Mostrar el registro sencillo del documento

dc.rights.licenseAtribución-NoComercial-SinDerivadas 4.0 Internacional
dc.contributor.advisorArboleda Granados, Humberto
dc.contributor.authorRodríguez Angarita, Óscar Gerardo
dc.date.accessioned2020-02-13T12:20:15Z
dc.date.available2020-02-13T12:20:15Z
dc.date.issued2018
dc.identifier.urihttps://repositorio.unal.edu.co/handle/unal/75585
dc.description.abstractEl TDAH es un trastorno comportamental altamente heredable, persistente y frecuente. La etiología del trastorno no está clara. Sin embargo, se han descrito similitudes neuroanatómicas entre trastorno de déficit de atención e hiperactividad (TDAH), trastorno obsesivo compulsivo (TOC) y espectro autista (EA). Por otra parte, se encontrado asociación significativa entre variantes genómicas en el gen BTBD3 y TOC y áreas diferencialmente metiladas en este mismo gen asociadas a TDAH. Este trabajo se propuso determinar el patrón de metilación de un área del gen BTBD3 en una muestra de pacientes Colombianos diagnosticados con TDAH. Adicionalmente se generaron comparaciones en los niveles de metilación en función de diversas variables clínicas. De manera complementaria, se desarrollaron análisis no considerados en los objetivos del trabajo, se generó un análisis de interacción proteína-proteína (PPi) alrededor del gen BTBD3 y un análisis de expresión y co-expresipon del gen en cerebro prenatal y adulto. Esto con el fin de tener un entendimiento más profundo de la función de BTBD3 en el neurodesarrollo y la conducta El diagnósticos de TDAH no se asocia con cambios en la metilación. Se observa relación de la metilación con estrés prenatal, el tipo de parto, estrés durante el embarazo, si fue o no un embarazo deseado, si hubo algún riesgo para el embarazo durante el tercer trimestre, tipo de parto enfermedades al nacer,tipo de alimentación, región de nacimiento edad del padre y de la madre. La red de interacciones entre proteínas está dominado por procesos asociados al metabolismo mediado por ubiquitinación. La desestabilización topológica mostró cambios significativos en topológica y enriquecimiento funcional. El análisis de expresión mostró que el gen BTBD3 tiende a estar más expresado en cerebro prenatal que en adulto pero con co-expresiones más débiles.
dc.description.abstractIntroduction: ADHD is a behavioral disorder highly heritable, persistent and frequent. The etiology of the disorder is not clear. Nevertheless, neuroanatomical similarities has been described between attention deficit hyperactivity disorder ADHD, obsessive compulsive disorder (OCD) and autistic spectrum (AS). Moreover, there is significant association between genomic variants in BTBD3 and OCD and differentially methylated areas in the before mentioned gene associated to ADHD. This work attempt to determine the methylation pattern in a region of BTBD3 in a sample of Colombian patients diagnosed with ADHD. Additional, methylation was compared as function of clinical variables. in a complementary way, it was performed several not before considered analysis in the goals of the investigation, it was developed a protein-protein (PPi) interaction analysis surrounding the BTBD3 gene and an expression and coexpression analysis of the gene in the prenatal and adult brain. This with the goal get a more profound understanding of the BTBD3 function in the neurodevelopment and in the behavior The diagnosis of ADHD is not associated with changes in methylation. Relations are observed when diagnosis interact with prenatal stress, type of delivery, stress during pregnancy, if it was a desired pregnant, risks in the third trimester of pregnancy, type of delivery, diseases at birth, type of feeding, region of birth, age of father and mother. The network of interactions between proteins is dominated by metabolism mediated by ubiquitination. Topological destabilization showed significant changes in topological and functional enrichment. The expression analysis show than the BTBD3 gene tend to be more expressed in the prenatal brain than in the adult brain but with lower coexpression
dc.format.extent151
dc.format.mimetypeapplication/pdf
dc.language.isospa
dc.rightsDerechos reservados - Universidad Nacional de Colombia
dc.rights.urihttp://creativecommons.org/licenses/by-nc-nd/4.0/
dc.subject.ddcNeurociencias
dc.titleAspectos epigenéticos del gen BTBD3 en niños diagnosticados con TDAH en una muestra de pacientes colombianos
dc.typeDocumento de trabajo
dc.rights.spaAcceso abierto
dc.description.additionalMagister en Neurociencias.
dc.type.driverinfo:eu-repo/semantics/workingPaper
dc.type.versioninfo:eu-repo/semantics/acceptedVersion
dc.contributor.researchgroupGrupo de Neurociencias
dc.description.degreelevelMaestría
dc.publisher.branchUniversidad Nacional de Colombia - Sede Bogotá
dc.relation.referencesAdkins, R. M., Thomas, F., Tylavsky, F. A., & Krushkal, J. (2011). Parental ages and levels of DNA methylation in the newborn are correlated. BMC Medical Genetics, 12, 47. Allin, M. P. G., Salaria, S., Nosarti, C., Wyatt, J., Rifkin, L., & Murray, R. M. (2005). Vermis and lateral lobes of the cerebellum in adolescents born very preterm. Neuroreport, 16(16), 1821–1824. Ameis, S. H., Lerch, J. P., Taylor, M. J., Lee, W., Viviano, J. D., Pipitone, J., … Anagnostou, E. (2016). A Diffusion Tensor Imaging Study in Children With ADHD, Autism Spectrum Disorder, OCD, and Matched Controls: Distinct and Non-Distinct White Matter Disruption and Dimensional Brain-Behavior Relationships. The American Journal of Psychiatry, 173(12), 1213–1222. American Psychiatric Association. (2009a). American Psychiatric Association Practice Guidelines for the Treatment of Psychiatric Disorders. American Psychiatric Association. (2009b). American Psychiatric Association Practice Guidelines for the Treatment of Psychiatric Disorders. Angeles Fernández-Gil, M., Palacios-Bote, R., Leo-Barahona, M., & Mora-Encinas, J. P. (2010). Anatomy of the brainstem: a gaze into the stem of life. Seminars in Ultrasound, CT, and MR, 31(3), 196–219. Bardwell, V. J., & Treisman, R. (1994). The POZ domain: a conserved protein-protein interaction motif. Genes & Development, 8(14), 1664–1677. Barkley, R. A. (1997). Behavioral inhibition, sustained attention, and executive functions: constructing a unifying theory of ADHD. Psychological Bulletin, 121(1), 65–94. Barkley, R. A. (2005). Attention-Deficit Hyperactivity Disorder, Third Edition: A Handbook for Diagnosis and Treatment. Guilford Press. Bartolomei, M. S., & Ferguson-Smith, A. C. (2011). Mammalian genomic imprinting. Cold Spring Harbor Perspectives in Biology, 3(7). https://doi.org/10.1101/cshperspect.a002592 Baylin, S. B., & Jones, P. A. (2016). Epigenetic Determinants of Cancer. Cold Spring Harbor Perspectives in Biology, 8(9). https://doi.org/10.1101/cshperspect.a019505 Bell, A. S. (2011a). A critical review of ADHD diagnostic criteria: what to address in the DSM-V. Journal of Attention Disorders, 15(1), 3–10. Bell, A. S. (2011b). A critical review of ADHD diagnostic criteria: what to address in the DSM-V. Journal of Attention Disorders, 15(1), 3–10. Benevento, L. A., Rezak, M., & Santos-Anderson, R. (1977). An autoradiographic study of the projections of the pretectum in the rhesus monkey (macaca mulatta): evidence for sensorimotor links to the thalamus and oculomotor nuclei. Brain Research, 127(2), 197–218. Berger, S. L., Kouzarides, T., Shiekhattar, R., & Shilatifard, A. (2009). An operational definition of epigenetics. Genes & Development, 23(7), 781–783. Bestor, T. H. (1998). The host defence function of genomic methylation patterns. Novartis Foundation Symposium, 214, 187–195; discussion 195–199, 228–232. Bind, M.-A., Zanobetti, A., Gasparrini, A., Peters, A., Coull, B., Baccarelli, A., … Schwartz, J. (2014). Effects of temperature and relative humidity on DNA methylation. Epidemiology , 25(4), 561–569. Bohacek, J., & Mansuy, I. M. (2015a). Molecular insights into transgenerational non-genetic inheritance of acquired behaviours. Nature Reviews. Genetics, 16(11), 641–652. Bohacek, J., & Mansuy, I. M. (2015b). Molecular insights into transgenerational non-genetic inheritance of acquired behaviours. Nature Reviews. Genetics, 16(11), 641–652. Booth, M. J., Ost, T. W. B., Beraldi, D., Bell, N. M., Branco, M. R., Reik, W., & Balasubramanian, S. (2013a). Oxidative bisulfite sequencing of 5-methylcytosine and 5-hydroxymethylcytosine. Nature Protocols, 8(10), 1841–1851. Booth, M. J., Ost, T. W. B., Beraldi, D., Bell, N. M., Branco, M. R., Reik, W., & Balasubramanian, S. (2013b). Oxidative bisulfite sequencing of 5-methylcytosine and 5-hydroxymethylcytosine. Nature Protocols, 8(10), 1841–1851. Browne, H. A., Gair, S. L., Scharf, J. M., & Grice, D. E. (2014). Genetics of obsessive-compulsive disorder and related disorders. The Psychiatric Clinics of North America, 37(3), 319–335. BTBD3 BTB domain containing 3 [Homo sapiens (human)] - Gene - NCBI. (n.d.). Retrieved May 29, 2017, from https://www.ncbi.nlm.nih.gov/gene?cmd=retrieve&list_uids=22903 Burgess, N., Maguire, E. A., & O’Keefe, J. (2002). The Human Hippocampus and Spatial and Episodic Memory. Neuron, 35(4), 625–641. Champagne, F. A., Weaver, I. C. G., Diorio, J., Dymov, S., Szyf, M., & Meaney, M. J. (2006). Maternal care associated with methylation of the estrogen receptor-alpha1b promoter and estrogen receptor-alpha expression in the medial preoptic area of female offspring. Endocrinology, 147(6), 2909–2915. Chen, P.-C., Bhattacharyya, B. J., Hanna, J., Minkel, H., Wilson, J. A., Finley, D., … Wilson, S. M. (2011). Ubiquitin homeostasis is critical for synaptic development and function. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience, 31(48), 17505–17513. Clark, S. J., Statham, A., Stirzaker, C., Molloy, P. L., & Frommer, M. (2006). DNA methylation: Bisulphite modification and analysis. Nature Protocols, 1(5), 2353–2364. Cnattingius, S., Zack, M. M., Ekbom, A., Gunnarskog, J., Kreuger, A., Linet, M., & Adami, H.-O. (1995). Prenatal and Neonatal Risk Factors for Childhood Lymphatic Leukemia. JNCI Journal of the National Cancer Institute, 87(12), 908–914. Collewijn, H. (1975). Oculomotor areas in the rabbit’s midbrain and pretectum. Journal of Neurobiology, 6(1), 3–22. Costa, R. M., Honjo, T., & Silva, A. J. (2003). Learning and memory deficits in Notch mutant mice. Current Biology: CB, 13(15), 1348–1354. de Zwaan, M., Barbara, G., Astrid, M., Holmer, G., Alexandra, M., Heide, G., … Alexandra, P. (2011). The estimated prevalence and correlates of adult ADHD in a German community sample. European Archives of Psychiatry and Clinical Neuroscience, 262(1), 79–86. de Zwaan, M., Gruß, B., Müller, A., Graap, H., Martin, A., Glaesmer, H., … Philipsen, A. (2011). The estimated prevalence and correlates of adult ADHD in a German community sample. European Archives of Psychiatry and Clinical Neuroscience, 262(1), 79–86. Dias, B. G., & Ressler, K. J. (2014a). Parental olfactory experience influences behavior and neural structure in subsequent generations. Nature Neuroscience, 17(1), 89–96. Di Ruscio, A., Ebralidze, A. K., Benoukraf, T., Amabile, G., Goff, L. A., Terragni, J., … Tenen, D. G. (2013). DNMT1-interacting RNAs block gene-specific DNA methylation. Nature, 503(7476), 371–376. Dorph-Petersen, K.-A., & Lewis, D. A. (2017). Postmortem structural studies of the thalamus in schizophrenia. Schizophrenia Research, 180, 28–35. Du, J., Johnson, L. M., Jacobsen, S. E., & Patel, D. J. (2015). DNA methylation pathways and their crosstalk with histone methylation. Nature Reviews. Molecular Cell Biology, 16(9), 519–532. Durant, K., & Wagner, S. (2017). On the distribution of betweenness centrality in random trees. Theoretical Computer Science, 699, 33–52. Eiland, L., Ramroop, J., Hill, M. N., Manley, J., & McEwen, B. S. (2012). Chronic juvenile stress produces corticolimbic dendritic architectural remodeling and modulates emotional behavior in male and female rats. Psychoneuroendocrinology, 37(1), 39–47. Eiland, L., & Romeo, R. D. (2013). Stress and the developing adolescent brain. Neuroscience, 249, 162–171. Elliott, E., Ezra-Nevo, G., Regev, L., Neufeld-Cohen, A., & Chen, A. (2010). Resilience to social stress coincides with functional DNA methylation of the Crf gene in adult mice. Nature Neuroscience, 13(11), 1351–1353. Estrada, E. (2012). The Structure of Complex Networks: Theory and Applications. Oxford University Press. Faini, M., Beck, R., Wieland, F. T., & Briggs, J. A. G. (2013). Vesicle coats: structure, function, and general principles of assembly. Trends in Cell Biology, 23(6), 279–288. Feng, J., Jian, F., Yu, Z., Campbell, S. L., Thuc, L., En, L., … Guoping, F. (2010). Dnmt1 and Dnmt3a maintain DNA methylation and regulate synaptic function in adult forebrain neurons. Nature Neuroscience, 13(4), 423–430. Fiuza, U.-M., & Arias, A. M. (2007). Cell and molecular biology of Notch. The Journal of Endocrinology, 194(3), 459–474. Flach, P. (2012). Machine Learning: The Art and Science of Algorithms that Make Sense of Data. Cambridge University Press. Forget, H., Hélène, F., André, L., Isabelle, B., & Henri, C. (2016). Long-term cognitive effects of glucocorticoid excess in Cushing’s syndrome. Psychoneuroendocrinology, 65, 26–33. Franklin, T. B., Russig, H., Weiss, I. C., Gräff, J., Linder, N., Michalon, A., … Mansuy, I. M. (2010a). Epigenetic transmission of the impact of early stress across generations. Biological Psychiatry, 68(5), 408–415. Fu, J., Ivy Yu, H.-M., Maruyama, T., Mirando, A. J., & Hsu, W. (2011). Gpr177/mouse Wntless is essential for Wnt-mediated craniofacial and brain development. Developmental Dynamics: An Official Publication of the American Association of Anatomists, 240(2), 365–371. Fu, J., Jiang, M., Mirando, A. J., Yu, H.-M. I., & Hsu, W. (2009). Reciprocal regulation of Wnt and Gpr177/mouse Wntless is required for embryonic axis formation. Proceedings of the National Academy of Sciences of the United States of America, 106(44), 18598–18603. Gardiner-Garden, M., & Frommer, M. (1987a). CpG Islands in vertebrate genomes. Journal of Molecular Biology, 196(2), 261–282. Gene Ontology Consortium. (n.d.). AmiGO 2: Search. Retrieved December 26, 2019, from http://amigo.geneontology.org/amigo/search/ontology?q=Catabolic%20process%20of%20cellular%20protein,%20catabolic%20process%20of%20protein-mediated%20ubiquitin-dependent%20protein Giniger, E. (1998). A role for Abl in Notch signaling. Neuron, 20(4), 667–681. Grandjean, V., Gounon, P., Wagner, N., Martin, L., Wagner, K. D., Bernex, F., … Rassoulzadegan, M. (2009a). The miR-124-Sox9 paramutation: RNA-mediated epigenetic control of embryonic and adult growth. Development , 136(21), 3647–3655. Griffiths, B. B., & Hunter, R. G. (2014). Neuroepigenetics of stress. Neuroscience, 275, 420–435. Grodd, W., Hülsmann, E., Lotze, M., Wildgruber, D., & Erb, M. (2001). Sensorimotor mapping of the human cerebellum: fMRI evidence of somatotopic organization. Human Brain Mapping, 13(2), 55–73. Hackett, J. A., & Surani, M. A. (2013a). DNA methylation dynamics during the mammalian life cycle. Philosophical Transactions of the Royal Society of London. Series B, Biological Sciences, 368(1609), 20110328. Hackett, J. A., & Surani, M. A. (2013c). DNA methylation dynamics during the mammalian life cycle. Philosophical Transactions of the Royal Society of London. Series B, Biological Sciences, 368(1609), 20110328. Hackett, J. A., & Surani, M. A. (2013d). DNA methylation dynamics during the mammalian life cycle. Philosophical Transactions of the Royal Society of London. Series B, Biological Sciences, 368(1609), 20110328. Harkness, K. L., Stewart, J. G., & Wynne-Edwards, K. E. (2011). Cortisol reactivity to social stress in adolescents: role of depression severity and child maltreatment. Psychoneuroendocrinology, 36(2), 173–181. Hawrylycz, M. J., Lein, E. S., Guillozet-Bongaarts, A. L., Shen, E. H., Ng, L., Miller, J. A., … Jones, A. R. (2012). An anatomically comprehensive atlas of the adult human brain transcriptome. Nature, 489(7416), 391–399. Hegarty, J. P., 2nd, Gu, M., Spielman, D. M., Cleveland, S. C., Hallmayer, J. F., Lazzeroni, L. C., … Hardan, A. Y. (2018). A proton MR spectroscopy study of the thalamus in twins with autism spectrum disorder. Progress in Neuro-Psychopharmacology & Biological Psychiatry, 81, 153–160. Hirvikoski, T., Lindholm, T., Nordenström, A., Nordström, A.-L., & Lajic, S. (2009). High self-perceived stress and many stressors, but normal diurnal cortisol rhythm, in adults with ADHD (attention-deficit/hyperactivity disorder). Hormones and Behavior, 55(3), 418–424. Hirvikoski, T., Tatja, H., Torun, L., Anna, N., Anna-Lena, N., & Svetlana, L. (2009). High self-perceived stress and many stressors, but normal diurnal cortisol rhythm, in adults with ADHD (attention-deficit/hyperactivity disorder). Hormones and Behavior, 55(3), 418–424. Huang, E. J., Li, H., Tang, A. A., Wiggins, A. K., Neve, R. L., Zhong, W., … Jan, Y. N. (2005). Targeted deletion of numb and numblike in sensory neurons reveals their essential functions in axon arborization. Genes & Development, 19(1), 138–151. Human hg19 chr20:11,870,949-11,871,848 UCSC Genome Browser v362. (n.d.). Retrieved March 29, 2018, from https://genome.ucsc.edu/cgi-bin/hgTracks?db=hg19&lastVirtModeType=default&lastVirtModeExtraState=&virtModeType=default&virtMode=0&nonVirtPosition=&position=chr20%3A11870949-11871848&hgsid=660489131_NJVVIFwb4veLsQjFif8oiYPu7Cb9 Huynh, K. D., & Bardwell, V. J. (1998). The BCL-6 POZ domain and other POZ domains interact with the co-repressors N-CoR and SMRT. Oncogene, 17(19), 2473–2484. Igarashi, K., Hoshino, H., Muto, A., Suwabe, N., Nishikawa, S., Nakauchi, H., & Yamamoto, M. (1998). Multivalent DNA binding complex generated by small Maf and Bach1 as a possible biochemical basis for beta-globin locus control region complex. The Journal of Biological Chemistry, 273(19), 11783–11790. Igarashi, K., Kurosaki, T., & Roychoudhuri, R. (2017). BACH transcription factors in innate and adaptive immunity. Nature Reviews. Immunology. https://doi.org/10.1038/nri.2017.26 Jagannath, A., Peirson, S. N., & Foster, R. G. (2013). Sleep and circadian rhythm disruption in neuropsychiatric illness. Current Opinion in Neurobiology, 23(5), 888–894. Jin, P., Alisch, R. S., & Warren, S. T. (2004). RNA and microRNAs in fragile X mental retardation. Nature Cell Biology, 6(11), 1048–1053. Johnson, A. C. (2015a). Developmental pathways to attention-deficit/hyperactivity disorder and disruptive behavior disorders: Investigating the impact of the stress response on executive functioning. Clinical Psychology Review, 36, 1–12. Kelly, K. F., & Daniel, J. M. (2006). POZ for effect--POZ-ZF transcription factors in cancer and development. Trends in Cell Biology, 16(11), 578–587. Kim, J.-M., J.-M., K., To, T. K., & Seki, M. (2012). An Epigenetic Integrator: New Insights into Genome Regulation, Environmental Stress Responses and Developmental Controls by HISTONE DEACETYLASE 6. Plant & Cell Physiology, 53(5), 794–800. Klein, A.-L., Zilian, O., Suter, U., & Taylor, V. (2004). Murine numb regulates granule cell maturation in the cerebellum. Developmental Biology, 266(1), 161–177. Komiya, Y., & Habas, R. (2008). Wnt signal transduction pathways. Organogenesis, 4(2), 68–75. Koubeissi, M. Z., Bartolomei, F., Beltagy, A., & Picard, F. (2014). Electrical stimulation of a small brain area reversibly disrupts consciousness. Epilepsy & Behavior: E&B, 37, 32–35. Kouzarides, T., & Tony, K. (2007). Chromatin Modifications and Their Function. Cell, 128(4), 693–705. Kruskal, W. H., & Wallis, W. A. (1952). Use of Ranks in One-Criterion Variance Analysis. Journal of the American Statistical Association, 47(260), 583–621. Ku, J.-L., Jeon, Y.-K., & Park, J.-G. (2011a). Methylation-Specific PCR. In Methods in Molecular Biology (pp. 23–32 Laht, P., Otsus, M., Remm, J., & Veske, A. (2014). B-plexins control microtubule dynamics and dendrite morphology of hippocampal neurons. Experimental Cell Research, 326(1), 174–184. LaSalle, J. M., Powell, W. T., & Yasui, D. H. (2013a). Epigenetic layers and players underlying neurodevelopment. Trends in Neurosciences, 36(8), 460–470. Lebel, C., Walker, L., Leemans, A., Phillips, L., & Beaulieu, C. (2008). Microstructural maturation of the human brain from childhood to adulthood. NeuroImage, 40(3), 1044–1055. Lerario, A. M., Moraitis, A., & Hammer, G. D. (2014). Genetics and epigenetics of adrenocortical tumors. Molecular and Cellular Endocrinology, 386(1-2), 67–84. Lewin, J., Schmitt, A. O., Adorján, P., Hildmann, T., & Piepenbrock, C. (2004). Quantitative DNA methylation analysis based on four-dye trace data from direct sequencing of PCR amplificates. Bioinformatics , 20(17), 3005–3012. Li, E., & Zhang, Y. (2014). DNA methylation in mammals. Cold Spring Harbor Perspectives in Biology, 6(5), a019133. Limperopoulos, C., Chilingaryan, G., Guizard, N., Robertson, R. L., & Du Plessis, A. J. (2010). Cerebellar injury in the premature infant is associated with impaired growth of specific cerebral regions. Pediatric Research, 68(2), 145–150. Linley, S. B., Gallo, M. M., & Vertes, R. P. (2016). Lesions of the ventral midline thalamus produce deficits in reversal learning and attention on an odor texture set shifting task. Brain Research, 1649(Pt A), 110–122. Lucassen, P. J., Bosch, O. J., Jousma, E., Krömer, S. A., Andrew, R., Seckl, J. R., & Neumann, I. D. (2009). Prenatal stress reduces postnatal neurogenesis in rats selectively bred for high, but not low, anxiety: possible key role of placental 11beta-hydroxysteroid dehydrogenase type 2. The European Journal of Neuroscience, 29(1), 97–103. Ma, L., Liang, M., Yan-Hui, C., Hui, C., Yan-Yan, L., & Yan-Xia, W. (2011). The function of hypothalamus–pituitary–adrenal axis in children with ADHD. Brain Research, 1368, 159–162. Massat, I., Slama, H., Kavec, M., Linotte, S., Mary, A., Baleriaux, D., … Peigneux, P. (2012). Working memory-related functional brain patterns in never medicated children with ADHD. PloS One, 7(11), e49392. Matsui, A., Tran, M., Yoshida, A. C., Kikuchi, S. S., U, M., Ogawa, M., & Shimogori, T. (2013). BTBD3 controls dendrite orientation toward active axons in mammalian neocortex. Science, 342(6162), 1114–1118. Mattfeld, A. T., Whitfield-Gabrieli, S., Biederman, J., Spencer, T., Brown, A., Fried, R., & Gabrieli, J. D. E. (2016). Dissociation of working memory impairments and attention-deficit/hyperactivity disorder in the brain. NeuroImage. Clinical, 10, 274–282. Mattick, J. S. (2004). Opinion: RNA regulation: a new genetics? Nature Reviews. Genetics, 5(4), 316–323. Maunakea, A. K., Chepelev, I., Cui, K., & Zhao, K. (2013). Intragenic DNA methylation modulates alternative splicing by recruiting MeCP2 to promote exon recognition. Cell Research, 23(11), 1256–1269. McEwen, B. S., Gray, J., & Nasca, C. (2015). Recognizing Resilience: Learning from the Effects of Stress on the Brain. Neurobiology of Stress, 1, 1–11. Medin, T., Rinholm, J. E., Owe, S. G., Sagvolden, T., Gjedde, A., Storm-Mathisen, J., & Bergersen, L. H. (2013). Low dopamine D5 receptor density in hippocampus in an animal model of attention-deficit/hyperactivity disorder (ADHD). Neuroscience, 242, 11–20. Messaoudi, I., Young, J. E., Colman, R. J., Handy, A. M., Roth, G. S., Ingram, D. K., & Mattison, J. A. (2010). Aging and the Effect of Calorie Restriction in Rhesus Monkeys. Calorie Restriction, Aging and Longevity, pp. 55–78. https://doi.org/10.1007/978-90-481-8556-6_4 Mesulam, M.-M. (2000). Principles of Behavioral and Cognitive Neurology. Oxford University Press. MethPrimer-Design MSP/BSP primers and predict CpG islands - Li Lab, PUMCH. (n.d.). Retrieved September 12, 2016, from http://www.urogene.org/cgi-bin/methprimer/methprimer.cgi Mifsud, K. R., María, G.-M., Trollope, A. F., Andrew, C., Saunderson, E. A., & Reul, J. M. H. M. (2011). Epigenetic mechanisms in stress and adaptation. Brain, Behavior, and Immunity, 25(7), 1305–1315. Millan, M. J. (2013a). An epigenetic framework for neurodevelopmental disorders: from pathogenesis to potential therapy. Neuropharmacology, 68, 2–82. Miller, A. M., Miller, R. B., Obermeyer, W. H., Behan, M., & Benca, R. M. (1999). The pretectum mediates rapid eye movement sleep regulation by light. Behavioral Neuroscience, 113(4), 755–765. Miller, J. A., Ding, S.-L., Sunkin, S. M., Smith, K. A., Ng, L., Szafer, A., … Lein, E. S. (2014). Transcriptional landscape of the prenatal human brain. Nature, 508(7495), 199–206. Mori, F., Nishie, M., Piao, Y.-S., Kito, K., Kamitani, T., Takahashi, H., & Wakabayashi, K. (2005). Accumulation of NEDD8 in neuronal and glial inclusions of neurodegenerative disorders. Neuropathology and Applied Neurobiology, 31(1), 53–61. Murgatroyd, C., & Spengler, D. (2014). Polycomb binding precedes early-life stress responsive DNA methylation at the Avp enhancer. PloS One, 9(3), e90277. Nigg, J. T., Hill Goldsmith, H., & Jennifer, S. (2004). Temperament and Attention Deficit Hyperactivity Disorder: The Development of a Multiple Pathway Model. Journal of Clinical Child and Adolescent Psychology: The Official Journal for the Society of Clinical Child and Adolescent Psychology, American Psychological Association, Division 53, 33(1), 42–53. Nishida, M., Pearsall, J., Buckner, R. L., & Walker, M. P. (2009). REM sleep, prefrontal theta, and the consolidation of human emotional memory. Cerebral Cortex , 19(5), 1158–1166. Nussbaum, R. L., McInnes, R. R., & Willard, H. F. (2007). Preface. In Thompson & Thompson Genetics in Medicine (p. v). Nussbaum, R. L., McInnes, R. R., & Willard, H. F. (2015). Thompson & Thompson Genetics in Medicine. Elsevier Health Sciences. Okano, M. (1998). Dnmt2 is not required for de novo and maintenance methylation of viral DNA in embryonic stem cells. Nucleic Acids Research, 26(11), 2536–2540. Oktafiani, D. (n.d.). DNA extraction by Wizard® Genomic DNA Purification Kit v1 (protocols.io.7j6hkre). Protocols.io. https://doi.org/10.17504/protocols.io.7j6hkre O’Mara, S. (2006). Controlling hippocampal output: the central role of subiculum in hippocampal information processing. Behavioural Brain Research, 174(2), 304–312. Pandey, G. N., Rizavi, H. S., Ren, X., Dwivedi, Y., & Palkovits, M. (2013). Region-specific alterations in glucocorticoid receptor expression in the postmortem brain of teenage suicide victims. Psychoneuroendocrinology, 38(11), 2628–2639. Patterson, K., Molloy, L., Qu, W., & Clark, S. (2011). DNA methylation: bisulphite modification and analysis. Journal of Visualized Experiments: JoVE, (56). https://doi.org/10.3791/3170 Provençal, N., & Binder, E. B. (2015). The effects of early life stress on the epigenome: From the womb to adulthood and even before. Experimental Neurology, 268, 10–20. Rando, O. J. (2012). Combinatorial complexity in chromatin structure and function: revisiting the histone code. Current Opinion in Genetics & Development, 22(2), 148–155. Rangarajan, B., Suresh, S., & Mahanand, B. S. (2014). Identification of potential biomarkers in the hippocampus region for the diagnosis of ADHD using PBL-McRBFN approach. 2014 13th International Conference on Control Automation Robotics & Vision (ICARCV). https://doi.org/10.1109/icarcv.2014.7064272 Raz, S., & Leykin, D. (2015). Psychological and cortisol reactivity to experimentally induced stress in adults with ADHD. Psychoneuroendocrinology, 60, 7–17. RefSeq: NCBI Reference Sequence Database. (n.d.). Retrieved March 28, 2017, from https://www.ncbi.nlm.nih.gov/refseq/ Reichardt, J., & Bornholdt, S. (2006). Statistical mechanics of community detection. Physical Review. E, Statistical, Nonlinear, and Soft Matter Physics, 74(1 Pt 2), 016110. Rice, J. C., & Allis, C. D. (2001). Histone methylation versus histone acetylation: new insights into epigenetic regulation. Current Opinion in Cell Biology, 13(3), 263–273. Romanos, M., Freitag, C., Jacob, C., Craig, D. W., Dempfle, A., Nguyen, T. T., … Lesch, K. P. (2008). Genome-wide linkage analysis of ADHD using high-density SNP arrays: novel loci at 5q13.1 and 14q12. Molecular Psychiatry, 13(5), 522–530. Roth, T. L., Lubin, F. D., Funk, A. J., & Sweatt, J. D. (2009). Lasting epigenetic influence of early-life adversity on the BDNF gene. Biological Psychiatry, 65(9), 760–769. Salinas, P. C. (2007). Modulation of the microtubule cytoskeleton: a role for a divergent canonical Wnt pathway. Trends in Cell Biology, 17(7), 333–342. Schlinzig, T., Johansson, S., Gunnar, A., Ekström, T. J., & Norman, M. (2009). Epigenetic modulation at birth - altered DNA-methylation in white blood cells after Caesarean section. Acta Paediatrica, 98(7), 1096–1099. Schroll, H., Horn, A., Gröschel, C., Brücke, C., Lütjens, G., Schneider, G.-H., … Hamker, F. H. (2015). Differential contributions of the globus pallidus and ventral thalamus to stimulus–response learning in humans. NeuroImage, 122, 233–245. Shaw, P., Eckstrand, K., Sharp, W., Blumenthal, J., Lerch, J. P., Greenstein, D., … Rapoport, J. L. (2007). Attention-deficit/hyperactivity disorder is characterized by a delay in cortical maturation. Proceedings of the National Academy of Sciences of the United States of America, 104(49), 19649–19654. Siggs, O. M., & Beutler, B. (2012). The BTB-ZF transcription factors. Cell Cycle , 11(18), 3358–3369. Siprashvili, Z., Zurab, S., Webster, D. E., Markus, K., Danielle, J., Rinn, J. L., … Khavari, P. A. (2012). Identification of proteins binding coding and non-coding human RNAs using protein microarrays. BMC Genomics, 13(1), 633. Soloway, P. D. (2006). [Review of Genetics: paramutable possibilities]. Nature, 441(7092), 413–414. Stankiewicz, A. M., Swiergiel, A. H., & Lisowski, P. (2013). Epigenetics of stress adaptations in the brain. Brain Research Bulletin, 98, 76–92. Stewart, S. E., Yu, D., Scharf, J. M., Neale, B. M., Fagerness, J. A., Mathews, C. A., … Pauls, D. L. (2013). Genome-wide association study of obsessive-compulsive disorder. Molecular Psychiatry, 18(7), 788–798. Stogios, P. J., Chen, L., & Privé, G. G. (2007). Crystal structure of the BTB domain from the LRF/ZBTB7 transcriptional regulator. Protein Science: A Publication of the Protein Society, 16(2), 336–342. Stogios, P. J., Downs, G. S., Jauhal, J. J. S., Nandra, S. K., & Privé, G. G. (2005). Sequence and structural analysis of BTB domain proteins. Genome Biology, 6(10), R82. Stoodley, C. J., & Limperopoulos, C. (2016). Structure-function relationships in the developing cerebellum: Evidence from early-life cerebellar injury and neurodevelopmental disorders. Seminars in Fetal & Neonatal Medicine, 21(5), 356–364. Stoodley, C. J., & Schmahmann, J. D. (2010). Evidence for topographic organization in the cerebellum of motor control versus cognitive and affective processing. Cortex; a Journal Devoted to the Study of the Nervous System and Behavior, 46(7), 831–844. Szklarczyk, D., Morris, J. H., Cook, H., Kuhn, M., Wyder, S., Simonovic, M., … von Mering, C. (2017). The STRING database in 2017: quality-controlled protein-protein association networks, made broadly accessible. Nucleic Acids Research, 45(D1), D362–D368. Tahiliani, M., Koh, K. P., Shen, Y., Pastor, W. A., Bandukwala, H., Brudno, Y., … Rao, A. (2009). Conversion of 5-methylcytosine to 5-hydroxymethylcytosine in mammalian DNA by MLL partner TET1. Science, 324(5929), 930–935. Thapar, A., & Cooper, M. (2016). Attention deficit hyperactivity disorder. The Lancet, 387(10024), 1240–1250. Thapar, A., Cooper, M., Eyre, O., & Langley, K. (2013a). What have we learnt about the causes of ADHD? Journal of Child Psychology and Psychiatry, and Allied Disciplines, 54(1), 3–16. Thapar, A., Cooper, M., Eyre, O., & Langley, K. (2013c). What have we learnt about the causes of ADHD? Journal of Child Psychology and Psychiatry, and Allied Disciplines, 54(1), 3–16. Tsigos, C., Constantine, T., & Chrousos, G. P. (2002). Hypothalamic–pituitary–adrenal axis, neuroendocrine factors and stress. Journal of Psychosomatic Research, 53(4), 865–871. Tusnády, G. E., Simon, I., Váradi, A., & Arányi, T. (2005). BiSearch: primer-design and search tool for PCR on bisulfite-treated genomes. Nucleic Acids Research, 33(1), e9. van der Knaap, L. J., Oldehinkel, A. J., Verhulst, F. C., van Oort, F. V. A., & Riese, H. (2015). Glucocorticoid receptor gene methylation and HPA-axis regulation in adolescents. The TRAILS study. Psychoneuroendocrinology, 58, 46–50. van der Knaap, L. J., Riese, H., Hudziak, J. J., Verbiest, M. M. P. J., Verhulst, F. C., Oldehinkel, A. J., & van Oort, F. V. A. (2014). Glucocorticoid receptor gene (NR3C1) methylation following stressful events between birth and adolescence. The TRAILS study. Translational Psychiatry, 4, e381. Vukojevic, V., Kolassa, I.-T., Fastenrath, M., Gschwind, L., Spalek, K., Milnik, A., … de Quervain, D. J.-F. (2014). Epigenetic modification of the glucocorticoid receptor gene is linked to traumatic memory and post-traumatic stress disorder risk in genocide survivors. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience, 34(31), 10274–10284. Waddington, C. H. (1956). Embryology, Epigenetics and Biogenetics. Nature, 177(4522), 1241–1241. Wang, C.-F., Hsing, H.-W., Zhuang, Z.-H., Wen, M.-H., Chang, W.-J., Briz, C. G., … Chou, S.-J. (2017). Lhx2 Expression in Postmitotic Cortical Neurons Initiates Assembly of the Thalamocortical Somatosensory Circuit. Cell Reports, 18(4), 849–856. Weaver, I. C. G., Cervoni, N., Champagne, F. A., D’Alessio, A. C., Sharma, S., Seckl, J. R., … Meaney, M. J. (2004). Epigenetic programming by maternal behavior. Nature Neuroscience, 7(8), 847–854. Whelan, C. D., Hibar, D. P., van Velzen, L. S., Zannas, A. S., Carrillo-Roa, T., McMahon, K., … Alzheimer’s Disease Neuroimaging Initiative. (2016). Heritability and reliability of automatically segmented human hippocampal formation subregions. NeuroImage, 128, 125–137. Wolf, O. T. (2003). HPA axis and memory. Best Practice & Research. Clinical Endocrinology & Metabolism, 17(2), 287–299. Wolfram|Alpha: Making the world’s knowledge computable. (n.d.). Retrieved April 10, 2018, from www.wolframalpha.com/ Wulff, K., Gatti, S., Wettstein, J. G., & Foster, R. G. (2010). Sleep and circadian rhythm disruption in psychiatric and neurodegenerative disease. Nature Reviews. Neuroscience, 11(8), 589–599. Wu, S. C., & Zhang, Y. (2010). Active DNA demethylation: many roads lead to Rome. Nature Reviews. Molecular Cell Biology, 11(9), 607–620. Yang, B.-Z., Zhang, H., Ge, W., Weder, N., Douglas-Palumberi, H., Perepletchikova, F., … Kaufman, J. (2013). Child abuse and epigenetic mechanisms of disease risk. American Journal of Preventive Medicine, 44(2), 101–107. Yehuda, R., Flory, J. D., Bierer, L. M., Henn-Haase, C., Lehrner, A., Desarnaud, F., … Meaney, M. J. (2015). Lower methylation of glucocorticoid receptor gene promoter 1F in peripheral blood of veterans with posttraumatic stress disorder. Biological Psychiatry, 77(4), 356–364. Yektaei-Karin, E., Moshfegh, A., Lundahl, J., Berggren, V., Hansson, L.-O., & Marchini, G. (2007). The stress of birth enhances in vitro spontaneous and IL-8-induced neutrophil chemotaxis in the human newborn. Pediatric Allergy and Immunology: Official Publication of the European Society of Pediatric Allergy and Immunology, 18(8), 643–651. Yoon, K., & Gaiano, N. (2005). Notch signaling in the mammalian central nervous system: insights from mouse mutants. Nature Neuroscience, 8(6), 709–715. Yoshida, C., Tokumasu, F., Hohmura, K. I., Bungert, J., Hayashi, N., Nagasawa, T., … Igarashi, K. (1999). Long range interaction of cis-DNA elements mediated by architectural transcription factor Bach1. Genes to Cells: Devoted to Molecular & Cellular Mechanisms, 4(11), 643–655. Yue, W., Cheng, W., Liu, Z., Tang, Y., Lu, T., Zhang, D., … Huang, Y. (2016). Genome-wide DNA methylation analysis in obsessive-compulsive disorder patients. Scientific Reports, 6, 31333. Yung, P. Y. K., Stuetzer, A., Fischle, W., Martinez, A.-M., & Cavalli, G. (2015). Histone H3 Serine 28 Is Essential for Efficient Polycomb-Mediated Gene Repression in Drosophila. Cell Reports, 11(9), 1437–1445. Zhang, L., Xu, Y., Zhuang, J., Peng, H., Wu, H., Zhao, Z., … Zhao, Z. (2016). Metabolic abnormality of pontine tegmentum in patients with REM sleep behavior disorder analyzed using magnetic resonance spectroscopy. Clinical Neurology and Neurosurgery, 148, 137–141. Zhang, T.-Y., Hellstrom, I. C., Bagot, R. C., Wen, X., Diorio, J., & Meaney, M. J. (2010). Maternal care and DNA methylation of a glutamic acid decarboxylase 1 promoter in rat hippocampus. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience, 30(39), 13130–13137. Zhou, K., Dempfle, A., Arcos-Burgos, M., Bakker, S. C., Banaschewski, T., Biederman, J., … Asherson, P. (2008). Meta-analysis of genome-wide linkage scans of attention deficit hyperactivity disorder. American Journal of Medical Genetics. Part B, Neuropsychiatric Genetics: The Official Publication of the International Society of Psychiatric Genetics, 147B(8), 1392–1398.
dc.rights.accessrightsinfo:eu-repo/semantics/openAccess
dc.subject.proposalADHD
dc.subject.proposalTDAH
dc.subject.proposalMethylation
dc.subject.proposalMetilación
dc.subject.proposalPPi
dc.subject.proposalPPi
dc.subject.proposalBTBD3
dc.subject.proposalBTBD3
dc.subject.proposalTopological destabilization
dc.subject.proposalDesestabilización topológica
dc.subject.proposalBSP PCR
dc.subject.proposalBSP PCR
dc.subject.proposalAnálisis de expresión
dc.subject.proposalExpression analysis
dc.type.coarhttp://purl.org/coar/resource_type/c_8042
dc.type.coarversionhttp://purl.org/coar/version/c_ab4af688f83e57aa
dc.type.contentText
dc.type.redcolhttp://purl.org/redcol/resource_type/WP
oaire.accessrightshttp://purl.org/coar/access_right/c_abf2


Archivos en el documento

Thumbnail
Thumbnail

Este documento aparece en la(s) siguiente(s) colección(ones)

Mostrar el registro sencillo del documento

Atribución-NoComercial-SinDerivadas 4.0 InternacionalEsta obra está bajo licencia internacional Creative Commons Reconocimiento-NoComercial 4.0.Este documento ha sido depositado por parte de el(los) autor(es) bajo la siguiente constancia de depósito