Mostrar el registro sencillo del ítem

dc.contributor.authorNavarro Quiroz, Elkinspa
dc.contributor.authorChavez-Estrada, Valeriaspa
dc.contributor.authorMacias-Ochoa, Karimespa
dc.contributor.authorAyala-Navarro, María Fernandaspa
dc.contributor.authorFlores-Aguilar, Aniyensy Saraispa
dc.contributor.authorMorales Navarrete, Franciscospa
dc.contributor.authorde la Cruz Lopez, Fernandospa
dc.contributor.authorGOMEZ ESCORCIA, LORENAspa
dc.contributor.authorMusso, Carlosspa
dc.contributor.authorAroca Martínez, Gustavospa
dc.contributor.authorGonzales Torres, Henryspa
dc.contributor.authorDíaz Pérez, Andersonspa
dc.contributor.authorCadena Bonfanti, Andres Angélospa
dc.contributor.authorSarmiento Gutiérrez, Joanyspa
dc.contributor.authorMeza, Jainyspa
dc.contributor.authorDiaz Arroyo, Esperanzaspa
dc.contributor.authorBello-Lemus, Yesitspa
dc.contributor.authorAhmad, Mostaphaspa
dc.contributor.authorNavarro Quiroz, Robertospa
dc.date.accessioned2020-03-06T19:37:56Z
dc.date.available2020-03-06T19:37:56Z
dc.date.issued2019-11-13
dc.identifier.issn1422-0067spa
dc.identifier.urihttps://hdl.handle.net/11323/6084spa
dc.description.abstractThe complex physiology of eukaryotic cells is regulated through numerous mechanisms, including epigenetic changes and posttranslational modifications. The wide-ranging diversity of these mechanisms constitutes a way of dynamic regulation of the functionality of proteins, their activity, and their subcellular localization as well as modulation of the di erential expression of genes in response to external and internal stimuli that allow an organism to respond or adapt to accordingly. However, alterations in these mechanisms have been evidenced in several autoimmune diseases, including systemic lupus erythematosus (SLE). The present review aims to provide an approach to the current knowledge of the implications of these mechanisms in SLE pathophysiology.spa
dc.language.isoeng
dc.publisherInternational Journal of Molecular Sciencesspa
dc.rightsCC0 1.0 Universalspa
dc.rights.urihttp://creativecommons.org/publicdomain/zero/1.0/spa
dc.subjectPosttranslational modificationsspa
dc.subjectEpigenetic mechanismsspa
dc.subjectSystemic lupus erythematosusspa
dc.subjectUbiquitinationspa
dc.subjectSUMOylationspa
dc.subjectGlycosylationspa
dc.subjectHydroxylationspa
dc.subjectPhosphorylationspa
dc.subjectSulfationspa
dc.subjectAcetylationspa
dc.titleEpigenetic mechanisms and posttranslational Modifications in systemic lupus erythematosusspa
dc.typeArtículo de revistaspa
dc.rights.accessrightsinfo:eu-repo/semantics/openAccessspa
dc.identifier.doidoi:10.3390/ijms20225679spa
dc.identifier.instnameCorporación Universidad de la Costaspa
dc.identifier.reponameREDICUC - Repositorio CUCspa
dc.identifier.repourlhttps://repositorio.cuc.edu.co/spa
dc.relation.references1. Wu, H.; Zhao, M.; Tan, L.; Lu, Q. The key culprit in the pathogenesis of systemic lupus erythematosus: Aberrant DNA methylation. Autoimmun. Rev. 2016, 15, 684–689. [CrossRef]spa
dc.relation.references2. Rhodes, B.; Vyse, T.J. The genetics of SLE: An update in the light of genome-wide association studies. Rheumatology (Oxford) 2008, 47, 1603–1611. [CrossRef]spa
dc.relation.references3. Quddus, J.; Johnson, K.J.; Gavalchin, J.; Amento, E.P.; Chrisp, C.E.; Yung, R.L.; Richardson, B.C. Treating activated CD4+ T cells with either of two distinct DNA methyltransferase inhibitors, 5-azacytidine or procainamide, is sufficient to cause a lupus-like disease in syngeneic mice. J. Clin. Invest. 1993, 92, 38–53. [CrossRef] [PubMed]spa
dc.relation.references4. Coit, P.; Yalavarthi, S.; Ognenovski, M.; Zhao, W.; Hasni, S.; Wren, J.D.; Kaplan, M.J.; Sawalha, A.H. EpigenomeprofilingrevealssignificantDNAdemethylationofinterferonsignaturegenesinlupusneutrophils. J. Autoimmun. 2015, 58, 59–66. [CrossRef]spa
dc.relation.references5. Javierre, B.M.; Richardson, B. A New Epigenetic Challenge: Systemic Lupus Erythematosus. In Epigenetic Contributions in Autoimmune Disease. Advances in Experimental Medicine and Biology; Ballestar, E., Ed.; Springer: Boston, MA, USA, 2011; Volume 711, pp. 117–136.spa
dc.relation.references6. Zhao, M.; Zhou, Y.; Zhu, B.; Wan, M.; Jiang, T.; Tan, Q.; Liu, Y.; Jiang, J.; Luo, S.; Tan, Y.; et al. IFI44L promoter methylation as a blood biomarker for systemic lupus erythematosus. Ann. Rheum. Dis. 2016, 75, 1998–2006. [CrossRef] [PubMed]spa
dc.relation.references7. Cai, L.; Sutter, B.M.; Li, B.; Tu, B.P. Acetyl-CoA induces cell growth and proliferation by promoting the acetylation of histones at growth genes. Mol. Cell 2011, 42, 426–437. [CrossRef]spa
dc.relation.references8. Brooks, W.H.; Le Dantec, C.; Pers, J.O.; Youinou, P.; Renaudineau, Y. Epigenetics and autoimmunity. J. Autoimmun. 2010, 34, J207–J219. [CrossRef] [PubMed]spa
dc.relation.references9. Patel, D.R.; Richardson, B.C. Epigenetic mechanisms in lupus. Curr. Opin. Rheumatol. 2010, 22, 478–482. [CrossRef]spa
dc.relation.references10. Zouali, M. Epigenetics in lupus. Ann. N. Y. Acad. Sci. 2011, 1217, 154–165. [CrossRef]spa
dc.relation.references11. Coit, P.; Jeffries, M.; Altorok, N.; Dozmorov, M.G.; Koelsch, K.A.; Wren, J.D.; Merrill, J.T.; McCune, W.J.; Sawalha, A.H. Genome-wide DNA methylation study suggests epigenetic accessibility and transcriptional poising of interferon-regulated genes in naive CD4+ T cells from lupus patients. J. Autoimmun. 2013, 43, 78–84. [CrossRef]spa
dc.relation.references12. Pieterse, E.; Hofstra, J.; Berden, J.; Herrmann, M.; Dieker, J.; van der Vlag, J. Acetylated histones contribute to the immunostimulatory potential of neutrophil extracellular traps in systemic lupus erythematosus. Clin. Exp. Immunol. 2015, 179, 68–74. [CrossRef] [PubMed]spa
dc.relation.references13. Sujashvili, R. Advantages of Extracellular Ubiquitin in Modulation of Immune Responses. Mediators Inflamm. 2016, 2016, 1–6. [CrossRef] [PubMed]spa
dc.relation.references14. Téllez Castillo, N.; Siachoque Jara, J.J.; Siachoque Jara, J.S.; Siachoque Jara, M.A.; Siachoque Montañez, H.O. Activación de la célula T, alteraciones en el lupus eritematoso sistémico, una revisión narrativa. Rev. Colomb. Reumatol. 2018, 25, 38–54. [CrossRef]spa
dc.relation.references15. Barrera-Vargas, A.; Gómez-Martín, D.; Carmona-Rivera, C.; Merayo-Chalico, J.; Torres-Ruiz, J.; Manna, Z.; Hasni, S.; Alcocer-Varela, J.; Kaplan, M.J. Differential ubiquitination in NETs regulates macrophage responses in systemic lupus erythematosus. Ann. Rheum. Dis. 2018, 77, 944–950. [CrossRef] [PubMed]spa
dc.relation.references16. Nakasone, M.A.; Livnat-Levanon, N.; Glickman, M.H.; Cohen, R.E.; Fushman, D. Mixed-linkage ubiquitin chains send mixed messages. Structure 2013, 21, 727–740. [CrossRef]spa
dc.relation.references17. Erpapazoglou, Z.; Walker, O.; Haguenauer-Tsapis, R. Versatile roles of k63-linked ubiquitin chains in trafficking. Cells 2014, 3, 1027–1088. [CrossRef]spa
dc.relation.references18. Saavedra Hernández, D. La molécula CD28 y su función en la activación de células T. Rev. Cuba. Hematol. Inmunol. Hemoter. 2013, 29, 359–367.spa
dc.relation.references19. Ding, X.; Wang, A.; Ma, X.; Demarque, M.; Jin, W.; Xin, H.; Dejean, A.; Dong, C. Protein SUMOylation Is Required for Regulatory T Cell Expansion and Function. Cell Rep. 2016, 16, 1055–1066. [CrossRef]spa
dc.relation.references20. Rider, V.; Abdou, N.I.; Kimler, B.F.; Lu, N.; Brown, S.; Fridley, B.L. Gender bias in human systemic lupus erythematosus: A problem of steroid receptor action? Front. Immunol. 2018, 9, 1–10. [CrossRef]spa
dc.relation.references21. Barry, R.; John, S.W.; Liccardi, G.; Tenev, T.; Jaco, I.; Chen, C.H.; Choi, J.; Kasperkiewicz, P.; Fernandes-Alnemri, T.; Alnemri, E.; et al. SUMO-mediated regulation of NLRP3 modulates inflammasome activity. Nat. Commun. 2018, 9, 3001. [CrossRef]spa
dc.relation.references22. Guzmán-Flores, J.M.; Portales-Pérez, D.P. Mecanismos de supresión de las células T reguladoras (Treg). Gac. Med. Mex. 2013, 149, 630–638.spa
dc.relation.references23. Hernández, A.S. Células colaboradoras (TH1, TH2, TH17) y reguladoras (Treg, TH3, NKT) en la artritis reumatoide. Reumatol. Clin. Supl. 2009, 5 (Suppl. 1), 1–5. [CrossRef]spa
dc.relation.references24. Crabtree, G.R.; Schreiber, S.L. Snapshot: Calcium-calcineurin-NFAT signaling. Cell 2010, 138, 1–4.spa
dc.relation.references25. Biermann, M.H.; Griffante, G.; Podolska, M.J.; Boeltz, S.; Stürmer, J.; Muñoz, L.E.; Bilyy, R.; Herrmann, M. Sweet but dangerous–The role of immunoglobulin G glycosylation in autoimmunity and inflammation. Lupus 2016, 25, 934–942. [CrossRef] [PubMed]spa
dc.relation.references26. Magnelli, P.E.; Bielik, A.M.; Guthrie, E.P. Identification and characterization of protein glycosylation using specific endo- and exoglycosidases. J. Vis. Exp. 2011, e3749. [CrossRef]spa
dc.relation.references27. Valliere-Douglass, J.F.; Kodama, P.; Mujacic, M.; Brady, L.J.; Wang, W.; Wallace, A.; Yan, B.; Reddy, P.; Treuheit, M.J.; Balland, A. Asparagine-linked oligosaccharides present on a non-consensus amino acid sequence in the CH1 domain of human antibodies. J. Biol. Chem. 2009, 284, 32493–32506. [CrossRef]spa
dc.relation.references28. Hashii, N.; Kawasaki, N.; Itoh, S.; Nakajima, Y.; Kawanishi, T.; Yamaguchi, T. Alteration of N-glycosylation in the kidney in a mouse model of systemic lupus erythematosus: Relative quantification of N-glycans using an isotope-tagging method. Immunology 2009, 126, 336–345. [CrossRef]spa
dc.relation.references29. Vidarsson, G.; Dekkers, G.; Rispens, T. IgG subclasses and allotypes: From structure to effector functions. Front. Immunol. 2014, 5, 520. [CrossRef]spa
dc.relation.references30. Abès, R.; Teillaud, J.-L. Impact of Glycosylation on Effector Functions of Therapeutic IgG. Pharmaceuticals 2010, 3, 146–157. [CrossRef]spa
dc.relation.references31. Jennewein, M.F.; Alter, G. The Immunoregulatory Roles of Antibody Glycosylation. Trends Immunol. 2017, 38, 358–372. [CrossRef]spa
dc.relation.references32. Reily, C.; Stewart, T.J.; Renfrow, M.B.; Novak, J. Glycosylation in health and disease. Nat. Rev. Nephrol. 2019, 15, 346–366. [CrossRef] [PubMed]spa
dc.relation.references33. Anthony, R.M.; Ravetch, J.V. A Novel Role for the IgG Fc Glycan: The Anti-inflammatory Activity of Sialylated IgG Fcs. J. Clin. Immunol. 2010, 30, 9–14. [CrossRef] [PubMed]spa
dc.relation.references34. Saxena, A.; Wu, D. Advances in Therapeutic Fc Engineering - Modulation of IgG-Associated Effector Functions and Serum Half-life. Front. Immunol. 2016, 7, 580. [CrossRef] [PubMed]spa
dc.relation.references35. Leong, K.W.; Ding, J.L. The unexplored roles of human serum IgA. DNA Cell Biol. 2014, 33, 823–829. [CrossRef]spa
dc.relation.references36. Papista, C.; Berthelot, L.; Monteiro, R.C. Dysfunctions of the Iga system: A common link between intestinal and renal diseases. Cell. Mol. Immunol. 2011, 8, 126–134. [CrossRef]spa
dc.relation.references37. Kawa,I.A.; Masood,A.; Amin,S.; Mustafa,M.F.; Rashid,F.Chapter2—ClinicalPerspectiveofPosttranslational Modifications. In Protein Modificomics; Dar, T.A., Singh, L.R., Eds.; Academic Press: London, UK, 2019; pp. 37–68.spa
dc.relation.references38. Zurlo, G.; Guo, J.; Takada, M.; Wei, W.; Zhang, Q. New Insights into Protein Hydroxylation and Its Important Role in Human Diseases. Biochim. Biophys. Acta 2016, 1866, 208–220. [CrossRef]spa
dc.relation.references39. Mansoor, F.; Ali, A.; Ali, R. Binding of circulating SLE autoantibodies to oxygen free radical damage chromatin. Autoimmunity 2005, 38, 431–438. [CrossRef]spa
dc.relation.references40. Lahita, R.G.; Bradlow, L.; Fishman, J.; Kunkel, H.G. Estrogen metabolism in systemic lupus erythematosus. Patients and family members. Arthritis Rheum. 1982, 25, 843–846. [CrossRef]spa
dc.relation.references41. Garg, D.K.; Ali, R. Reactive oxygen species modified polyguanylic acid: Immunogenicity and implications for systemic autoimmunity. J. Autoimmun. 1998, 11, 371–378. [CrossRef]spa
dc.relation.references42. Ardito, F.; Giuliani, M.; Perrone, D.; Troiano, G.; Lo Muzio, L. The crucial role of protein phosphorylation in cell signalingand its use as targeted therapy (Review). Int. J. Mol. Med. 2017, 40, 271–280. [CrossRef]spa
dc.relation.references43. Skourti-Stathaki, K.; Proudfoot, N. Histone 3 S10 Phosphorylation: ‘Caught in the R Loop!’. Mol. Cell 2013, 52, 470–472. [CrossRef]spa
dc.relation.references44. Eichten, S.R.; Schmit, R.J.; Springer, N.M. Epigenetics: Beyond chromatin modifications and complex genetic regulation. Plant Physiol. 2014, 165, 933–947. [CrossRef]spa
dc.relation.references45. Rossetto, D.; Avvakumov, N.; Côté, J. Histone phosphorylation. Epigenetics 2012, 7, 1098–1108. [CrossRef]spa
dc.relation.references46. Rossy, J.; Williamson, D.J.; Gaus, K. How does the kinase Lck phosphorylate the T cell receptor? Spatial organization as a regulatory mechanism. Front. Immunol. 2012, 3, 1–6. [CrossRef]spa
dc.relation.references47. Wu, T.; Xie, C.; Han, J.; Ye, Y.; Weiel, J.; Li, Q.; Blanco, I.; Ahn, C.; Olsen, N.; Putterman, C.; et al. Metabolic disturbances associated with systemic lupus erythematosus. PLoS ONE 2012, 7, e37210. [CrossRef]spa
dc.relation.references48. Hsu, W.; Rosenquist, G.L.; Ansari, A.A.; Gershwin, M.E. Autoimmunity and tyrosine sulfation. Autoimmun. Rev. 2005, 4, 429–435. [CrossRef]spa
dc.relation.references49. Tonks, N.K. Protein tyrosine phosphatases: From genes, to function, to disease. Nat. Rev. Mol. Cell Biol. 2006, 7, 833–834. [CrossRef]spa
dc.relation.references50. Kehoe, J.W.; Bertozzi, C.R. Tyrosine sulfation: A modulator of extracellular protein-protein interactions. Chem. Biol. 2000, 7, 57–61. [CrossRef]spa
dc.relation.references51. Seibert, C.; Sakmar, T.P. Toward a framework for sulfoproteomics: Synthesis and characterization of sulfotyrosine-containing peptides. Biopolym. 2008, 90, 459–477. [CrossRef]spa
dc.relation.references52. Farzan, M.; Mirzabekov, T.; Kolchinsky, P.; Wyatt, R.; Cayabyab, M.; Gerard, N.P.; Gerard, C.; Sodroski, J.; Choe, H. Tyrosine sulfation of the amino terminus of CCR5 facilitates HIV-1 entry. Cell 1999, 96, 667–676. [CrossRef]spa
dc.relation.references53. Carvalho, C.; Calvisi, S.L.; Leal, B.; Bettencourt, A.; Marinho, A.; Almeida, I.; Farinha, F.; Costa, P.P.; Silva, B.M.; Vasconcelos, C. CCR5-Delta32: Implications in SLE development. Int. J. Immunogenet. 2014, 41, 236–241. [CrossRef] [PubMed]spa
dc.relation.references54. Ren, J.; Panther, E.; Liao, X.; Grammer, A.C.; Lipsky, P.E.; Reilly, C.M. The Impact of Protein Acetylation/Deacetylation on Systemic Lupus Erythematosus. Int. J. Mol. Sci. 2018, 19, 4007. [CrossRef] [PubMed]spa
dc.relation.references55. Shahbazian, M.D.; Grunstein, M. Functions of site-specific histone acetylation and deacetylation. Annu. Rev. Biochem. 2007, 76, 75–100. [CrossRef] [PubMed]spa
dc.relation.references56. Cheung, W.L.; Briggs, S.D.; Allis, C.D. Acetylation and chromosomal functions. Curr. Opin. Cell Biol. 2000, 12, 326–333. [CrossRef]spa
dc.relation.references57. Wang, Z.; Chang, C.; Peng, M.; Lu, Q. Translating epigenetics into clinic: Focus on lupus. Clin. Epigenetics 2017, 9, 1–15. [CrossRef]spa
dc.relation.references58. Bannister, A.J.; Kouzarides, T. Regulation of chromatin by histone modifications. Cell Res. 2011, 21, 381–395. [CrossRef]spa
dc.relation.references59. Drazic, A.; Myklebust, L.M.; Ree, R.; Arnesen, T. The world of protein acetylation. Biochim. Biophys. Acta 2016, 1864, 1372–1401. [CrossRef]spa
dc.relation.references60. Parthun, M.R. Hat1: The emerging cellular roles of a type B histone acetyltransferase. Oncogene 2007, 26, 5319–5328. [CrossRef]spa
dc.relation.references61. Leung, Y.T.; Shi, L.; Maurer, K.; Song, L.; Zhang, Z.; Petri, M.; Sullivan, K.E. Interferon regulatory factor 1 and histone H4 acetylation in systemic lupus erythematosus. Epigenetics 2015, 10, 191–199. [CrossRef]spa
dc.relation.references62. Tsai, K.L.; Liao, C.C.; Chang, Y.S.; Huang, C.W.; Huang, Y.C.; Chen, J.H.; Lin, S.H.; Tai, C.C.; Lin, Y.F.; Lin, C.Y. Low Levels of IgM and IgA Recognizing Acetylated C1-Inhibitor Peptides Are Associated with Systemic Lupus Erythematosus in Taiwanese Women. Molecules 2019, 24, 1645. [CrossRef]spa
dc.relation.references63. Nettis, E.; Colanardi, M.C.; Loria, M.P.; Vacca, A. Acquired C1-inhibitor deficiency in a patient with systemic lupus erythematosus: A case report and review of the literature. Eur. J. Clin. Invest. 2005, 35, 781–784. [CrossRef] [PubMed]spa
dc.relation.references64. Dunn, J.; Simmons, R.; Thabet, S.; Jo, H. The role of epigenetics in the endothelial cell shear stress response and atherosclerosis. Int. J. Biochem. Cell Biol. 2015, 67, 167–176. [CrossRef] [PubMed]spa
dc.relation.references65. Rodríguez-Dorantes, M.; Téllez-Ascencio, N.; Cerbón, M.A.; Lez, M.; Cervantes, A. Metilación del ADN: Un fenómeno epigenético de importancia Médica. Rev. Invest. Clin. 2004, 56, 56–71. [PubMed]spa
dc.relation.references66. Pedroza Díaz, N.J.; Ortiz Reyes, B.L.; Vásquez Duque, G.M. Protein Biomarkers in Neuropsychiatric Lupus. Rev. Colomb. Reumatol. 2012, 19, 158–171.spa
dc.relation.references67. Godsell, J.; Rudloff, I.; Kandane-Rathnayake, R.; Hoi, A.; Nold, M.F.; Morand, M.F.; Harris, J. Clinical associations of IL-10 and IL-37 in systemic lupus erythematosus. Sci. Rep. 2016, 6, 1–10. [CrossRef]spa
dc.relation.references68. Lu, Q.; Wu, A.; Richardson, B.C. Demethylation of the same promoter sequence increases CD70 expression in lupus T cells and T cells treated with lupus-inducing drugs. J. Immunol. 2005, 174, 6212–6219. [CrossRef]spa
dc.relation.references69. Pretel, M.; Marquès, l.; España, A. Lupus eritematoso inducido por fármacos. Actas Dermosifiliogr. 2012, 105, 18–30. [CrossRef]spa
dc.relation.references70. Richardson, B. Epigenetically Altered T Cells Contribute to Lupus Flares. Cells 2019, 8, 127. [CrossRef]spa
dc.relation.references71. Teruel, M.; Sawalha, A.H. Epigenetic Variability in Systemic Lupus Erythematosus: What We Learned from Genome-Wide DNA Methylation Studies. Curr. Rheumatol. Rep. 2017, 19, 32. [CrossRef]spa
dc.relation.references72. Díaz, J.P.; Muñoz Vahos, C.H.; Luján Chavarría, T.P.; Vásquez Duque, G.M.; Ortiz Reyes, B.L. Análisis proteómico del líquido cefalorraquídeo de pacientes con lupus neuropsiquiátrico, un abordaje inicial para la búsqueda de biomarcadores. Rev. Colomb. Reumatol. 2014, 21, 115–124. [CrossRef]spa
dc.relation.references73. Cheung, P.; Lau, P. Epigenetic Regulation by Histone Methylation and Histone Variants. Mol. Endocrinol. 2005, 19, 563–573. [CrossRef] [PubMed]spa
dc.relation.references74. Mondal, S.; Gong, X.; Zhang, X.; Salinger, A.J.; Zheng, L.; Sen, S.; Weerapana, E.; Zhang, X.; Thompson, P.R. Halogen Bonding Increases the Potency and Isozyme-selectivity of Protein Arginine Deiminase 1 Inhibitors. Angew. Chemie 2019, 58, 12476–12480. [CrossRef] [PubMed]spa
dc.relation.references75. Knuckley, B.; Causey, C.P.; Jones, J.E.; Bhatia, M.; Dreyton, C.J.; Osborne, T.C.; Takahara, H.; Thompson, P.R. Substrate specificity and kinetic studies of PADs 1, 3, and 4 identify potent and selective inhibitors of protein arginine deiminase 3. Biochemistry 2010, 49, 4852–4863. [CrossRef] [PubMed]spa
dc.relation.references76. Nakashima, K.; Hagiwara, T.; Yamada, M. Nuclear localization of peptidylarginine deiminase V and histone deimination in granulocytes. J. Biol. Chem. 2002, 277, 49562–49568. [CrossRef]spa
dc.relation.references77. Kakumanu, P.; Sobel, E.S.; Narain, S.; Li, Y.; Akaogi, J.; Yamasaki, Y.; Segal, M.S.; Hahn, P.C.; Chan, E.K.; Reeves, W.H.; et al. Citrulline dependence of anti-cyclic citrullinated peptide antibodies in systemic lupus erythematosus as a marker of deforming/erosive arthritis. J. Rheumatol. 2009, 36, 2682–2690. [CrossRef]spa
dc.relation.references78. Muller, S.; Radic, M. Citrullinated Autoantigens: From Diagnostic Markers to Pathogenetic Mechanisms. Clin. Rev. Allergy Immunol. 2015, 49, 232–239. [CrossRef]spa
dc.relation.references79. Navarro Quiroz, E.; Navarro Quiroz, R.; Pacheco Lugo, L.; Aroca Martínez, G.; Gómez Escorcia, L.; Gonzalez Torres, H.; Cadena Bonfanti, A.; Marmolejo, M.D.C.; Sanchez, E.; Villarreal Camacho, J.L.; et al. Integrated analysis of microRNA regulation and its interaction with mechanisms of epigenetic regulation in the etiology of systemic lupus erythematosus. PLoS ONE 2019, 14, e0218116. [CrossRef]spa
dc.relation.references80. Kronimus, Y.; Dodel, R.; Galuska, S.P.; Neumann, S. IgG Fc N-glycosylation: Alterations in neurologic diseases and potential therapeutic target? J. Autoimmun. 2019, 96, 14–23. [CrossRef]spa
dc.relation.references81. Gruszewska, E.; Chludzinska, A.; Chrostek, L.; Cylwik, B.; Gindzienska-Sieskiewicz, E.; Szmitkowski, M.; Sierakowski, S. Carbohydrate-deficient transferrin depends on disease activity in rheumatoid arthritis and systemic sclerosis. Scand. J. Rheumatol. 2013, 42, 203–206. [CrossRef]spa
dc.relation.references82. Pozo, M.C. Inestabilidad Genética y Cambios en la Cromatina en Mutantes del Complejo THO en Mitosis y Meiosis de Eucariotas Modelo. Ph.D. Thesis, Universidad de Sevilla, Seville, Spain, December 2013.spa
dc.type.coarhttp://purl.org/coar/resource_type/c_6501spa
dc.type.contentTextspa
dc.type.driverinfo:eu-repo/semantics/articlespa
dc.type.redcolhttp://purl.org/redcol/resource_type/ARTspa
dc.type.versioninfo:eu-repo/semantics/acceptedVersionspa
dc.type.coarversionhttp://purl.org/coar/version/c_ab4af688f83e57aaspa
dc.rights.coarhttp://purl.org/coar/access_right/c_abf2spa


Ficheros en el ítem

Thumbnail
Thumbnail

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

  • Artículos científicos [3154]
    Artículos de investigación publicados por miembros de la comunidad universitaria.

Mostrar el registro sencillo del ítem

CC0 1.0 Universal
Excepto si se señala otra cosa, la licencia del ítem se describe como CC0 1.0 Universal