, 754, 3–29. https://doi.org/10.1007/978-1-4419-9967-2_1spa
dc.relation.referencesKabacik, S., Lowe, D., Fransen, L., Leonard, M., Ang, S.-L., Whiteman, C., Corsi, S., Cohen, H., Felton, S., Bali, R., Horvath, S., & Raj, K. (2022). The relationship between epigenetic age and the hallmarks of aging in human cells. Nature Aging, 2(6), 484–493. https://doi.org/10.1038/s43587-022-00220-0spa
dc.relation.referencesKipling, D., Davis, T., Ostler, E. L., & Faragher, R. G. A. (2004). What Can Progeroid Syndromes Tell Us About Human Aging? Science, 305(5689), 1426–1431. https://doi.org/10.1126/science.1102587spa
dc.relation.referencesKling, T., & Carén, H. (2019). Methylation Analysis Using Microarrays: Analysis and Interpretation. In Methods in Molecular Biology (Vol. 1908, Issue July, pp. 205–217). Humana Press. https://doi.org/10.1007/978-1-4939-9004-7_14spa
dc.relation.referencesKoval, A. P., Veniaminova, N. A., & Kramerov, D. A. (2011). Additional box B of RNA polymerase III promoter in SINE B1 can be functional. Gene, 487(2), 113–117. https://doi.org/10.1016/j.gene.2011.08.001spa
dc.relation.referencesKuzmina, N. S., Lapteva, N. S., & Rubanovich, A. V. (2016). Hypermethylation of gene promoters in peripheral blood leukocytes in humans long term after radiation exposure. Environmental Research, 146, 10–17. https://doi.org/10.1016/j.envres.2015.12.008spa
dc.relation.referencesLander, E. S., Linton, L. M., Birren, B., Nusbaum, C., Zody, M. C., Baldwin, J., Devon, K., Dewar, K., Doyle, M., Fitzhugh, W., Funke, R., Gage, D., Harris, K., Heaford, A., Howland, J., Kann, L., Lehoczky, J., Levine, R., McEwan, P., … Chen, Y. J. (2001). Erratum: Initial sequencing and analysis of the human genome: International Human Genome Sequencing Consortium (Nature (2001) 409 (860-921)). Nature, 412(6846), 565–566. https://doi.org/10.1038/35087627spa
dc.relation.referencesLee, K.-A., Flores, R. R., Jang, I. H., Saathoff, A., & Robbins, P. D. (2022). Immune Senescence, Immunosenescence and Aging. Frontiers in Aging, 3. https://doi.org/10.3389/fragi.2022.900028spa
dc.relation.referencesLevine, M. E., Lu, A. T., Quach, A., Chen, B. H., Assimes, T. L., Bandinelli, S., Hou, L., Baccarelli, A. A., Stewart, J. D., Li, Y., Whitsel, E. A., Wilson, J. G., Reiner, A. P., Aviv, A., Lohman, K., Liu, Y., Ferrucci, L., & Horvath, S. (2018). An epigenetic biomarker of aging for lifespan and healthspan. AGING, 10(4). www.aging-us.comspa
dc.relation.referencesLiu, N., Yang, R., Shi, Y., Chen, L., Liu, Y., Wang, Z., Liu, S., Ouyang, L., Wang, H., Lai, W., Mao, C., Wang, M., Cheng, Y., Liu, S., Wang, X., Zhou, H., Cao, Y., Xiao, D., & Tao, Y. (2020). The cross-talk between methylation and phosphorylation in lymphoid-specific helicase drives cancer stem-like properties. Signal Transduction and Targeted Therapy 2020 5:1, 5(1), 1–14. https://doi.org/10.1038/s41392-020-00249-wspa
dc.relation.referencesLiu, Z., Leung, D., Thrush, K., Zhao, W., Ratliff, S., Tanaka, T., Schmitz, L. L., Smith, J. A., Ferrucci, L., & Levine, M. E. (2020). Underlying features of epigenetic aging clocks in vivo and in vitro. Aging Cell, 19(10). https://doi.org/10.1111/acel.13229spa
dc.relation.referencesLoaeza-Loaeza, J., Beltran, A. S., & Hernández-Sotelo, D. (2020). Dnmts and impact of cpg content, transcription factors, consensus motifs, lncrnas, and histone marks on dna methylation. In Genes (Vol. 11, Issue 11, pp. 1–19). MDPI AG. https://doi.org/10.3390/genes11111336spa
dc.relation.referencesLópez-Otín, C., Blasco, M. A., Partridge, L., Serrano, M., & Kroemer, G. (2013). The Hallmarks of Aging. Cell, 153(6), 1194–1217. https://doi.org/10.1016/j.cell.2013.05.039spa
dc.relation.referencesLuo, R., Bai, C., Yang, L., Zheng, Z., Su, G., Gao, G., Wei, Z., Zuo, Y., & Li, G. (2018). Correction to: DNA methylation subpatterns at distinct regulatory regions in human early embryos (Open Biology (2018) 8 (180131) DOI: 10.1098/rsob.180131). Open Biology, 8(12), 1–9. https://doi.org/10.1098/rsob.180215spa
dc.relation.referencesMagalingam, K. B., Somanath, S. D., & Radhakrishnan, A. K. (2023). A Glimpse into the Genome-wide DNA Methylation Changes in 6-hydroxydopamine-induced In Vitro Model of Parkinson’s Disease. Experimental Neurobiology, 32(3), 119–132. https://doi.org/10.5607/en22035spa
dc.relation.referencesMelo dos Santos, L. S., Trombetta-Lima, M., Eggen, B. J. L., & Demaria, M. (2024). Cellular senescence in brain aging and neurodegeneration. In Ageing Research Reviews (Vol. 93). Elsevier Ireland Ltd. https://doi.org/10.1016/j.arr.2023.102141spa
dc.relation.referencesMillan, J., Lesarri, A., Fernández, J. A., & Martínez, R. (2021). Exploring Epigenetic Marks by Analysis of Noncovalent Interactions. ChemBioChem, 22(2), 408–415. https://doi.org/10.1002/cbic.202000380spa
dc.relation.referencesMinnerop, M., Kurzwelly, D., Wagner, H., Soehn, A. S., Reichbauer, J., Tao, F., Rattay, T. W., Peitz, M., Rehbach, K., Giorgetti, A., Pyle, A., Thiele, H., Altmüller, J., Timmann, D., Karaca, I., Lennarz, M., Baets, J., Hengel, H., Synofzik, M., … Schüle, R. (2017). Hypomorphic mutations in POLR3A are a frequent cause of sporadic and recessive spastic ataxia. Brain, 140(6), 1561–1578. https://doi.org/10.1093/brain/awx095spa
dc.relation.referencesMoore, L. D., Le, T., & Fan, G. (2013). DNA Methylation and Its Basic Function. Neuropsychopharmacology, 38(1), 23–38. https://doi.org/10.1038/npp.2012.112spa
dc.relation.referencesMorris, T., Stirling, L., Feber, A., & Teschendorff, A. (2024). Package ‘ ChAMP .’spa
dc.relation.referencesMuse, M. E., Titus, A. J., Salas, L. A., Wilkins, O. M., Mullen, C., Gregory, K. J., Schneider, S. S., Crisi, G. M., Jawale, R. M., Otis, C. N., Christensen, B. C., & Arcaro, K. F. (2020). Enrichment of CpG island shore region hypermethylation in epigenetic breast field cancerization. Epigenetics, 15(10), 1093–1106. https://doi.org/10.1080/15592294.2020.1747748spa
dc.relation.referencesNelson, R. (2019). POLR3A Identified as Major Locus for Autosomal Recessive Wiedemann-Rautenstrauch Syndrome: New findings show “compelling evidence” that POLR3A mutations underlie the etiology of autosomal-recessive WRS. American Journal of Medical Genetics, Part A, 179(2), 146–147. https://doi.org/10.1002/ajmg.a.61040spa
dc.relation.referencesPanja, S., Hayati, S., Epsi, N. J., Parrott, J. S., & Mitrofanova, A. (2018). Integrative (epi) Genomic Analysis to Predict Response to Androgen-Deprivation Therapy in Prostate Cancer. EBioMedicine, 31, 110–121. https://doi.org/10.1016/j.ebiom.2018.04.007spa
dc.relation.referencesPaolacci, S., Bertola, D., Franco, J., Mohammed, S., Tartaglia, M., Wollnik, B., & Hennekam, R. C. (2017). Wiedemann–Rautenstrauch syndrome: A phenotype analysis. American Journal of Medical Genetics, Part A, 173(7), 1763–1772. https://doi.org/10.1002/ajmg.a.38246spa
dc.relation.referencesPaolacci, S., Li, Y., Agolini, E., Bellacchio, E., Arboleda-Bustos, C. E., Carrero, D., Bertola, D., Al-Gazali, L., Alders, M., Altmuller, J., Arboleda, G., Beleggia, F., Bruselles, A., Ciolfi, A., Gillessen-Kaesbach, G., Krieg, T., Mohammed, S., Muller, C., Novelli, A., … Hennekam, R. C. (2018). Specific combinations of biallelic POLR3A variants cause Wiedemann-Rautenstrauch syndrome. Journal of Medical Genetics, 55(12), 837–846. https://doi.org/10.1136/jmedgenet-2018-105528spa
dc.relation.referencesPark, J. L., Lee, Y. S., Kunkeaw, N., Kim, S. Y., Kim, I. H., & Lee, Y. S. (2017). Epigenetic regulation of noncoding RNA transcription by mammalian RNA polymerase III. Epigenomics, 9(2), 171–187. https://doi.org/10.2217/epi-2016-0108spa
dc.relation.referencesProud, C. G. (2019). Phosphorylation and Signal Transduction Pathways in Translational Control. Cold Spring Harbor Perspectives in Biology, 11(7). https://doi.org/10.1101/CSHPERSPECT.A033050spa
dc.relation.referencesPuig, N., & Agrelo, R. (2012). From aging to cancer: a DNA methylation journey. Ageing Research, 3(1), 4. https://doi.org/10.4081/ar.2012.e4spa
dc.relation.referencesRautenstrauch, T., Snigula, F., Krieg, T., Gay, S., & Müller, P. K. (1977). Progeria: A cell culture study and clinical report of familial incidence. European Journal of Pediatrics, 124(2), 101–111. https://doi.org/10.1007/BF00477545spa
dc.relation.referencesReale, A., Tagliatesta, S., Zardo, G., & Zampieri, M. (2022). Counteracting aged DNA methylation states to combat ageing and age-related diseases. Mechanisms of Ageing and Development, 206(June), 111695. https://doi.org/10.1016/j.mad.2022.111695spa
dc.relation.referencesSakaki, M., Ebihara, Y., Okamura, K., Nakabayashi, K., Igarashi, A., Matsumoto, K., Hata, K., Kobayashi, Y., & Maehara, K. (2017). Potential roles of DNA methylation in the initiation and establishment of replicative senescence revealed by array-based methylome and transcriptome analyses. PLoS ONE, 12(2). https://doi.org/10.1371/journal.pone.0171431spa
dc.relation.referencesSaneyasu, T., Fukuzo, S., Kitashiro, A., Nagata, K., Honda, K., & Kamisoyama, H. (2019). Central administration of insulin and refeeding lead to the phosphorylation of AKT, but not FOXO1, in the hypothalamus of broiler chicks. Physiology and Behavior, 210(August), 112644. https://doi.org/10.1016/j.physbeh.2019.112644spa
dc.relation.referencesSchmauck-Medina, T., Molière, A., Lautrup, S., Zhang, J., Chlopicki, S., Madsen, H. B., Cao, S., Soendenbroe, C., Mansell, E., Vestergaard, M. B., Li, Z., Shiloh, Y., Opresko, P. L., Egly, J. M., Kirkwood, T., Verdin, E., Bohr, V. A., Cox, L. S., Stevnsner, T., … Fang, E. F. (2022). New hallmarks of ageing: a 2022 Copenhagen ageing meeting summary. Aging, 14(16), 6829–6839. https://doi.org/10.18632/AGING.204248spa
dc.relation.referencesSilver, B. B., & Nelson, C. M. (2018). The Bioelectric Code: Reprogramming Cancer and Aging From the Interface of Mechanical and Chemical Microenvironments. Frontiers in Cell and Developmental Biology, 6. https://doi.org/10.3389/fcell.2018.00021spa
dc.relation.referencesSpangle, J. M., Roberts, T. M., & Zhao, J. J. (2017). The emerging role of PI3K/AKT-mediated epigenetic regulation in cancer. Biochimica et Biophysica Acta, 1868(1), 123. https://doi.org/10.1016/J.BBCAN.2017.03.002spa
dc.relation.referencesStasenko, D. V., Tatosyan, K. A., Borodulina, O. R., & Kramerov, D. A. (2023). Nucleotide Context Can Modulate Promoter Strength in Genes Transcribed by RNA Polymerase III. Genes, 14(4), 802. https://doi.org/10.3390/genes14040802spa
dc.relation.referencesTahiliani, M., Koh, K. P., Shen, Y., Pastor, W. A., Bandukwala, H., Brudno, Y., Agarwal, S., Iyer, L. M., Liu, D. R., Aravind, L., & Rao, A. (2009). Conversion of 5-Methylcytosine to 5-Hydroxymethylcytosine in Mammalian DNA by MLL Partner TET1. Science, 324(5929), 930–935. https://doi.org/10.1126/science.1170116spa
dc.relation.referencesTemel, S. G., Ergoren, M. C., Manara, E., Paolacci, S., Tuncel, G., Gul, S., & Bertelli, M. (2020). Unique combination and in silico modeling of biallelic POLR3A variants as a cause of Wiedemann–Rautenstrauch syndrome. European Journal of Human Genetics, 28(12), 1675–1680. https://doi.org/10.1038/s41431-020-0673-1spa
dc.relation.referencesVisone, R., Bacalini, M. G., Franco, S. Di, Ferracin, M., Colorito, M. L., Pagotto, S., Laprovitera, N., Licastro, D., Marco, M. Di, Scavo, E., Bassi, C., Saccenti, E., Nicotra, A., Grzes, M., Garagnani, P., Laurenzi, V. De, Valeri, N., Mariani-Costantini, R., Negrini, M., … Veronese, A. (2019a). DNA methylation of shelf, shore and open sea CpG positions distinguish high microsatellite instability from low or stable microsatellite status colon cancer stem cells. Epigenomics, 11(6), 587–604. https://doi.org/10.2217/epi-2018-0153spa
dc.relation.referencesVisone, R., Bacalini, M. G., Franco, S. Di, Ferracin, M., Colorito, M. L., Pagotto, S., Laprovitera, N., Licastro, D., Marco, M. Di, Scavo, E., Bassi, C., Saccenti, E., Nicotra, A., Grzes, M., Garagnani, P., Laurenzi, V. De, Valeri, N., Mariani-Costantini, R., Negrini, M., … Veronese, A. (2019b). DNA methylation of shelf, shore and open sea CpG positions distinguish high microsatellite instability from low or stable microsatellite status colon cancer stem cells. Epigenomics, 11(6), 587–604. https://doi.org/10.2217/epi-2018-0153spa
dc.relation.referencesWambach, J. A., Wegner, D. J., Patni, N., Kircher, M., Willing, M. C., Baldridge, D., Xing, C., Agarwal, A. K., Vergano, S. A. S., Patel, C., Grange, D. K., Kenney, A., Najaf, T., Nickerson, D. A., Bamshad, M. J., Cole, F. S., & Garg, A. (2018). Bi-allelic POLR3A Loss-of-Function Variants Cause Autosomal-Recessive Wiedemann-Rautenstrauch Syndrome. American Journal of Human Genetics, 103(6), 968–975. https://doi.org/10.1016/j.ajhg.2018.10.010spa
dc.relation.referencesWan, R., Srikaram, P., Guntupalli, V., Hu, C., Chen, Q., & Gao, P. (2023). Cellular senescence in asthma: from pathogenesis to therapeutic challenges. www.thelancet.comspa
dc.relation.referencesWang, Q., Xiong, F., Wu, G., Liu, W., Chen, J., Wang, B., & Chen, Y. (2022). Gene body methylation in cancer: molecular mechanisms and clinical applications. Clinical Epigenetics, 14(1), 1–14. https://doi.org/10.1186/s13148-022-01382-9spa
dc.relation.referencesWang, Y., Huang, W., Zheng, S., Wang, L., Zhang, L., & Pei, X. (2024). Construction of an immune-related risk score signature for gastric cancer based on multi-omics data. Scientific Reports, 14(1), 1422. https://doi.org/10.1038/s41598-024-52087-3spa
dc.relation.referencesWeidner, C., Lin, Q., Koch, C., Eisele, L., Beier, F., Ziegler, P., Bauerschlag, D., Jöckel, K.-H., Erbel, R., Mühleisen, T., Zenke, M., Brümmendorf, T., & Wagner, W. (2014). Aging of blood can be tracked by DNA methylation changes at just three CpG sites. Genome Biology, 15(2), R24. https://doi.org/10.1186/gb-2014-15-2-r24spa
dc.relation.referencesWelsh, H., Batalha, C. M. P. F., Li, W., Mpye, K. L., Souza-Pinto, N. C., Naslavsky, M. S., & Parra, E. J. (2023). A systematic evaluation of normalization methods and probe replicability using infinium EPIC methylation data. Clinical Epigenetics, 15(1), 41. https://doi.org/10.1186/s13148-023-01459-zspa
dc.relation.referencesWu, X., Chen, W., Lin, F., Huang, Q., Zhong, J., Gao, H., Song, Y., & Liang, H. (2019). DNA methylation profile is a quantitative measure of biological aging in children. Aging, 11(22), 10031–10051. https://doi.org/10.18632/aging.102399spa
dc.relation.referencesXiao, F.-H., Wang, H.-T., & Kong, Q.-P. (2019). Dynamic DNA Methylation During Aging: A “Prophet” of Age-Related Outcomes. Frontiers in Genetics, 10. https://doi.org/10.3389/fgene.2019.00107spa
dc.relation.referencesXie, W., Baylin, S. B., & Easwaran, H. (2019). DNA methylation in senescence, aging and cancer Origin of cancer epigenome from cycling aging cells (Vol. 6, Issue 2). www.impactjournals.com/oncoscience/spa
dc.relation.referencesYadav, M. L., & Mohapatra, B. (2018). Intergenic. Encyclopedia of Animal Cognition and Behavior, 1–5. https://doi.org/10.1007/978-3-319-47829-6_64-1spa
dc.relation.referencesYen, B. L., Hwa, H. L., Hsu, P. J., Chen, P. M., Wang, L. T., Jiang, S. S., Liu, K. J., Sytwu, H. K., & Yen, M. L. (2020). Hla-g expression in human mesenchymal stem cells (Mscs) is related to unique methylation pattern in the proximal promoter as well as gene body dna. International Journal of Molecular Sciences, 21(14), 1–14. https://doi.org/10.3390/ijms21145075spa
dc.relation.referencesYuan, T., Jiao, Y., de Jong, S., Ophoff, R. A., Beck, S., & Teschendorff, A. E. (2015). An Integrative Multi-scale Analysis of the Dynamic DNA Methylation Landscape in Aging. PLoS Genetics, 11(2), 1–21. https://doi.org/10.1371/journal.pgen.1004996spa
dc.relation.referencesYukawa, Y. (2023). Plant Molecular Biology Lab. https://www.nsc.nagoya-cu.ac.jp/~yyuk/e-index.htmlspa
dc.relation.referencesZane, L., Sharma, V., & Misteli, T. (2014). Common features of chromatin in aging and cancer: cause or coincidence? Trends in Cell Biology, 24(11), 686–694. https://doi.org/10.1016/j.tcb.2014.07.001spa
dc.relation.referencesZhang, Y., Wang, Y., Luo, M., Xu, F., Lu, Y., Zhou, X., Cui, W., & Miao, L. (2019). Elabela protects against podocyte injury in mice with streptozocin-induced diabetes by associating with the PI3K/Akt/mTOR pathway. Peptides, 114(February), 29–37. https://doi.org/10.1016/j.peptides.2019.04.005spa
dc.relation.referencesZouali, M. (2021). DNA methylation signatures of autoimmune diseases in human B lymphocytes. Clinical Immunology, 222, 108622. https://doi.org/10.1016/j.clim.2020.108622spa
dc.relation.referencesZuo, S., Shi, G., Fan, J., Fan, B., Zhang, X., Liu, S., Hao, Y., Wei, Z., Zhou, X., & Feng, S. (2021). Identification of adhesion-associated DNA methylation patterns in the peripheral nervous system. Experimental and Therapeutic Medicine, 21(1). https://doi.org/10.3892/ETM.2020.9479spa
dc.rights.accessrightsinfo:eu-repo/semantics/openAccessspa
dc.rights.licenseAtribución-NoComercial 4.0 Internacionalspa
dc.rights.urihttp://creativecommons.org/licenses/by-nc/4.0/spa
dc.subject.ddc610 - Medicina y saludspa
dc.subject.ddc570 - Biología::572 - Bioquímicaspa
dc.subject.ddc610 - Medicina y salud::618 - Ginecología, obstetricia, pediatría, geriatríaspa
dc.subject.decsMetilación de ADNspa
dc.subject.decsDNA methylationeng
dc.subject.decsEpigénesis genéticaspa
dc.subject.decsEpigenesis, geneticeng
dc.subject.decsEnvejecimiento prematurospa
dc.subject.decsAging, prematureeng
dc.subject.decsTransducción de señalspa
dc.subject.decsSignal transductioneng
dc.subject.proposalMetilación de ADNspa
dc.subject.proposalProgeriaspa
dc.subject.proposalSenescenciaspa
dc.subject.proposalTransducción de señalesspa
dc.subject.proposalSWRspa
dc.subject.proposalDNA methylationeng
dc.subject.proposalSenescenceeng
dc.subject.proposalSignal transductioneng
dc.subject.umlsSíndrome de Wiedemann-Rauternstrauchspa
dc.subject.umlsWiedemann-Rautenstrauch syndromeeng
dc.subject.umlsTrastorno metabólico neonatalspa
dc.subject.umlsNeonatal metabolic disordereng
dc.subject.umlsARN Polimerasa IIIspa
dc.subject.umlsRNA Polymerase IIIeng
dc.titleEstudio del perfil de metilación de ADN en pacientes con síndrome progeroide neonatal (síndrome de Wiedemann-Rautenstrauch)spa
dc.title.translatedStudy of DNA methylation profile in patients with neonatal progeroid syndrome (Wiedemann-Rautenstrauch syndrome)eng
dc.typeTrabajo de grado - Maestríaspa
dc.type.coarhttp://purl.org/coar/resource_type/c_bdccspa
dc.type.coarversionhttp://purl.org/coar/version/c_ab4af688f83e57aaspa
dc.type.contentTextspa
dc.type.driverinfo:eu-repo/semantics/masterThesisspa
dc.type.redcolhttp://purl.org/redcol/resource_type/TMspa
dc.type.versioninfo:eu-repo/semantics/acceptedVersionspa
dcterms.audience.professionaldevelopmentInvestigadoresspa
oaire.accessrightshttp://purl.org/coar/access_right/c_abf2spa
oaire.awardtitleEstudio del Perfil de Metilación de ADN En Pacientes con Síndrome Progeroide Neonatal (Síndrome de Wiedemann-Rautenstrauch)spa
oaire.fundernameMinisterio de Ciencia, Tecnología e Innovaciónspa

Archivos

Bloque original

Mostrando 1 - 1 de 1
Cargando...
Miniatura
Nombre:
1072657220.2024.pdf
Tamaño:
2.1 MB
Formato:
Adobe Portable Document Format
Descripción:
Tesis de Magíster en Ciencias-Bioquímica

Bloque de licencias

Mostrando 1 - 2 de 2
Cargando...
Miniatura
Nombre:
license.txt
Tamaño:
5.74 KB
Formato:
Item-specific license agreed upon to submission
Descripción:
Cargando...
Miniatura
Nombre:
Herman Fredy Barrera Torres embargo.pdf
Tamaño:
856.93 KB
Formato:
Adobe Portable Document Format
Descripción: