Ítem
Acceso Abierto

Identificación de factores de transcripción de unión directa a la región promotora de STOX1

dc.contributor.advisorLaissue, Paul
dc.creatorBello Uyaban, Sandra
dc.creator.degreeMagíster en Ciencias con Énfasis en Genética Humana
dc.date.accessioned2018-06-27T22:21:01Z
dc.date.available2018-06-27T22:21:01Z
dc.date.created2018-06-07
dc.date.issued2018
dc.descriptionLa Preeclampsia (PE) es una enfermedad exclusiva del embarazo caracterizada por hipertensión arterial y compromiso multisistémico. Afecta entre el 2% y el 8% de las mujeres gestantes en el mundo y es la principal causa de morbilidad y mortalidad materna y neonatal. El principal evento relacionado con la fisiopatología de la PE es la disfunción endotelial, la cual depende del control poligénico de múltiples cascadas moleculares. Diversas proteínas han sido estudiadas con el objetivo de esclarecer la fisiopatología de la enfermedad, entre ellas el factor de transcripción STOX1. Esta proteína regula diferentes procesos biológicos implicados en la fisiopatología de la PE como la angiogénesis, el estrés oxidativo, la inflamación y la apoptosis. Sin embargo, se desconoce qué factores de transcripción se fijan al promotor de STOX1 participando en su regulación. Durante el presente trabajo de tesis se propuso identificar los factores que interactúan potencialmente con la región promotora de STOX1 e identificar, por medio de secuenciación del promotor, variantes que puedan afectar sitios de unión a factores de transcripción en una muestra de pacientes francesas afectadas con PE. Para cumplir este objetivo fueron utilizadas técnicas experimentales in vivo como el ensayo de monohíbrido en levaduras y bioinformáticas in silico. Se identificó al factor de transcripción HEY-L como un potencial regulador de STOX1 y fue encontrada la variante c.-735T>G en la región donde potencialmente interactúa este factor de transcripción. Sin embargo, deben ser realizados estudios funcionales para validar la interacción definitiva y si la mutación genera una modificación en la transactivación del promotor.spa
dc.format.mimetypeapplication/pdf
dc.identifier.doihttps://doi.org/10.48713/10336_18135
dc.identifier.urihttp://repository.urosario.edu.co/handle/10336/18135
dc.language.isospa
dc.publisherUniversidad del Rosariospa
dc.publisher.departmentFacultad de medicinaspa
dc.publisher.programMaestría en Ciencias con Énfasis en Genética Humanaspa
dc.rightsAtribución-NoComercial-SinDerivadas 2.5 Colombiaspa
dc.rights.accesRightsinfo:eu-repo/semantics/openAccess
dc.rights.accesoAbierto (Texto Completo)spa
dc.rights.licenciaEL AUTOR, manifiesta que la obra objeto de la presente autorización es original y la realizó sin violar o usurpar derechos de autor de terceros, por lo tanto la obra es de exclusiva autoría y tiene la titularidad sobre la misma. PARGRAFO: En caso de presentarse cualquier reclamación o acción por parte de un tercero en cuanto a los derechos de autor sobre la obra en cuestión, EL AUTOR, asumirá toda la responsabilidad, y saldrá en defensa de los derechos aquí autorizados; para todos los efectos la universidad actúa como un tercero de buena fe. EL AUTOR, autoriza a LA UNIVERSIDAD DEL ROSARIO, para que en los términos establecidos en la Ley 23 de 1982, Ley 44 de 1993, Decisión andina 351 de 1993, Decreto 460 de 1995 y demás normas generales sobre la materia, utilice y use la obra objeto de la presente autorización. -------------------------------------- POLITICA DE TRATAMIENTO DE DATOS PERSONALES. Declaro que autorizo previa y de forma informada el tratamiento de mis datos personales por parte de LA UNIVERSIDAD DEL ROSARIO para fines académicos y en aplicación de convenios con terceros o servicios conexos con actividades propias de la academia, con estricto cumplimiento de los principios de ley. Para el correcto ejercicio de mi derecho de habeas data cuento con la cuenta de correo habeasdata@urosario.edu.co, donde previa identificación podré solicitar la consulta, corrección y supresión de mis datos.spa
dc.rights.urihttp://creativecommons.org/licenses/by-nc-nd/2.5/co/
dc.source.bibliographicCitationAli, S. M., & Khalil, R. A. (2015). Genetic, immune and vasoactive factors in the vascular dysfunction associated with hypertension in pregnancy. Expert Opin Ther Targets, 19(11), 1495-1515. doi:10.1517/14728222.2015.1067684
dc.source.bibliographicCitationAmaral, L. M., Cunningham, M. W., Cornelius, D. C., & LaMarca, B. (2015). Preeclampsia: long-term consequences for vascular health. Vascular Health and Risk Management, 11, 403-415. doi:10.2147/VHRM.S64798
dc.source.bibliographicCitationBallermann, B. J. (2005). Glomerular endothelial cell differentiation. Kidney Int, 67(5), 1668-1671. doi:10.1111/j.1523-1755.2005.00260.x
dc.source.bibliographicCitationBerends, A. L., Bertoli-Avella, A. M., de Groot, C. J., van Duijn, C. M., Oostra, B. A., & Steegers, E. A. (2007). STOX1 gene in pre-eclampsia and intrauterine growth restriction. Bjog, 114(9), 1163-1167. doi:10.1111/j.1471-0528.2007.01414.x
dc.source.bibliographicCitationBiagi, G., De Rosa, V., Pelusi, G., Scagliarini, G., Sani, G., & Coccheri, S. (1990). Increased placental production of leukotriene B4 in gestational hypertension. Thromb Res, 60(5), 377-384.
dc.source.bibliographicCitationBuas, M. F., Kabak, S., & Kadesch, T. (2010). The Notch effector Hey1 associates with myogenic target genes to repress myogenesis. J Biol Chem, 285(2), 1249-1258. doi:10.1074/jbc.M109.046441
dc.source.bibliographicCitationCaburet, S., Zavadakova, P., Ben-Neriah, Z., Bouhali, K., Dipietromaria, A., Charon, C., . . . Fellous, M. (2012). Genome-Wide Linkage in a Highly Consanguineous Pedigree Reveals Two Novel Loci on Chromosome 7 for Non-Syndromic Familial Premature Ovarian Failure. PLoS ONE, 7(3), e33412. doi:10.1371/journal.pone.0033412
dc.source.bibliographicCitationCarlosama, C., Elzaiat, M., Patino, L. C., Mateus, H. E., Veitia, R. A., & Laissue, P. (2017). A homozygous donor splice-site mutation in the meiotic gene MSH4 causes primary ovarian insufficiency. Hum Mol Genet, 26(16), 3161-3166. doi:10.1093/hmg/ddx199
dc.source.bibliographicCitationCartharius, K., Frech, K., Grote, K., Klocke, B., Haltmeier, M., Klingenhoff, A., . . . Werner, T. (2005). MatInspector and beyond: promoter analysis based on transcription factor binding sites. Bioinformatics, 21(13), 2933-2942. doi:10.1093/bioinformatics/bti473
dc.source.bibliographicCitationCartharius, K., Frech, K., Karas, H., Wingender, E., & Werner, T. (1995). Quandt K, Frech K, Karas H, Wingender E, Werner T.. MatInd and MatInspector: new fast and versatile tools for detection of consensus matches in nucleotide sequence data. Nucl Acids Res 23: 4878-4884 (Vol. 23)
dc.source.bibliographicCitationCarty, D. M., Delles, C., & Dominiczak, A. F. (2008). Novel biomarkers for predicting preeclampsia. Trends Cardiovasc Med, 18(5), 186-194. doi:10.1016/j.tcm.2008.07.002
dc.source.bibliographicCitationCastro, T., Mateus, H. E., Fonseca, D. J., Forero, D., Restrepo, C. M., Talero, C., . . . Laissue, P. (2013). Sequence analysis of the ADRA2A coding region in children affected by attention deficit hyperactivity disorder. Neurol Sci, 34(12), 2219-2222. doi:10.1007/s10072-013-1569-4
dc.source.bibliographicCitationCerdeira, A. S., & Karumanchi, S. A. (2012). Angiogenic factors in preeclampsia and related disorders. Cold Spring Harb Perspect Med, 2(11). doi:10.1101/cshperspect.a006585
dc.source.bibliographicCitationChen, C. W., Jaffe, I. Z., & Karumanchi, S. A. (2014). Pre-eclampsia and cardiovascular disease. Cardiovascular Research, 101(4), 579-586. doi:10.1093/cvr/cvu018
dc.source.bibliographicCitationCraici, I. M., Wagner, S. J., Weissgerber, T. L., Grande, J. P., & Garovic, V. D. (2014). Advances in the pathophysiology of pre-eclampsia and related podocyte injury. Kidney international, 86(2), 275-285. doi:10.1038/ki.2014.17
dc.source.bibliographicCitationDekker, G. A., & Sibai, B. M. (1998). Etiology and pathogenesis of preeclampsia: current concepts. Am J Obstet Gynecol, 179(5), 1359-1375.
dc.source.bibliographicCitationDeplancke, B., Vermeirssen, V., Arda, H. E., Martinez, N. J., & Walhout, A. J. (2006). Gateway-compatible yeast one-hybrid screens. CSH Protoc, 2006(5). doi:10.1101/pdb.prot4590
dc.source.bibliographicCitationDiggle, C. P., Parry, D. A., Logan, C. V., Laissue, P., Rivera, C., Restrepo, C. M., . . . Bonthron, D. T. (2012). Prostaglandin transporter mutations cause pachydermoperiostosis with myelofibrosis. Hum Mutat, 33(8), 1175-1181. doi:10.1002/humu.22111
dc.source.bibliographicCitationDoridot, L., Chatre, L., Ducat, A., Vilotte, J. L., Lombes, A., Mehats, C., . . . Vaiman, D. (2014). Nitroso-redox balance and mitochondrial homeostasis are regulated by STOX1, a pre-eclampsia-associated gene. Antioxid Redox Signal, 21(6), 819-834. doi:10.1089/ars.2013.5661
dc.source.bibliographicCitationDoridot, L., Passet, B., Mehats, C., Rigourd, V., Barbaux, S., Ducat, A., . . . Vaiman, D. (2013). Preeclampsia-like symptoms induced in mice by fetoplacental expression of STOX1 are reversed by aspirin treatment. Hypertension, 61(3), 662-668. doi:10.1161/hypertensionaha.111.20299
dc.source.bibliographicCitationDucat, A., Doridot, L., Calicchio, R., Méhats, C., Vilotte, J.-L., Castille, J., . . . Vaiman, D. (2016). Endothelial cell dysfunction and cardiac hypertrophy in the STOX1 model of preeclampsia. Scientific Reports, 6, 19196. doi:10.1038/srep19196
dc.source.bibliographicCitationEiland, E., Nzerue, C., & Faulkner, M. (2012). Preeclampsia 2012. Journal of Pregnancy, 2012, 586578. doi:10.1155/2012/586578
dc.source.bibliographicCitationFonseca, D. J., Garzon, E., Lakhal, B., Braham, R., Ojeda, D., Elghezal, H., . . . Laissue, P. (2012). Screening for mutations of the FOXO4 gene in premature ovarian failure patients. Reprod Biomed Online, 24(3), 339-341. doi:10.1016/j.rbmo.2011.11.017
dc.source.bibliographicCitationFonseca, D. J., Ortega-Recalde, O., Esteban-Perez, C., Moreno-Ortiz, H., Patino, L. C., Bermudez, O. M., . . . Laissue, P. (2014). BMP15 c.-9C>G promoter sequence variant may contribute to the cause of non-syndromic premature ovarian failure. Reprod Biomed Online, 29(5), 627-633. doi:10.1016/j.rbmo.2014.07.018
dc.source.bibliographicCitationFonseca, D. J., Patino, L. C., Suarez, Y. C., de Jesus Rodriguez, A., Mateus, H. E., Jimenez, K. M., . . . Laissue, P. (2015). Next generation sequencing in women affected by nonsyndromic premature ovarian failure displays new potential causative genes and mutations. Fertil Steril, 104(1), 154-162.e152. doi:10.1016/j.fertnstert.2015.04.016
dc.source.bibliographicCitationFonseca, D. J., Prada, C. F., Siza, L. M., Angel, D., Gomez, Y. M., Restrepo, C. M., . . . Laissue, P. (2012). A de novo 14q12q13.3 interstitial deletion in a patient affected by a severe neurodevelopmental disorder of unknown origin. Am J Med Genet A, 158a(3), 689-693. doi:10.1002/ajmg.a.35215
dc.source.bibliographicCitationFonseca, D. J., Rojas, R. F., Vergara, J. I., Rios, X., Uribe, C., Chavez, L., . . . Laissue, P. (2013). A severe familial phenotype of Ichthyosis Curth-Macklin caused by a novel mutation in the KRT1 gene. Br J Dermatol, 168(2), 456-458. doi:10.1111/j.1365-2133.2012.11181.x
dc.source.bibliographicCitationForero, D. A., Wonkam, A., Wang, W., Laissue, P., Lopez-Correa, C., Fernandez-Lopez, J. C., . . . Perry, G. (2016). Current needs for human and medical genomics research infrastructure in low and middle income countries. J Med Genet, 53(7), 438-440. doi:10.1136/jmedgenet-2015-103631
dc.source.bibliographicCitationFounds, S. A., Conley, Y. P., Lyons-Weiler, J. F., Jeyabalan, A., Hogge, W. A., & Conrad, K. P. (2009). Altered global gene expression in first trimester placentas of women destined to develop preeclampsia. Placenta, 30(1), 15-24. doi:10.1016/j.placenta.2008.09.015
dc.source.bibliographicCitationFragkiadaki, P., Soulitzis, N., Sifakis, S., Koutroulakis, D., Gourvas, V., Vrachnis, N., & Spandidos, D. A. (2015). Downregulation of notch signaling pathway in late preterm and term placentas from pregnancies complicated by preeclampsia. PLoS One, 10(5), e0126163. doi:10.1371/journal.pone.0126163
dc.source.bibliographicCitationGeorge, E. M. (2014). New approaches for managing preeclampsia: clues from clinical and basic research. Clin Ther, 36(12), 1873-1881. doi:10.1016/j.clinthera.2014.09.023
dc.source.bibliographicCitationGeorge, E. M., & Bidwell, G. L. (2013). STOX1: A new player in preeclampsia? Hypertension, 61(3), 561-563. doi:10.1161/HYPERTENSIONAHA.111.00721
dc.source.bibliographicCitationGoel, A., & Rana, S. (2013). Angiogenic factors in preeclampsia: potential for diagnosis and treatment. Curr Opin Nephrol Hypertens, 22(6), 643-650. doi:10.1097/MNH.0b013e328365ad98
dc.source.bibliographicCitationGraves, J. A. (1998). Genomic imprinting, development and disease--is pre-eclampsia caused by a maternally imprinted gene? Reprod Fertil Dev, 10(1), 23-29
dc.source.bibliographicCitationGrill, S., Rusterholz, C., Zanetti-Dallenbach, R., Tercanli, S., Holzgreve, W., Hahn, S., & Lapaire, O. (2009). Potential markers of preeclampsia--a review. Reprod Biol Endocrinol, 7, 70. doi:10.1186/1477-7827-7-70
dc.source.bibliographicCitationGubelmann, C., Waszak, S. M., Isakova, A., Holcombe, W., Hens, K., Iagovitina, A., . . . Deplancke, B. (2013). A yeast one-hybrid and microfluidics-based pipeline to map mammalian gene regulatory networks. Molecular Systems Biology, 9, 682-682. doi:10.1038/msb.2013.38
dc.source.bibliographicCitationGuenther, C., & Garriga, G. (1996). Asymmetric distribution of the C. elegans HAM-1 protein in neuroblasts enables daughter cells to adopt distinct fates. Development, 122(11), 3509-3518.
dc.source.bibliographicCitationHaberle, V., & Lenhard, B. (2016). Promoter architectures and developmental gene regulation. Seminars in Cell & Developmental Biology, 57, 11-23. doi:https://doi.org/10.1016/j.semcdb.2016.01.014
dc.source.bibliographicCitationHahn, S., Lapaire, O., & Than, N. G. (2015). Biomarker development for presymptomatic molecular diagnosis of preeclampsia: feasible, useful or even unnecessary? Expert Rev Mol Diagn, 15(5), 617-629. doi:10.1586/14737159.2015.1025757
dc.source.bibliographicCitationHaram, K., Mortensen, J. H., & Nagy, B. (2014). Genetic Aspects of Preeclampsia and the HELLP Syndrome. Journal of Pregnancy, 2014, 910751. doi:10.1155/2014/910751
dc.source.bibliographicCitationHawfield, A., & Freedman, B. I. (2009). Pre-eclampsia: the pivotal role of the placenta in its pathophysiology and markers for early detection. Ther Adv Cardiovasc Dis, 3(1), 65-73. doi:10.1177/1753944708097114
dc.source.bibliographicCitationHellman, L. M., & Fried, M. G. (2007). Electrophoretic mobility shift assay (EMSA) for detecting protein-nucleic acid interactions. Nat Protoc, 2(8), 1849-1861. doi:10.1038/nprot.2007.249
dc.source.bibliographicCitationJebbink, J., Wolters, A., Fernando, F., Afink, G., van der Post, J., & Ris-Stalpers, C. (2012). Molecular genetics of preeclampsia and HELLP syndrome — A review. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease, 1822(12), 1960-1969. doi:http://dx.doi.org/10.1016/j.bbadis.2012.08.004
dc.source.bibliographicCitationJeyabalan, A. (2013). Epidemiology of preeclampsia: impact of obesity. Nutr Rev, 71 Suppl 1, S18-25. doi:10.1111/nure.12055
dc.source.bibliographicCitationKanasaki, K., & Kalluri, R. (2009). The Biology of Preeclampsia. Kidney international, 76(8), 831-837. doi:10.1038/ki.2009.284
dc.source.bibliographicCitationKivinen, K., Peterson, H., Hiltunen, L., Laivuori, H., Heino, S., Tiala, I., . . . Kere, J. (2007). Evaluation of STOX1 as a preeclampsia candidate gene in a population-wide sample. Eur J Hum Genet, 15(4), 494-497. doi:10.1038/sj.ejhg.5201788
dc.source.bibliographicCitationKleinrouweler, C. E., van Uitert, M., Moerland, P. D., Ris-Stalpers, C., van der Post, J. A., & Afink, G. B. (2013). Differentially expressed genes in the pre-eclamptic placenta: a systematic review and meta-analysis. PLoS One, 8(7), e68991. doi:10.1371/journal.pone.0068991
dc.source.bibliographicCitationKubota, K., Kent, L. N., Rumi, M. A. K., Roby, K. F., & Soares, M. J. (2015). Dynamic Regulation of AP-1 Transcriptional Complexes Directs Trophoblast Differentiation. In Mol Cell Biol (Vol. 35, pp. 3163-3177). 1752 N St., N.W., Washington, DC.
dc.source.bibliographicCitationL'Hote, D., Laissue, P., Serres, C., Montagutelli, X., Veitia, R. A., & Vaiman, D. (2010). Interspecific resources: a major tool for quantitative trait locus cloning and speciation research. Bioessays, 32(2), 132-142. doi:10.1002/bies.200900027
dc.source.bibliographicCitationLaissue, P. (2015). Aetiological coding sequence variants in non-syndromic premature ovarian failure: From genetic linkage analysis to next generation sequencing. Mol Cell Endocrinol, 411, 243-257. doi:10.1016/j.mce.2015.05.005
dc.source.bibliographicCitationLaissue, P. (2018). The molecular complexity of primary ovarian insufficiency aetiology and the use of massively parallel sequencing. Mol Cell Endocrinol, 460, 170-180. doi:10.1016/j.mce.2017.07.021
dc.source.bibliographicCitationLaissue, P., Burgio, G., l'Hote, D., Renault, G., Marchiol-Fournigault, C., Fradelizi, D., . . . Vaiman, D. (2009). Identification of Quantitative Trait Loci responsible for embryonic lethality in mice assessed by ultrasonography. Int J Dev Biol, 53(4), 623-629. doi:10.1387/ijdb.082613pl
dc.source.bibliographicCitationLaissue, P., L'Hote, D., Serres, C., & Vaiman, D. (2009). Mouse models for identifying genes modulating fertility parameters. Animal, 3(1), 55-71. doi:10.1017/s1751731108003315
dc.source.bibliographicCitationLaissue, P., Lakhal, B., Benayoun, B. A., Dipietromaria, A., Braham, R., Elghezal, H., . . . Veitia, R. A. (2009). Functional evidence implicating FOXL2 in non-syndromic premature ovarian failure and in the regulation of the transcription factor OSR2. J Med Genet, 46(7), 455-457. doi:10.1136/jmg.2008.065086
dc.source.bibliographicCitationLaissue, P., Lakhal, B., Vatin, M., Batista, F., Burgio, G., Mercier, E., . . . Vaiman, D. (2016). Association of FOXD1 variants with adverse pregnancy outcomes in mice and humans. Open Biol, 6(10). doi:10.1098/rsob.160109
dc.source.bibliographicCitationLaissue, P., Restrepo, C. M., & Ortiz, A. M. (2017). Improving the evaluation of milestones for students completing a clinical genetics elective. Genet Med, 19(12). doi:10.1038/gim.2017.62
dc.source.bibliographicCitationLakhal, B., Laissue, P., Braham, R., Elghezal, H., Saad, A., Fellous, M., & Veitia, R. A. (2010). BMP15 and premature ovarian failure: causal mutations, variants, polymorphisms? In Clin Endocrinol (Oxf) (Vol. 72, pp. 425-426). England.
dc.source.bibliographicCitationLakhal, B., Laissue, P., Elghezal, H., & Fellous, M. (2008). [Genetic analysis of premature ovarian failure: role of forkhead and TGF-beta genes]. Gynecol Obstet Fertil, 36(9), 862-871. doi:10.1016/j.gyobfe.2008.07.002
dc.source.bibliographicCitationLavery, D. N., Villaronga, M. A., Walker, M. M., Patel, A., Belandia, B., & Bevan, C. L. (2011). Repression of androgen receptor activity by HEYL, a third member of the Hairy/Enhancer-of-split-related family of Notch effectors. J Biol Chem, 286(20), 17796-17808. doi:10.1074/jbc.M110.198655
dc.source.bibliographicCitationLonergan, M., Aponso, D., Marvin, K. W., Helliwell, R. J. A., Sato, T. A., Mitchell, M. D., . . . Keelan, J. A. (2003). Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand (TRAIL), TRAIL Receptors, and the Soluble Receptor Osteoprotegerin in Human Gestational Membranes and Amniotic Fluid during Pregnancy and Labor at Term and Preterm. The Journal of Clinical Endocrinology & Metabolism, 88(8), 3835-3844. doi:10.1210/jc.2002-021905
dc.source.bibliographicCitationLynch, A. M., & Salmon, J. E. (2010). Dysregulated complement activation as a common pathway of injury in preeclampsia and other pregnancy complications. Placenta, 31(7), 561-567. doi:10.1016/j.placenta.2010.03.010
dc.source.bibliographicCitationMaharaj, A. S., Saint-Geniez, M., Maldonado, A. E., & D'Amore, P. A. (2006). Vascular endothelial growth factor localization in the adult. Am J Pathol, 168(2), 639-648. doi:10.2353/ajpath.2006.050834
dc.source.bibliographicCitationMahony, S., & Pugh, B. F. (2015). Protein-DNA binding in high-resolution. Critical reviews in biochemistry and molecular biology, 50(4), 269-283. doi:10.3109/10409238.2015.1051505
dc.source.bibliographicCitationMaier, M. M., & Gessler, M. (2000). Comparative analysis of the human and mouse Hey1 promoter: Hey genes are new Notch target genes. Biochem Biophys Res Commun, 275(2), 652-660. doi:10.1006/bbrc.2000.3354
dc.source.bibliographicCitationMateus, H. E., Perez, A. M., Mesa, M. L., Escobar, G., Galvez, J. M., Montano, J. I., . . . Laissue, P. (2017). A first description of the Colombian national registry for rare diseases. BMC Res Notes, 10(1), 514. doi:10.1186/s13104-017-2840-1
dc.source.bibliographicCitationMaynard, S. E., Min, J. Y., Merchan, J., Lim, K. H., Li, J., Mondal, S., . . . Karumanchi, S. A. (2003). Excess placental soluble fms-like tyrosine kinase 1 (sFlt1) may contribute to endothelial dysfunction, hypertension, and proteinuria in preeclampsia. J Clin Invest, 111(5), 649-658. doi:10.1172/jci17189
dc.source.bibliographicCitationMelchiorre, K., Sharma, R., & Thilaganathan, B. (2014). Cardiovascular implications in preeclampsia: an overview. Circulation, 130(8), 703-714. doi:10.1161/circulationaha.113.003664
dc.source.bibliographicCitationMing, Q., Roske, Y., Schuetz, A., Walentin, K., Ibraimi, I., Schmidt-Ott, K. M., & Heinemann, U. (2018). Structural basis of gene regulation by the Grainyhead/CP2 transcription factor family. Nucleic Acids Res, 46(4), 2082-2095. doi:10.1093/nar/gkx1299
dc.source.bibliographicCitationMitropoulos, K., Al Jaibeji, H., Forero, D. A., Laissue, P., Wonkam, A., Lopez-Correa, C., . . . Patrinos, G. P. (2015). Success stories in genomic medicine from resource-limited countries. Hum Genomics, 9, 11. doi:10.1186/s40246-015-0033-3
dc.source.bibliographicCitationMutter, W. P., & Karumanchi, S. A. (2008). Molecular mechanisms of preeclampsia. Microvasc Res, 75(1), 1-8. doi:10.1016/j.mvr.2007.04.009
dc.source.bibliographicCitationMyatt, L., & Webster, R. P. (2009). Vascular biology of preeclampsia. J Thromb Haemost, 7(3), 375-384. doi:10.1111/j.1538-7836.2008.03259.x
dc.source.bibliographicCitationNaljayan, M. V., & Karumanchi, S. A. (2013). NEW DEVELOPMENTS IN THE PATHOGENESIS OF PREECLAMPSIA. Advances in chronic kidney disease, 20(3), 265-270. doi:10.1053/j.ackd.2013.02.003
dc.source.bibliographicCitationNicolaides, K. H., Bindra, R., Turan, O. M., Chefetz, I., Sammar, M., Meiri, H., . . . Cuckle, H. S. (2006). A novel approach to first-trimester screening for early pre-eclampsia combining serum PP-13 and Doppler ultrasound. Ultrasound Obstet Gynecol, 27(1), 13-17. doi:10.1002/uog.2686
dc.source.bibliographicCitationNino, M. Y., Mateus, H. E., Fonseca, D. J., Kroos, M. A., Ospina, S. Y., Mejia, J. F., . . . Laissue, P. (2013). Identification and Functional Characterization of GAA Mutations in Colombian Patients Affected by Pompe Disease. JIMD Rep, 7, 39-48. doi:10.1007/8904_2012_138
dc.source.bibliographicCitationOjeda, D., Lakhal, B., Fonseca, D. J., Braham, R., Landolsi, H., Mateus, H. E., . . . Laissue, P. (2011). Sequence analysis of the CDKN1B gene in patients with premature ovarian failure reveals a novel mutation potentially related to the phenotype. Fertil Steril, 95(8), 2658-2660.e2651. doi:10.1016/j.fertnstert.2011.04.045
dc.source.bibliographicCitationOrtega-Recalde, O., Beltran, O. I., Galvez, J. M., Palma-Montero, A., Restrepo, C. M., Mateus, H. E., & Laissue, P. (2015). Biallelic HERC1 mutations in a syndromic form of overgrowth and intellectual disability. Clin Genet, 88(4), e1-3. doi:10.1111/cge.12634
dc.source.bibliographicCitationOrtega-Recalde, O., Fonseca, D. J., Patino, L. C., Atuesta, J. J., Rivera-Nieto, C., Restrepo, C. M., . . . Laissue, P. (2013). A novel familial case of diffuse leukodystrophy related to NDUFV1 compound heterozygous mutations. Mitochondrion, 13(6), 749-754. doi:10.1016/j.mito.2013.03.010
dc.source.bibliographicCitationOrtega-Recalde, O., Moreno, M. B., Vergara, J. I., Fonseca, D. J., Rojas, R. F., Mosquera, H., . . . Laissue, P. (2015). A novel TGM1 mutation, leading to multiple splicing rearrangements, is associated with autosomal recessive congenital ichthyosis. Clin Exp Dermatol, 40(7), 757-760. doi:10.1111/ced.12627
dc.source.bibliographicCitationOrtega-Recalde, O., Silgado, D., Fetiva, C., Fonseca, D. J., & Laissue, P. (2016). Transcriptomic analysis of skin in a case of ichthyosis Curth-Macklin caused by a KRT1 mutation. Br J Dermatol, 175(6), 1372-1375. doi:10.1111/bjd.149
dc.source.bibliographicCitationOrtega-Recalde, O., Vergara, J. I., Fonseca, D. J., Rios, X., Mosquera, H., Bermudez, O. M., . . . Laissue, P. (2014). Whole-exome sequencing enables rapid determination of xeroderma pigmentosum molecular etiology. PLoS One, 8(6), e64692. doi:10.1371/journal.pone.0064692
dc.source.bibliographicCitationOuwerkerk, P. B., & Meijer, A. H. (2001). Yeast one-hybrid screening for DNA-protein interactions. Curr Protoc Mol Biol, Chapter 12, Unit 12.12. doi:10.1002/0471142727.mb1212s5
dc.source.bibliographicCitationPalei, A. C., Spradley, F. T., Warrington, J. P., George, E. M., & Granger, J. P. (2013). Pathophysiology of Hypertension in Preeclampsia: A Lesson in Integrative Physiology. Acta physiologica (Oxford, England), 208(3), 224-233. doi:10.1111/apha.12106
dc.source.bibliographicCitationPares-Matos, E. I. (2013). Electrophoretic mobility-shift and super-shift assays for studies and characterization of protein-DNA complexes. Methods Mol Biol, 977, 159-167. doi:10.1007/978-1-62703-284-1_12
dc.source.bibliographicCitationPark, H. J., Shim, S. S., & Cha, D. H. (2015). Combined Screening for Early Detection of Pre-Eclampsia. Int J Mol Sci, 16(8), 17952-17974. doi:10.3390/ijms160817952
dc.source.bibliographicCitationPatino, L. C., Battu, R., Ortega-Recalde, O., Nallathambi, J., Anandula, V. R., Renukaradhya, U., & Laissue, P. (2014). Exome sequencing is an efficient tool for variant late-infantile neuronal ceroid lipofuscinosis molecular diagnosis. PLoS One, 9(10), e109576. doi:10.1371/journal.pone.0109576
dc.source.bibliographicCitationPatino, L. C., Beau, I., Carlosama, C., Buitrago, J. C., Gonzalez, R., Suarez, C. F., . . . Laissue, P. (2017). New mutations in non-syndromic primary ovarian insufficiency patients identified via whole-exome sequencing. Hum Reprod, 32(7), 1512-1520. doi:10.1093/humrep/dex089
dc.source.bibliographicCitationPatino, L. C., Silgado, D., & Laissue, P. (2017). A potential functional association between mutant BMPR2 and primary ovarian insufficiency. Syst Biol Reprod Med, 63(3), 145-149. doi:10.1080/19396368.2017.1291767
dc.source.bibliographicCitationPatino, L. C., Walton, K. L., Mueller, T. D., Johnson, K. E., Stocker, W., Richani, D., . . . Harrison, C. A. (2017). BMP15 Mutations Associated With Primary Ovarian Insufficiency Reduce Expression, Activity, or Synergy With GDF9. J Clin Endocrinol Metab, 102(3), 1009-1019. doi:10.1210/jc.2016-3503
dc.source.bibliographicCitationPennington, K. A., Schlitt, J. M., Jackson, D. L., Schulz, L. C., & Schust, D. J. (2012). Preeclampsia: multiple approaches for a multifactorial disease. Dis Model Mech, 5(1), 9-18. doi:10.1242/dmm.008516
dc.source.bibliographicCitationPetla, L. T., Chikkala, R., Ratnakar, K. S., Kodati, V., & Sritharan, V. (2013). Biomarkers for the management of pre-eclampsia in pregnant women. Indian J Med Res, 138, 60-67.
dc.source.bibliographicCitationPowe, C. E., Levine, R. J., & Karumanchi, S. A. (2011). Preeclampsia, a disease of the maternal endothelium: the role of antiangiogenic factors and implications for later cardiovascular disease. Circulation, 123(24), 2856-2869. doi:10.1161/circulationaha.109.853127
dc.source.bibliographicCitationPrada, C. F., & Laissue, P. (2014). A high resolution map of mammalian X chromosome fragile regions assessed by large-scale comparative genomics. Mamm Genome, 25(11-12), 618-635. doi:10.1007/s00335-014-9537-8
dc.source.bibliographicCitationQueimado, L., Lopes, C., Du, F., Martins, C., Bowcock, A. M., Soares, J., & Lovett, M. (1999). Pleomorphic adenoma gene 1 is expressed in cultured benign and malignant salivary gland tumor cells. Lab Invest, 79(5), 583-589.
dc.source.bibliographicCitationQuintero-Ronderos, P., Mercier, E., Fukuda, M., Gonzalez, R., Suarez, C. F., Patarroyo, M. A., . . . Laissue, P. (2017). Novel genes and mutations in patients affected by recurrent pregnancy loss. PLoS One, 12(10), e0186149. doi:10.1371/journal.pone.0186149
dc.source.bibliographicCitationQuintero-Ronderos, P., Mercier, E., Gris, J. C., Esteban-Perez, C., Moreno-Ortiz, H., Fonseca, D. J., . . . Laissue, P. (2017). THBD sequence variants potentially related to recurrent pregnancy loss. Reprod Biol Endocrinol, 15(1), 92. doi:10.1186/s12958-017-0311-0
dc.source.bibliographicCitationRamma, W., & Ahmed, A. (2011). Is inflammation the cause of pre-eclampsia? Biochem Soc Trans, 39(6), 1619-1627. doi:10.1042/bst20110672
dc.source.bibliographicCitationRigourd, V., Chauvet, C., Chelbi, S. T., Rebourcet, R., Mondon, F., Letourneur, F., . . . Vaiman, D. (2008). STOX1 Overexpression in Choriocarcinoma Cells Mimics Transcriptional Alterations Observed in Preeclamptic Placentas. PLoS ONE, 3(12), e3905. doi:10.1371/journal.pone.0003905
dc.source.bibliographicCitationRigourd, V., Chelbi, S., Chauvet, C., Rebourcet, R., Barbaux, S., Bessieres, B., . . . Vaiman, D. (2009). Re-evaluation of the role of STOX1 transcription factor in placental development and preeclampsia. J Reprod Immunol, 82(2), 174-181. doi:10.1016/j.jri.2009.05.001
dc.source.bibliographicCitationRoland, C. S., Hu, J., Ren, C. E., Chen, H., Li, J., Varvoutis, M. S., . . . Jiang, S. W. (2016). Morphological changes of placental syncytium and their implications for the pathogenesis of preeclampsia. Cell Mol Life Sci, 73(2), 365-376. doi:10.1007/s00018-015-2069-x
dc.source.bibliographicCitationRoy, A. L., & Singer, D. S. (2015). Core promoters in transcription: old problem, new insights. Trends Biochem Sci, 40(3), 165-171. doi:10.1016/j.tibs.2015.01.007
dc.source.bibliographicCitationSalonen Ros, H., Lichtenstein, P., Lipworth, L., & Cnattingius, S. (2000). Genetic effects on the liability of developing pre-eclampsia and gestational hypertension. Am J Med Genet, 91(4), 256-260.
dc.source.bibliographicCitationSimoncini, S., Njock, M. S., Robert, S., Camoin-Jau, L., Sampol, J., Harle, J. R., . . . Anfosso, F. (2009). TRAIL/Apo2L mediates the release of procoagulant endothelial microparticles induced by thrombin in vitro: a potential mechanism linking inflammation and coagulation. Circ Res, 104(8), 943-951. doi:10.1161/circresaha.108.183285
dc.source.bibliographicCitationStaff, A. C., Benton, S. J., von Dadelszen, P., Roberts, J. M., Taylor, R. N., Powers, R. W., . . . Redman, C. W. (2013). Redefining preeclampsia using placenta-derived biomarkers. Hypertension, 61(5), 932-942. doi:10.1161/hypertensionaha.111.00250
dc.source.bibliographicCitationSIVIGILA. (2013). Mortalidad Materna. In.
dc.source.bibliographicCitationThulluru, H. K., Park, C., Dufort, D., Kleiverda, G., Oudejans, C., & van Dijk, M. (2013). Maternal Nodal inversely affects NODAL and STOX1 expression in the fetal placenta. Front Genet, 4, 170. doi:10.3389/fgene.2013.00170
dc.source.bibliographicCitationUzan, J., Carbonnel, M., Piconne, O., Asmar, R., & Ayoubi, J. M. (2011). Pre-eclampsia: pathophysiology, diagnosis, and management. Vasc Health Risk Manag, 7, 467-474. doi:10.2147/vhrm.s20181
dc.source.bibliographicCitationVaiman, D., & Miralles, F. (2016). Targeting STOX1 in the therapy of preeclampsia. Expert Opin Ther Targets, 20(12), 1433-1443. doi:10.1080/14728222.2016.1253682
dc.source.bibliographicCitationValenzuela, F. J., Pérez-Sepúlveda, A., Torres, M. J., Correa, P., Repetto, G. M., & Illanes, S. E. (2012). Pathogenesis of Preeclampsia: The Genetic Component. Journal of Pregnancy, 2012, 632732. doi:10.1155/2012/632732
dc.source.bibliographicCitationValero-Rubio, D., Jimenez, K. M., Fonseca, D. J., Payan-Gomez, C., & Laissue, P. (2018). Transcriptomic analysis of FUCA1 knockdown in keratinocytes reveals new insights in the pathogenesis of fucosidosis skin lesions. Exp Dermatol. doi:10.1111/exd.13532
dc.source.bibliographicCitationvan Abel, D., Michel, O., Veerhuis, R., Jacobs, M., van Dijk, M., & Oudejans, C. B. (2012). Direct downregulation of CNTNAP2 by STOX1A is associated with Alzheimer's disease. J Alzheimers Dis, 31(4), 793-800. doi:10.3233/jad-2012-120472
dc.source.bibliographicCitationvan Dijk, M., Drewlo, S., & Oudejans, C. B. (2010). Differential methylation of STOX1 in human placenta. Epigenetics, 5(8), 736-742. doi:10.4161/epi.5.8.13084
dc.source.bibliographicCitationvan Dijk, M., Mulders, J., Poutsma, A., Konst, A. A., Lachmeijer, A. M., Dekker, G. A., . . . Oudejans, C. B. (2005). Maternal segregation of the Dutch preeclampsia locus at 10q22 with a new member of the winged helix gene family. Nat Genet, 37(5), 514-519. doi:10.1038/ng1541
dc.source.bibliographicCitationvan Dijk, M., & Oudejans, C. B. M. (2011). STOX1: Key Player in Trophoblast Dysfunction Underlying Early Onset Preeclampsia with Growth Retardation. Journal of Pregnancy, 2011, 521826. doi:10.1155/2011/521826
dc.source.bibliographicCitationvan Dijk, M., van Bezu, J., van Abel, D., Dunk, C., Blankenstein, M. A., Oudejans, C. B., & Lye, S. J. (2010). The STOX1 genotype associated with pre-eclampsia leads to a reduction of trophoblast invasion by alpha-T-catenin upregulation. Hum Mol Genet, 19(13), 2658-2667. doi:10.1093/hmg/ddq152
dc.source.bibliographicCitationVatin, M., Bouvier, S., Bellazi, L., Montagutelli, X., Laissue, P., Ziyyat, A., . . . Gris, J. C. (2014). Polymorphisms of human placental alkaline phosphatase are associated with in vitro fertilization success and recurrent pregnancy loss. Am J Pathol, 184(2), 362-368. doi:10.1016/j.ajpath.2013.10.024
dc.source.bibliographicCitationWang, A., Rana, S., & Karumanchi, S. A. (2009). Preeclampsia: the role of angiogenic factors in its pathogenesis. Physiology (Bethesda), 24, 147-158. doi:10.1152/physiol.00043.2008
dc.source.bibliographicCitationWiley, S. R., Schooley, K., Smolak, P. J., Din, W. S., Huang, C. P., Nicholl, J. K., . . . et al. (1995). Identification and characterization of a new member of the TNF family that induces apoptosis. Immunity, 3(6), 673-682.
dc.source.bibliographicCitationWilliams, P. J., & Broughton Pipkin, F. (2011). The genetics of pre-eclampsia and other hypertensive disorders of pregnancy. Best Pract Res Clin Obstet Gynaecol, 25(4), 405-417. doi:10.1016/j.bpobgyn.2011.02.007
dc.source.bibliographicCitationWilliams, P. J., & Broughton Pipkin, F. (2011). The genetics of pre-eclampsia and other hypertensive disorders of pregnancy. Best Practice & Research. Clinical Obstetrics & Gynaecology, 25(4-4), 405-417. doi:10.1016/j.bpobgyn.2011.02.007
dc.source.bibliographicCitationXie, Z., Hu, S., Qian, J., Blackshaw, S., & Zhu, H. (2011). Systematic characterization of protein-DNA interactions. Cell Mol Life Sci, 68(10), 1657-1668. doi:10.1007/s00018-010-0617-y
dc.source.instnameinstname:Universidad del Rosariospa
dc.source.reponamereponame:Repositorio Institucional EdocURspa
dc.subjectPreeclampsiaspa
dc.subjectSTOX1spa
dc.subjectPlacentaspa
dc.subjectDisfunción endotelialspa
dc.subject.ddcGinecología & otras especialidades médicas
dc.subject.keywordPreeclampsiaeng
dc.subject.keywordSTOX1eng
dc.subject.keywordPlacentaeng
dc.subject.keywordDysfunctional endothelialeng
dc.subject.lembComplicaciones del embarazospa
dc.subject.lembPreeclampsiaspa
dc.subject.lembObstetricia :: Investigacionesspa
dc.titleIdentificación de factores de transcripción de unión directa a la región promotora de STOX1spa
dc.typemasterThesiseng
dc.type.hasVersioninfo:eu-repo/semantics/acceptedVersion
dc.type.spaTesis de maestríaspa
Archivos
Bloque original
Mostrando1 - 1 de 1
Cargando...
Miniatura
Nombre:
BelloUyaban-SandraPatricia-2018.pdf
Tamaño:
860.24 KB
Formato:
Adobe Portable Document Format
Descripción:
Documento principal