Publicación:
Efectos inmunomoduladores de la cistatina de Ascaris lumbricoides sobre los linfocitos B

dc.contributor.advisorZakzuk Sierra, Josefina
dc.contributor.authorRodríguez Cantillo, Jonathan José
dc.date.accessioned2022-04-08T17:54:13Z
dc.date.available2022-04-08T17:54:13Z
dc.date.issued2022
dc.description.abstractLa actividad aberrante de los linfocitos B se ha asociado con distintas enfermedades autoinmunitarias como la artritis reumatoide (AR) (1). Esta enfermedad se caracteriza por tener un fuerte componente inflamatorio por lo que han sido distintos los mecanismos estudiados para crear dianas terapéuticas. Las cistatinas helmínticas han demostrado tener efectos antiinflamatorios en modelos de inflamación como la AR experimental. rAlCPI, una cistatina helmíntica, ha demostrado un papel inmunorregulador en modelos inflamatorios. Uno de los mecanismos por medio del cual lo hace es a través de la inducción de la citoquina antiinflamatoria IL-10. Hasta ahora se desconoce el papel de rAl-CPI en la inducción de IL-10 en los linfocitos B de donadores sanos y pacientes con AR. En este estudio se analizó la inducción de IL-10 por parte de linfocitos B purificados de PBMCs de donadores, y pacientes con AR pareados con controles por edad y sexo frente a la estimulación con rAl-CPI. Durante la selección de la concentración de rAl-CPI adecuada para realizar las estimulaciones de todos los experimentos, se encontró que la concentración 0,1 µM indujo los mayores niveles de IL-10 en comparación con las otras concentraciones (1 µM, 0,01 µM, 0,001 µM). Para averiguar si los bajos niveles de IL-10 en la concentración 1 µM se debieron a efectos citotóxicos, se realizaron estudios de viabilidad de PBMCs y linfocitos B con los marcadores FVD y Anexina-V + yoduro de propidio frente a las cuatro concentraciones de rAl-CPI utilizadas. Se encontró que rAlCPI 1 µM fue citotóxica para los linfocitos B de donadores sanos, a diferencia de la concentración de rAl-CPI 0.1 µM. Posteriormente, cuando se evaluaron los niveles de IL10 en sobrenadantes de PBMCs provenientes de donadores sanos mediante ELISA se encontraron unos niveles más altos en la condición estimulada con rAl-CPI 0.1 µM comparada con la condición control (p=0,012). Para verificar la inducción en linfocitos B de marcadores asociados a Bregs, las PBMCs de los donadores sanos fueron sometidas a tinción de marcadores de superficie e intracelular (CD25, CD1d, CD71, CD73, CD5 e IL.10). Se encontró un descenso estadísticamente significativo en la expresión del marcador CD25 en la condición rAl-CPI 0.1 µM + CpG comparada con CpG (p=0.037), mientras que no se hallaron diferencias significativas en los demás marcadores. Después, se midieron los niveles de IL-10 en los sobrenadantes de linfocitos B purificados de PBMCs de donadores sanos mediante ELISA y se encontraron unos niveles más altos en la condición estimulada con rAl-CPI comparada con la condición control (p=0,0037). Posteriormente, se compararon los niveles de IL-10 en los sobrenadantes de linfocitos B purificados de pacientes con AR y sus controles pareados mediante ELISA, encontrándose diferencias significativas en los niveles de IL-10 cuando se comparó la condición RPMI entre los grupos (p=0,034). La medición de IL-10 e IL-6 mediante CBA no arrojó diferencias significativas cuando se comparó entre las condiciones y grupos. En conclusión, se demuestra que las altas concentraciones de rAl-CPI son citotóxicas para la subpoblación de linfocitos B. La estimulación con rAl-CPI sugiere que es capaz de inducir producción de IL-10 en linfocitos B de donadores sanos. Esta tendencia no fue tan clara en el grupo de casos y controles. La inducción de linfocitos B reguladores no es concluyente de acuerdo a las mediciones de marcadores de superficie e intracelulares. Se sugiere la realización de estos experimentos con otras técnicas de purificación de linfocitos B, así como un aumento en el tamaño de la muestra.spa
dc.description.degreelevelMaestríaspa
dc.description.degreenameMagíster en Inmunologíaspa
dc.format.extent87 hojasspa
dc.format.mimetypeapplication/pdfspa
dc.identifier.urihttps://hdl.handle.net/11227/14988
dc.identifier.urihttp://dx.doi.org/10.57799/11227/1350
dc.language.isospaspa
dc.publisherUniversidad de Cartagenaspa
dc.publisher.facultyFacultad de Medicinaspa
dc.publisher.placeCartagena de Indiasspa
dc.publisher.programMaestría en Inmunologíaspa
dc.rightsDerechos Reservados - Universidad de Cartagena, 2022spa
dc.rights.accessrightsinfo:eu-repo/semantics/openAccessspa
dc.rights.creativecommonsAtribución-NoComercial 4.0 Internacional (CC BY-NC 4.0)spa
dc.rights.urihttps://creativecommons.org/licenses/by-nc/4.0/spa
dc.subject.armarcInmunología
dc.subject.armarcSistema inmunológico - Enfermedades
dc.subject.armarcArtritis reumatoide
dc.subject.armarcEnfermedades autoinmunes
dc.subject.armarcEnfermedades inmunológicas
dc.titleEfectos inmunomoduladores de la cistatina de Ascaris lumbricoides sobre los linfocitos Bspa
dc.typeTrabajo de grado - Maestríaspa
dc.type.coarhttp://purl.org/coar/resource_type/c_bdccspa
dc.type.contentTextspa
dc.type.driverinfo:eu-repo/semantics/masterThesisspa
dc.type.redcolhttps://purl.org/redcol/resource_type/TMspa
dc.type.versioninfo:eu-repo/semantics/publishedVersionspa
dcterms.referencesAlivernini S, Tolusso B, Fedele AL, Di Mario C, Ferraccioli G, Gremese E. The B side of rheumatoid arthritis pathogenesis. Pharmacol Res. 2019 Nov;149(July):104465.
dcterms.referencesCaraballo L, Zakzuk J, Acevedo N. Helminth-derived cystatins: The immunomodulatory properties of an Ascaris lumbricoides cystatin. Parasitology. 2021;148(14):1744–56.
dcterms.referencesGiannini D, Antonucci M, Petrelli F, Bilia S, Alunno A, Puxeddu I. One year in review 2020: pathogenesis of rheumatoid arthritis. Clin Exp Rheumatol. 2020;38(3):387–97.
dcterms.referencesRodríguez Cantillo J, López Crespo J, Polo Ibarra J, Moreno Grau Á, Alvis Guzmán N, Zakzuk Sierra J. Medición de la calidad de vida de pacientes con artritis reumatoide atendidos en el Hospital Universitario del Caribe, Bolívar, Colombia. Rev Ciencias Biomédicas. 2021;10(4):246–55.
dcterms.referencesMartinec R. Quality of Life in Patients with Rheumatoid Arthritis – a Preliminary Study. Acta Clin Croat. 2019;58(1):157–66.
dcterms.referencesHuscher D, Mittendorf T, von Hinüber U, Kötter I, Hoese G, Pfäfflin A, et al. Evolution of cost structures in rheumatoid arthritis over the past decade. Ann Rheum Dis. 2015 Apr;74(4):738–45.
dcterms.referencesGierut A, Perlman H, Pope RM. Innate Immunity and Rheumatoid Arthritis. Rheum Dis Clin North Am. 2010 May;36(2):271–96.
dcterms.referencesKlareskog L, Amara K, Malmström V. Adaptive immunity in rheumatoid arthritis. Curr Opin Rheumatol. 2014 Jan;26(1):72–9.
dcterms.referencesAletaha D, Funovits J, Smolen JS. Physical disability in rheumatoid arthritis is associated with cartilage damage rather than bone destruction. Ann Rheum Dis. 2011 May 1;70(5):733–9.
dcterms.referencesKay J, Upchurch KS. ACR/EULAR 2010 rheumatoid arthritis classification criteria. Rheumatology. 2012 Dec 1;51(suppl 6):vi5–9.
dcterms.referencesNakayamada S, Kubo S, Yoshikawa M, Miyazaki Y, Yunoue N, Iwata S, et al. Differential effects of biological DMARDs on peripheral immune cell phenotypes in patients with rheumatoid arthritis. Rheumatology. 2018 Jan 1;57(1):164–74.
dcterms.referencesAl-Zifzaf DS, El Bakry SA, Mamdouh R, Shawarby LA, Ghaffar AYA, Amer HA, et al. FoxP3+T regulatory cells in Rheumatoid arthritis and the imbalance of the Treg/TH17 cytokine axis. Egypt Rheumatol. 2015 Jan;37(1):7–15.
dcterms.referencesAlunno A, Manetti M, Caterbi S, Ibba-Manneschi L, Bistoni O, Bartoloni E, et al. Altered Immunoregulation in Rheumatoid Arthritis: The Role of Regulatory T Cells and Proinflammatory Th17 Cells and Therapeutic Implications. Mediators Inflamm. 2015;2015:1–12.
dcterms.referencesUmmarino D. Defective IL-10-producing Breg cells. Nat Rev Rheumatol. 2017 Mar 2;13(3):132–132
dcterms.referencesMatsushita T. Regulatory and effector B cells: Friends or foes? J Dermatol Sci. 2019 Jan;93(1):2–7.
dcterms.referencesWolf SD, Dittel BN, Hardardottir F, Janeway CA. Experimental Autoimmune Encephalomyelitis Induction in Genetically B Cell–deficient Mice. J Exp Med. 1996 Dec 1;184(6):2271–8.
dcterms.referencesMizoguchi A, Mizoguchi E, Takedatsu H, Blumberg RS, Bhan AK. Chronic Intestinal Inflammatory Condition Generates IL-10-Producing Regulatory B Cell Subset Characterized by CD1d Upregulation. Immunity. 2002 Feb;16(2):219–30.
dcterms.referencesFillatreau S, Sweenie CH, McGeachy MJ, Gray D, Anderton SM. B cells regulate autoimmunity by provision of IL-10. Nat Immunol. 2002 Oct 3;3(10):944–50.
dcterms.referencesMauri C, Gray D, Mushtaq N, Londei M. Prevention of Arthritis by Interleukin 10–producing B Cells. J Exp Med. 2003 Feb 17;197(4):489–501.
dcterms.referencesMizoguchi A, Bhan AK. A Case for Regulatory B Cells. J Immunol. 2006 Jan 15;176(2):705–10.
dcterms.referencesGray M, Miles K, Salter D, Gray D, Savill J. Apoptotic cells protect mice from autoimmune inflammation by the induction of regulatory B cells. Proc Natl Acad Sci. 2007 Aug 28;104(35):14080–5.
dcterms.referencesCarter NA, Vasconcellos R, Rosser EC, Tulone C, Muñoz-Suano A, Kamanaka M, et al. Mice Lacking Endogenous IL10–Producing Regulatory B Cells Develop Exacerbated Disease and Present with an Increased Frequency of Th1/Th17 but a Decrease in Regulatory T Cells. J Immunol. 2011 May 15;186(10):5569–79.
dcterms.referencesSun J-B, Flach C-F, Czerkinsky C, Holmgren J. B Lymphocytes Promote Expansion of Regulatory T Cells in Oral Tolerance: Powerful Induction by Antigen Coupled to Cholera Toxin B Subunit. J Immunol. 2008 Dec 15;181(12):8278– 87.
dcterms.referencesTadmor T, Zhang Y, Cho H-M, Podack ER, Rosenblatt JD. The absence of B lymphocytes reduces the number and function of T-regulatory cells and enhances the anti-tumor response in a murine tumor model. Cancer Immunol Immunother. 2011 May 21;60(5):609–19.
dcterms.referencesMatsumoto M, Baba A, Yokota T, Nishikawa H, Ohkawa Y, Kayama H, et al. Interleukin-10-Producing Plasmablasts Exert Regulatory Function in Autoimmune Inflammation. Immunity. 2014 Dec;41(6):1040–51.
dcterms.referencesRay A, Dittel B. Mechanisms of Regulatory B cell Function in Autoimmune and Inflammatory Diseases beyond IL-10. J Clin Med. 2017 Jan 23;6(1):12.
dcterms.referencesBraza F, Chesne J, Castagnet S, Magnan A, Brouard S. Regulatory functions of B cells in allergic diseases. Allergy. 2014 Nov;69(11):1454–63.
dcterms.referencesZhang Y, Gallastegui N, Rosenblatt JD. Regulatory B cells in anti-tumor immunity. Int Immunol. 2015 Oct;27(10):521–30.
dcterms.referencesWortel C, Heidt S. Regulatory B cells: Phenotype, function and role in transplantation. Transpl Immunol. 2017 Mar;41:1– 9.
dcterms.referencesShen P, Fillatreau S. Suppressive functions of B cells in infectious diseases. Int Immunol. 2015 Oct;27(10):513–9.
dcterms.referencesMauri C, Bosma A. Immune Regulatory Function of B Cells. Annu Rev Immunol. 2012 Apr 23;30(1):221–41.
dcterms.referencesShen P, Fillatreau S. Antibody-independent functions of B cells: a focus on cytokines. Nat Rev Immunol. 2015 Jul 12;15(7):441–51.
dcterms.referencesBaba Y, Matsumoto M, Kurosaki T. Signals controlling the development and activity of regulatory B-lineage cells. Int Immunol. 2015 Oct;27(10):487–93.
dcterms.referencesFillatreau S. Natural regulatory plasma cells. Curr Opin Immunol. 2018 Dec;55:62–6.
dcterms.referencesRudensky AY. Regulatory T cells and Foxp3. Immunol Rev. 2011 May;241(1):260–8.
dcterms.referencesLampropoulou V, Hoehlig K, Roch T, Neves P, Gómez EC, Sweenie CH, et al. TLR-Activated B Cells Suppress T CellMediated Autoimmunity. J Immunol. 2008 Apr 1;180(7):4763–73.
dcterms.referencesYanaba K, Bouaziz J-D, Matsushita T, Tsubata T, Tedder TF. The Development and Function of Regulatory B Cells Expressing IL-10 (B10 Cells) Requires Antigen Receptor Diversity and TLR Signals. J Immunol. 2009 Jun 15;182(12):7459–72.
dcterms.referencesGiordani L, Sanchez M, Libri I, Quaranta MG, Mattioli B, Viora M. IFN-α amplifies human naïve B cell TLR-9-mediated activation and Ig production. J Leukoc Biol. 2009 Aug;86(2):261–71.
dcterms.referencesYang M, Sun L, Wang S, Ko K-H, Xu H, Zheng B-J, et al. Cutting Edge: Novel Function of B Cell-Activating Factor in the Induction of IL-10–Producing Regulatory B Cells. J Immunol. 2010 Apr 1;184(7):3321–5.
dcterms.referencesYoshizaki A, Miyagaki T, DiLillo DJ, Matsushita T, Horikawa M, Kountikov EI, et al. Regulatory B cells control T-cell autoimmunity through IL-21-dependent cognate interactions. Nature. 2012 Nov 14;491(7423):264–8.
dcterms.referencesRosser EC, Oleinika K, Tonon S, Doyle R, Bosma A, Carter NA, et al. Regulatory B cells are induced by gut microbiota– driven interleukin-1β and interleukin-6 production. Nat Med. 2014 Nov 19;20(11):1334–9.
dcterms.referencesWang R-X, Yu C-R, Dambuza IM, Mahdi RM, Dolinska MB, Sergeev Y V., et al. Interleukin-35 induces regulatory B cells that suppress autoimmune disease. Nat Med. 2014 Jun 17;20(6):633–41.
dcterms.referencesBaba Y, Saito Y, Kotetsu Y. Heterogeneous subsets of B-lineage regulatory cells (Breg cells). Int Immunol. 2020 Mar 7;32(3):155–62.
dcterms.referencesMatsushita T, Yanaba K, Bouaziz J-D, Fujimoto M, Tedder TF. Regulatory B cells inhibit EAE initiation in mice while other B cells promote disease progression. J Clin Invest. 2008 Sep 18;118(10):3420–30.
dcterms.referencesMeng X, Grötsch B, Luo Y, Knaup KX, Wiesener MS, Chen X-X, et al. Hypoxia-inducible factor-1α is a critical transcription factor for IL-10-producing B cells in autoimmune disease. Nat Commun. 2018 Dec 17;9(1):251.
dcterms.referencesYang X, Yang J, Chu Y, Xue Y, Xuan D, Zheng S, et al. T Follicular Helper Cells and Regulatory B Cells Dynamics in Systemic Lupus Erythematosus. Rieux-Laucat F, editor. PLoS One. 2014 Feb 14;9(2):e88441.
dcterms.referencesHussain S, Delovitch TL. Intravenous Transfusion of BCR-Activated B Cells Protects NOD Mice from Type 1 Diabetes in an IL-10-Dependent Manner. J Immunol. 2007 Dec 1;179(11):7225–32.
dcterms.referencesEvans JG, Chavez-Rueda KA, Eddaoudi A, Meyer-Bahlburg A, Rawlings DJ, Ehrenstein MR, et al. Novel Suppressive Function of Transitional 2 B Cells in Experimental Arthritis. J Immunol. 2007 Jun 15;178(12):7868–78.
dcterms.referencesXiao S, Brooks CR, Sobel RA, Kuchroo VK. Tim-1 Is Essential for Induction and Maintenance of IL-10 in Regulatory B Cells and Their Regulation of Tissue Inflammation. J Immunol. 2015 Feb 15;194(4):1602–8.
dcterms.referencesRafei M, Hsieh J, Zehntner S, Li M, Forner K, Birman E, et al. A granulocyte-macrophage colony–stimulating factor and interleukin-15 fusokine induces a regulatory B cell population with immune suppressive properties. Nat Med. 2009 Sep 9;15(9):1038–45.
dcterms.referencesMatsumoto M, Baba A, Yokota T, Nishikawa H, Ohkawa Y, Kayama H, et al. Interleukin-10-producing plasmablasts exert regulatory function in autoimmune inflammation. Immunity. 2014;41(6):1040–51.
dcterms.referencesIwata Y, Matsushita T, Horikawa M, DiLillo DJ, Yanaba K, Venturi GM, et al. Characterization of a rare IL-10-competent B-cell subset in humans that parallels mouse regulatory B10 cells. Blood. 2011;117(2):530–41.
dcterms.referencesFlores-Borja F, Bosma A, Ng D, Reddy V, Ehrenstein MR, Isenberg DA, et al. CD19+CD24hiCD38hi B Cells Maintain Regulatory T Cells While Limiting TH1 and TH17 Differentiation. Sci Transl Med. 2013 Feb 20;5(173):173ra23-173ra23.
dcterms.referencesDíaz-Alderete A, Crispin JC, Inés Vargas-Rojas M, Alcocer-Varela J. IL-10 production in B cells is confined to CD154+ cells in patients with systemic lupus erythematosus. J Autoimmun. 2004 Dec;23(4):379–83.
dcterms.referencesTian J, Zekzer D, Hanssen L, Lu Y, Olcott A, Kaufman DL. Lipopolysaccharide-Activated B Cells Down-Regulate Th1 Immunity and Prevent Autoimmune Diabetes in Nonobese Diabetic Mice. J Immunol. 2001 Jul 15;167(2):1081–9
dcterms.referencesKessel A, Haj T, Peri R, Snir A, Melamed D, Sabo E, et al. Human CD19+CD25high B regulatory cells suppress proliferation of CD4+ T cells and enhance Foxp3 and CTLA-4 expression in T-regulatory cells. Autoimmun Rev. 2012 Jul;11(9):670–7.
dcterms.referencesShen P, Roch T, Lampropoulou V, O’Connor RA, Stervbo U, Hilgenberg E, et al. IL-35-producing B cells are critical regulators of immunity during autoimmune and infectious diseases. Nature. 2014 Mar 23;507(7492):366–70.
dcterms.referencesAllen JE, Maizels RM. Diversity and dialogue in immunity to helminths. Nat Rev Immunol. 2011 Jun 25;11(6):375–88
dcterms.referencesvan Riet E, Hartgers FC, Yazdanbakhsh M. Chronic helminth infections induce immunomodulation: Consequences and
dcterms.referencesFigueiredo CA, Barreto ML, Rodrigues LC, Cooper PJ, Silva NB, Amorim LD, et al. Chronic Intestinal Helminth Infections Are Associated with Immune Hyporesponsiveness and Induction of a Regulatory Network. Infect Immun. 2010 Jul;78(7):3160–7.
dcterms.referencesMalhotra I, Ouma JH, Wamachi A, Kioko J, Mungai P, Njzovu M, et al. Influence of Maternal Filariasis on Childhood Infection and Immunity to Wuchereria bancrofti in Kenya. Infect Immun. 2003 Sep;71(9):5231–7.
dcterms.referencesMahanty S, Mollis SN, Ravichandran M, Abrams JS, Kumaraswami V, Jayaraman K, et al. High Levels of Spontaneous and Parasite Antigen-Driven Interieukin-10 Production Are Associated with Antigen-Specific Hyporesponsiveness in Human Lymphatic Filariasis. J Infect Dis. 1996 Mar 1;173(3):769–72.
dcterms.referencesBabu S, Bhat SQ, Pavan Kumar N, Lipira AB, Kumar S, Karthik C, et al. Filarial Lymphedema Is Characterized by Antigen-Specific Th1 and Th17 Proinflammatory Responses and a Lack of Regulatory T Cells. Fujiwara RT, editor. PLoS Negl Trop Dis. 2009 Apr 21;3(4):e420.
dcterms.referencesMcSorley HJ, Maizels RM. Helminth Infections and Host Immune Regulation. Clin Microbiol Rev. 2012 Oct 1;25(4):585– 608.
dcterms.referencesCaraballo L, Acevedo N, Zakzuk J. Ascariasis as a model to study the helminth/allergy relationships. Parasite Immunol. 2019 Jun 6;41(6):e12595.
dcterms.referencesSmallwood TB, Giacomin PR, Loukas A, Mulvenna JP, Clark RJ, Miles JJ. Helminth immunomodulation in autoimmune disease. Front Immunol. 2017;8(APR).
dcterms.referencesHewitson JP, Grainger JR, Maizels RM. Helminth immunoregulation: The role of parasite secreted proteins in modulating host immunity. Mol Biochem Parasitol. 2009 Sep;167(1):1–11.
dcterms.referencesCoakley G, Maizels RM, Buck AH. Exosomes and Other Extracellular Vesicles: The New Communicators in Parasite Infections. Trends Parasitol. 2015 Oct;31(10):477–89.
dcterms.referencesCoakley G, Buck AH, Maizels RM. Host parasite communications—Messages from helminths for the immune system. Mol Biochem Parasitol. 2016 Jul;208(1):33–40.
dcterms.referencesSmits HH, Hammad H, van Nimwegen M, Soullie T, Willart MA, Lievers E, et al. Protective effect of Schistosoma mansoni infection on allergic airway inflammation depends on the intensity and chronicity of infection. J Allergy Clin Immunol. 2007 Oct;120(4):932–40.
dcterms.referencesWilson MS, Taylor MD, O’Gorman MT, Balic A, Barr TA, Filbey K, et al. Helminth-induced CD19+CD23hi B cells modulate experimental allergic and autoimmune inflammation. Eur J Immunol. 2010 Mar 19;40(6):1682–96.
dcterms.referencesAmu S, Saunders SP, Kronenberg M, Mangan NE, Atzberger A, Fallon PG. Regulatory B cells prevent and reverse allergic airway inflammation via FoxP3-positive T regulatory cells in a murine model. J Allergy Clin Immunol. 2010;125(5):1114-1124.e8.
dcterms.referencesMangan NE, Fallon RE, Smith P, van Rooijen N, McKenzie AN, Fallon PG. Helminth Infection Protects Mice from Anaphylaxis via IL-10-Producing B Cells. J Immunol. 2004 Nov 15;173(10):6346–56.
dcterms.referencesCorreale J, Farez M, Razzitte G. Helminth infections associated with multiple sclerosis induce regulatory B cells. Ann Neurol. 2008;64(2):187–99.
dcterms.referencesMangan NE, Fallon RE, Smith P, van Rooijen N, McKenzie AN, Fallon PG. Helminth Infection Protects Mice from Anaphylaxis via IL-10-Producing B Cells. J Immunol. 2004 Nov 15;173(10):6346–56.
dcterms.referencesCorreale J, Farez M, Razzitte G. Helminth infections associated with multiple sclerosis induce regulatory B cells. Ann Neurol. 2008;64(2):187–99.
dcterms.referencesCorreale J, Farez M. Association between parasite infection and immune responses in multiple sclerosis. Ann Neurol. 2007 Feb;61(2):97–108.
dcterms.referencesVray B, Hartmann S, Hoebeke J. Immunomodulatory properties of cystatins. Cell Mol Life Sci. 2002 Oct 1;59(9):1503–12.
dcterms.referencesBird PI, Trapani JA, Villadangos JA. Endolysosomal proteases and their inhibitors in immunity. Nat Rev Immunol. 2009 Dec;9(12):871–82.
dcterms.referencesPrunk M, Perišić Nanut M, Sabotič J, Kos J. Cystatins, cysteine peptidase inhibitors, as regulators of immune cell cytotoxicity. Period Biol. 2017 Mar 17;118(4):353–62.
dcterms.referencesHenskens YM, Veerman EC, Nieuw Amerongen A V. Review. Biol Chem Hoppe Seyler. 1996 Jan;377(2):71–120.
dcterms.referencesTurk B, Turk D, Turk V. Protease signalling: The cutting edge. EMBO J. 2012;31(7):1630–43.
dcterms.referencesCurtis MA, Aduse-Opoku J, Rangarajan M. Cysteine Proteases of Porphyromonas Gingivalis. Crit Rev Oral Biol Med. 2001 May 1;12(3):192–216.
dcterms.referencesKocholaty W, Weil L, Smith L. Proteinase secretion and growth of Clostridium histolyticum. Biochem J. 1938;32(10):1685–90.
dcterms.referencesKay J, Dunn BM. Viral proteinases: weakness in strength. BBA - Gene Struct Expr. 1990;1048(1):1–18.
dcterms.referencesBarrett AJ, Rawlings ND, Woesser JF. Cysteine peptidases. In: Handbook of Proteolytic Enzimes. London: Academic Pess; 1998. p. 543–798.
dcterms.referencesMcKerrow JH, Sun E, Rosenthal PJ, Bouvier J. The Proteases and Pathogenicity of Parasitic Protozoa. Annu Rev Microbiol. 1993 Oct;47(1):821–53.
dcterms.referencesBarrett AJ, Fritz H, Grubb A, Isemura S, Järvinen M, Katunuma N, et al. Nomenclature and classification of the proteins homologous with the cysteine-proteinase inhibitor chicken cystatin. Biochem J. 1986 May 15;236(1):312–312.
dcterms.referencesTurk B, Krizaj I, Kralj B, Dolenc I, Popovic T, Bieth JG, et al. Bovine stefin C, a new member of the stefin family. J Biol Chem. 1993 Apr 5;268(10):7323–9.
dcterms.referencesTurk V, Bode W. The cystatins: Protein inhibitors of cysteine proteinases. FEBS Lett. 1991 Jul 1;285(2):213–9.
dcterms.referencesRinne A, Dorn A, Järvinen M, Alavaikko M, Jokinen K, Hopsu-Havu VK. Immunoelectron microscopical location of the acid cysteine proteinase inhibitor in the lymphatic tissue of the tonsils. Acta Histochem. 1986 Jan;79(2):137–45.
dcterms.referencesFreije JP, Pendás AM, Velasco G, Roca A, Abrahamson M, López-Otín C. Localization of the human cystatin D gene (CST5) to chromosome 20p11.21 by in situ hybridization. Cytogenet Genome Res. 1993;62(1):29–31.
dcterms.referencesNi J, Abrahamson M, Zhang M, Fernandez MA, Grubb A, Su J, et al. Cystatin E is a Novel Human Cysteine Proteinase Inhibitor with Structural Resemblance to Family 2 Cystatins. J Biol Chem. 1997 Apr;272(16):10853–8.
dcterms.referencesSotiropoulou G, Anisowicz A, Sager R. Identification, Cloning, and Characterization of Cystatin M, a Novel Cysteine Proteinase Inhibitor, Down-regulated in Breast Cancer. J Biol Chem. 1997 Jan;272(2):903–10.
dcterms.referencesManoury B, Gregory WF, Maizels RM, Watts C. Bm-CPI-2, a cystatin homolog secreted by the filarial parasite Brugia malayi, inhibits class II MHC-restricted antigen processing. Curr Biol. 2001 Mar;11(6):447–51.
dcterms.referencesHartmann S, Lucius R. Modulation of host immune responses by nematode cystatins. Int J Parasitol. 2003 Sep;33(11):1291–302.
dcterms.referencesFong D, Chan MM, Hsieh WT. Gene mapping of human cathepsins and cystatins. Biomed Biochim Acta. 1991;50(4–6):595–8.
dcterms.referencesNakanishi S. Substance P precursor and kininogen: their structures, gene organizations, and regulation. Physiol Rev. 1987 Oct 1;67(4):1117–42.
dcterms.referencesSalvesen G, Parkes C, Abrahamson M, Grubb A, Barrett AJ. Human low-Mr kininogen contains three copies of a cystatin sequence that are divergent in structure and in inhibitory activity for cysteine proteinases. Biochem J. 1986 Mar 1;234(2):429–34.
dcterms.referencesDeLa Cadena RA, Colman RW. Structure and functions of human kininogens. Trends Pharmacol Sci. 1991Jan;12(C):272–5.
dcterms.referencesHall A, Dalboge H, Grubb A, Abrahamson M. Importance of the evolutionarily conserved glycine residue in the Nterminal region of human cystatin C (Gly-11) for cysteine endopeptidase inhibition. Biochem J. 1993;291(1):123–9.
dcterms.referencesAUERSWALD EA, GENENGER G, ASSFALG-MACHLEIDT I, MACHLEIDT W, ENGH RA, FRITZ H. Recombinant chicken egg white cystatin variants of the QLVSG region. Eur J Biochem. 1992 Nov;209(3):837–45.
dcterms.referencesJosiah Ochieng, Gautam Chaudhuri. Cystatin Superfamily. J Health Care Poor Underserved. 2010;21(1A):51–70
dcterms.referencesLiu J, Svärd SG, Klotz C. Giardia intestinalis cystatin is a potent inhibitor of papain, parasite cysteine proteases and, to a lesser extent, human cathepsin B. FEBS Lett. 2019 Jun 22;593(12):1313–25.
dcterms.referencesCoronado S, Zakzuk J, Regino R, Ahumada V, Benedetti I, Angelina A, et al. Ascaris lumbricoides Cystatin Prevents Development of Allergic Airway Inflammation in a Mouse Model. Front Immunol. 2019 Sep 27;10(SEP).
dcterms.referencesSchönemeyer A, Lucius R, Sonnenburg B, Brattig N, Sabat R, Schilling K, et al. Modulation of Human T Cell Responses and Macrophage Functions by Onchocystatin, a Secreted Protein of the Filarial Nematode Onchocerca volvulus. J Immunol. 2001 Sep 15;167(6):3207–15.
dcterms.referencesSun Y, Liu G, Li Z, Chen Y, Liu Y, Liu B, et al. Modulation of dendritic cell function and immune response by cysteine protease inhibitor from murine nematode parasite Heligmosomoides polygyrus. Immunology. 2013 Apr;138(4):370–81.
dcterms.referencesVenugopal G, Mueller M, Hartmann S, Steinfelder S. Differential immunomodulation in human monocytes versus macrophages by filarial cystatin. Sharma J, editor. PLoS One. 2017 Nov 15;12(11):e0188138.
dcterms.referencesCoronado S, Barrios L, Zakzuk J, Regino R, Ahumada V, Franco L, et al. A recombinant cystatin from Ascaris lumbricoides attenuates inflammation of DSS-induced colitis. Parasite Immunol. 2017 Apr;39(4):e12425.
dcterms.referencesDainichi T, Maekawa Y, Ishii K, Zhang T, Nashed BF, Sakai T, et al. Nippocystatin, a Cysteine Protease Inhibitor fromNippostrongylus brasiliensis, Inhibits Antigen Processing and Modulates Antigen-Specific Immune Response. Petri WA, editor. Infect Immun. 2001 Dec 1;69(12):7380–6.
dcterms.referencesKobpornchai P, Flynn RJ, Reamtong O, Rittisoonthorn N, Kosoltanapiwat N, Boonnak K, et al. A novel cystatin derived from Trichinella spiralis suppresses macrophage-mediated inflammatory responses. Hübner MP, editor. PLoS Negl Trop Dis. 2020 Apr 1;14(4):e0008192.
dcterms.referencesBisht N, Khatri V, Chauhan N, Kalyanasundaram R. Cystatin from Filarial Parasites Suppress the Clinical Symptoms and Pathology of Experimentally Induced Colitis in Mice by Inducing T-Regulatory Cells, B1-Cells, and Alternatively Activated Macrophages. Biomedicines. 2019 Oct 31;7(4):85.
dcterms.referencesKiyohara H, Sujino T, Teratani T, Miyamoto K, Arai MM, Nomura E, et al. Toll-Like Receptor 7 Agonist–Induced Dermatitis Causes Severe Dextran Sulfate Sodium Colitis by Altering the Gut Microbiome and Immune Cells. Cmgh. 2019;7(1):135–56.
dcterms.referencesPreisker S, Brethack A-K, Bokemeyer A, Bettenworth D, Sina C, Derer S. Crohn’s Disease Patients in Remission Display an Enhanced Intestinal IgM+ B Cell Count in Concert with a Strong Activation of the Intestinal Complement System. Cells. 2019 Jan 21;8(1):78.
dcterms.referencesYadav RSP, Khatri V, Amdare N, Goswami K, Shivkumar VB, Gangane N, et al. Immuno-Modulatory Effect and Therapeutic Potential of Brugia malayi Cystatin in Experimentally Induced Arthritis. Indian J Clin Biochem. 2016 Apr 23;31(2):203–8.
dcterms.referencesLiu F, Cheng W, Pappoe F, Hu X, Wen H, Luo Q, et al. Schistosoma japonicum cystatin attenuates murine collageninduced arthritis. Parasitol Res. 2016 Oct 8;115(10):3795–806.
dcterms.referencesLopez Crespo J. Efecto de las parasitosis intestinales en la presentación y severidad de artritis reumatoide y su impacto en linfocitos T y B reguladores. Universidad de Cartagena; 2019.
dcterms.referencesLozano Mendoza A. Evaluación de los efectos inmunomoduladores de la cistatina de Ascaris lumbricoides sobre poblaciones celulares humanas. Universidad de Cartagena; 2019.
dcterms.referencesKoopman G, Reutelingsperger CPM, Kuijten GAM, Keehnen RMJ, Pals ST, Van Oers MHJ. Annexin V for flow cytometric detection of phosphatidylserine expression on B cells undergoing apoptosis. Blood. 1994;84(5):1415–20.
dcterms.referencesMielle J, Audo R, Hahne M, Macia L, Combe B, Morel J, et al. IL-10 producing B cells ability to induce regulatory T cells is maintained in rheumatoid arthritis. Front Immunol. 2018;9(MAY):1–11.
dcterms.referencesHanten JA, Vasilakos JP, Riter CL, Neys L, Lipson KE, Alkan SS, et al. Comparison of human B cell activation by TLR7 and TLR9 agonists. BMC Immunol. 2008;9:1–15.
dcterms.referencesBrisslert M, Bokarewa M, Larsson P, Wing K, Vincent L, Tarkowski A. Phenotypic and functional characterization of human CD 25 + B cells
dcterms.referencesWilson EH, Katz E, Goodridge HS, Harnett MM, Harnett W. In vivo activation of murine peritoneal B1 cells by the filarial nematode phosphorylcholine-containing glycoprotein ES-62. Parasite Immunol. 2003;25(8–9):463–6.
dcterms.referencesHarnett W, McInnes IB, Harnett MM. ES-62, a filarial nematode-derived immunomodulator with anti-inflammatory potential. Immunol Lett. 2004;94(1–2):27–33.
dcterms.referencesZakzuk J, Mercado D, Bornacelly A, Sánchez J, Ahumada V, Acevedo N, et al. Hygienic conditions influence sensitization to Blomia tropicalis allergenic components: Results from the FRAAT birth cohort. Pediatr Allergy Immunol. 2019;30(2):172–8.
dcterms.referencesZakzuk J. Imunorregulação induzida por helmintos: Uma atualização. Iatreia. 2016;29(2):182–93.
dcterms.referencesBuendía E, Zakzuk J, Mercado D, Alvarez A, Caraballo L. The IgE response to Ascaris molecular components is associated with clinical indicators of asthma severity. World Allergy Organ J. 2015;8(1):8.
dcterms.referencesCaraballo L, Acevedo N. New Allergens of Relevance in Tropical Regions: The Impact of Ascaris lumbricoides Infections. World Allergy Organ J. 2011;4(5):77–84.
dcterms.referencesHaahtela T, Holgate S, Pawankar R, Akdis CA, Benjaponpitak S, Caraballo L, et al. The biodiversity hypothesis and allergic disease: world allergy organization position statement. World Allergy Organ J. 2013;6(1):3.
dcterms.referencesBach JF. The effect of infections on susceptibility to autoimmune and allergic diseases. N Engl J Med. 2002 Sep;347(12):911–20.
dcterms.referencesZakzuk J, Casadiego S, Mercado A, Alvis-Guzman N, Caraballo L. Ascaris lumbricoides infection induces both, reduction and increase of asthma symptoms in a rural community. Acta Trop. 2018 Nov;187(August 2017):1–4.
dcterms.referencesCaraballo L, Zakzuk J, Lee BW, Acevedo N, Soh JY, Sánchez-Borges M, et al. Particularities of allergy in the Tropics. World Allergy Organ J. 2016;9(1):1–44.
dcterms.referencesCaraballo L, Valenta R, Puerta L, Pomés A, Zakzuk J, Fernandez-Caldas E, et al. The allergenic activity and clinical impact of individual IgE-antibody binding molecules from indoor allergen sources. World Allergy Organ J. 2020 May;13(5):100118.
dcterms.referencesMartínez B, Simão F, Gomes V, Nguidi M, Amorim A, Carvalho EF, et al. Searching for the roots of the first free African American community. Sci Rep. 2020 Dec 26;10(1):20634.
dcterms.referencesAcevedo N, Benfeitas R, Katayama S, Bruhn S, Andersson A, Wikberg G, et al. Epigenetic alterations in skin homing CD4+CLA+ T cells of atopic dermatitis patients. Sci Rep. 2020 Dec 22;10(1):18020.
dcterms.referencesAhumada V, Zakzuk J, Coronado S, Aglaz L, Araujo G, Briza P, et al. Identification of a new, no cross-reacting allergen from Ascaris lumbricoides. World Allergy Organ J. 2020 Aug;13(8):100400.
dcterms.referencesAhumada V, Manotas M, Zakzuk J, Aglas L, Coronado S, Briza P, et al. Identification and Physicochemical Characterization of a New Allergen from Ascaris lumbricoides. Int J Mol Sci. 2020 Dec 21;21(24):9761.
dcterms.referencesLozano A, Zakzuk J, Caraballo L. Characterization of the allergenic properties of a pi-class glutathione transferase from Ascaris suum. World Allergy Organ J. 2020 Aug;13(8):100399.
dspace.entity.typePublication
oaire.accessrightshttp://purl.org/coar/access_right/c_abf2spa
oaire.versionhttp://purl.org/coar/version/c_970fb48d4fbd8a85spa

Archivos

Bloque original

Mostrando 1 - 2 de 2
Cargando...
Miniatura
Nombre:
Rodriguez-Cantillo,JJ. Tesis 2022. Maestría en Inmunología (Repositorio)).pdf
Tamaño:
3.86 MB
Formato:
Adobe Portable Document Format
Descripción:
Cargando...
Miniatura
Nombre:
AUTORIZACIÓN DE PUBLICACIÓN DE TRABAJO DE GRADO JONATHAN RODRÍGUEZ.pdf
Tamaño:
403.05 KB
Formato:
Adobe Portable Document Format
Descripción:

Bloque de licencias

Mostrando 1 - 1 de 1
Cargando...
Miniatura
Nombre:
license.txt
Tamaño:
1.71 KB
Formato:
Item-specific license agreed upon to submission
Descripción:

Colecciones

Datos de Contacto

Imagen Escudo Universidad de Cartagena

 

 

 

Línea de Atención

Línea Anticorrupción

Síguenos en: