Mostrar el registro sencillo del ítem

dc.contributor.advisorPuerta LLerena, Leonardo (Director/a)
dc.contributor.authorPérez Castillo, Jahaziel
dc.date.accessioned2018-09-24T18:34:14Z
dc.date.available2018-09-24T18:34:14Z
dc.date.issued2018
dc.identifier.citationTM616.97 / P415es
dc.identifier.urihttps://hdl.handle.net/11227/7052
dc.identifier.urihttp://dx.doi.org/10.57799/11227/1348
dc.descriptionTesis (Magister en Inmunología) -- Universidad de Cartagena. Instituto de Investigaciones Inmunológicas. Maestría en Inmunología, 2018es
dc.description.abstractEste proyecto de investigación tiene la finalidad, determinar si la proteína híbrida DPx4 derivada de ácaros domésticos induce la activación de linfocitos T de pacientes alérgicos.es
dc.description.abstractLas enfermedades alérgicas son un problema de salud mundial, con tendencia al aumento en la prevalencia en los últimos años. Estas enfermedades se caracterizan por una respuesta inmunológica principalmente tipo Th2, con secreción de citoquinas proinflamatorias, activación y proliferación de células que participan en los procesos inflamatorios alérgicos. Actualmente el único tratamiento capaz de modular esta respuesta alérgica es la inmunoterapia específica con extractos alergénicos. Se ha propuesto que el uso de vacunas basadas en alérgenos recombinantes o variantes de éstos puede llevar a una inmunoterapia alérgeno específica más segura y eficaz. En este trabajo se investigó la capacidad de la molécula DPx4 de inducir la proliferación de Linfocitos T y su producción de citoquinas mediante citometría de flujo usando cultivo celular de PBMC de pacientes alérgicos a los ácaros del polvo doméstico, un aspecto clave para evaluar su potencial uso para desarrollar preparaciones para inmunoterapia. DPx4 se diseñó como una proteína híbrida expresada en E. coli y se le ha evaluado la reactividad IgE en muestras serológicas de pacientes alérgicos a los ácaros. Aquí se muestran resultados sobre su efecto en linfocitos T. No se observó proliferación de células T específicas inducida por DPx4, y se recomienda hacer este tipo de análisis usando líneas celulares o clonas antígeno específicas. Sin embargo, en los sobrenadantes de los cultivos celulares se observó que esta proteína indujo, en comparación con el efecto del extracto D. pteronyssinus, niveles de IL-5 y TNF-α significativamente menores, niveles de INFγ mayores pero no estadísticamente significativos, al tiempo que los niveles de IL10 fueron significativamente mayores, lo que apoya resultados previos que indican que DPx4 tiene propiedades hipo-alergénicas.
dc.format.mediumapplication/pdf
dc.language.isospaes
dc.publisherUniversidad de Cartagenaes
dc.rights.urihttps://creativecommons.org/licenses/by-nc-nd/4.0
dc.subjectInmunología - Investigacioneses
dc.subjectAlergia - Enfermedadeses
dc.subjectÁcaroses
dc.subjectÁcaros - Alergiaes
dc.subjectSistema inmunees
dc.titleAnálisis de la respuesta de linfocitos T estimulados con una proteína híbrida derivada de alérgenos de ácaros domésticoses
dc.typeTrabajo de grado - Maestríaspa
dcterms.referencesPawankar, R., et al., World Allergy Organization (WAO) white book on allergy. Wisconsin: World Allergy Organisation. Available online at: http://www. worldallergy. org/UserFiles/file/WAO-White-Book-on-Allergy web. pdf, 2011.
dcterms.referencesDennis, R.J., et al., Prevalence of asthma and other allergic conditions in Colombia 2009–2010: a cross-sectional study. BMC pulmonary medicine, 2012. 12(1): p. 17.
dcterms.referencesLim, F.L., et al., Asthma, airway symptoms and rhinitis in office workers in Malaysia: associations with house dust mite (HDM) allergy, cat allergy and levels of house dust mite allergens in office dust. PloS one, 2015. 10(4): p. e0124905.
dcterms.referencesFernández-Caldas E, P.L., Caraballo L, Lockey RF, Allergens and Allergen Immunotherapy: 5th Edition. 2014. 181-201.
dcterms.referencesCaraballo, L., et al., Particularities of allergy in the Tropics. World Allergy Organization Journal, 2016. 9(1): p. 20.
dcterms.referencesBohle, B., et al., Sublingual immunotherapy induces IL-10-producing T regulatory cells, allergen-specific T-cell tolerance, and immune deviation. J Allergy Clin Immunol, 2007. 120(3): p. 707-13.
dcterms.referencesScadding, G.W., et al., Sublingual grass pollen immunotherapy is associated with increases in sublingual Foxp3-expressing cells and elevated allergen-specific immunoglobulin G4, immunoglobulin A and serum inhibitory activity for immunoglobulin E-facilitated allergen binding to B cells. Clin Exp Allergy, 2010. 40(4): p. 598-606.
dcterms.referencesAkdis, C.A. and M. Akdis, Mechanisms of allergen-specific immunotherapy. Journal of Allergy and Clinical Immunology, 2011. 127(1): p. 18-27.
dcterms.referencesReisinger, J., et al., Allergen-specific nasal IgG antibodies induced by vaccination with genetically modified allergens are associated with reduced nasal allergen sensitivity. Journal of allergy and clinical immunology, 2005. 116(2): p. 347-354.
dcterms.referencesChapman, M.D., et al., Recombinant allergens for diagnosis and therapy of allergic disease. Journal of allergy and clinical immunology, 2000. 106(3): p. 409-418.
dcterms.referencesRamos, J.D.A., G.R. Valmonte, and R.M. de Guia, Recombinant proteins and peptides as diagnostic and therapeutic reagents for arthropod allergies. Protein and peptide letters, 2007. 14(10): p. 992-1002.
dcterms.referencesKlimek, L., et al., Allergy immunotherapy with a hypoallergenic recombinant birch pollen allergen rBet v 1-FV in a randomized controlled trial. Clinical and translational allergy, 2015. 5(1): p. 28.
dcterms.referencesBachmann, M. and T. Kündig, Allergen‐specific immunotherapy: is it vaccination against toxins after all? Allergy, 2017. 72(1): p. 13-23.
dcterms.referencesValenta, R., et al., Vaccine development for allergen-specific immunotherapy based on recombinant allergens and synthetic allergen peptides: Lessons from the past and novel mechanisms of action for the future. Journal of Allergy and Clinical Immunology, 2016. 137(2): p. 351-357.
dcterms.referencesCromwell, O., D. Hafner, and A. Nandy, Recombinant allergens for specific immunotherapy. J Allergy Clin Immunol, 2011. 127(4): p. 865-72.
dcterms.referencesBanerjee, S., et al., Conversion of Der p 23, a new major house dust mite allergen, into a hypoallergenic vaccine. J Immunol, 2014. 192(10): p. 4867-75.
dcterms.referencesChen, K.W., et al., Hypoallergenic Der p 1/Der p 2 combination vaccines for immunotherapy of house dust mite allergy. J Allergy Clin Immunol, 2012. 130(2): p. 435-43 e4.
dcterms.referencesReginald, K. and F.T. Chew, Conformational IgE Epitope Mapping of Der p 2 and the Evaluations of Two Candidate Hypoallergens for Immunotherapy. Sci Rep, 2018. 8(1): p. 3391.
dcterms.referencesMartínez, M.E., Hipersensibilidad tipo I frente a los ácaros del polvo doméstico: mecanismos inmunológicos y diagnóstico. Medicina & Laboratorio, 2012. 18: p. 513-536.
dcterms.referencesBousquet, J., et al., Allergic Rhinitis and its Impact on Asthma (ARIA) 2008 update (in collaboration with the World Health Organization, GA(2)LEN and AllerGen). Allergy, 2008. 63 Suppl 86: p. 8-160.
dcterms.referencesPawankar, R., et al., Overview on the pathomechanisms of allergic rhinitis. Asia Pac Allergy, 2011. 1(3): p. 157-67.
dcterms.referencesAkdis, M., et al., Interleukins, from 1 to 37, and interferon-γ: receptors, functions, and roles in diseases. Journal of allergy and clinical immunology, 2011. 127(3): p. 701-721. e70.
dcterms.referencesKalesnikoff, J. and S.J. Galli, New developments in mast cell biology. Nature immunology, 2008. 9(11): p. 1215-1223.
dcterms.referencesKnol, E.F., Requirements for effective IgE cross‐linking on mast cells and basophils. Molecular nutrition & food research, 2006. 50(7): p. 620-624.
dcterms.referencesHesla, H.M., et al., Allergy-related disease in relation to early life exposures—the ALADDIN birth cohort. Journal of Allergy and Clinical Immunology, 2017. 139(2): p. 686-688.
dcterms.referencesAkdis, M., Healthy immune response to allergens: T regulatory cells and more. Current opinion in immunology, 2006. 18(6): p. 738-744.
dcterms.referencesAkkoc, T., M. Akdis, and C.A. Akdis, Update in the mechanisms of allergen-specific immunotheraphy. Allergy, asthma & immunology research, 2011. 3(1): p. 11-20.
dcterms.referencesWang, Y.-H., et al., IL-25 augments type 2 immune responses by enhancing the expansion and functions of TSLP-DC–activated Th2 memory cells. Journal of Experimental Medicine, 2007. 204(8): p. 1837-1847.
dcterms.referencesBilsborough, J., et al., IL-31 is associated with cutaneous lymphocyte antigen–positive skin homing T cells in patients with atopic dermatitis. Journal of Allergy and Clinical Immunology, 2006. 117(2): p. 418-425.
dcterms.referencesLiew, F.Y., N.I. Pitman, and I.B. McInnes, Disease-associated functions of IL-33: the new kid in the IL-1 family. Nature Reviews Immunology, 2010. 10(2): p. 103-110.
dcterms.referencesGri, G., et al., CD4+ CD25+ regulatory T cells suppress mast cell degranulation and allergic responses through OX40-OX40L interaction. Immunity, 2008. 29(5): p. 771-781
dcterms.referencesKearley, J., et al., Resolution of airway inflammation and hyperreactivity after in vivo transfer of CD4+ CD25+ regulatory T cells is interleukin 10 dependent. Journal of Experimental Medicine, 2005. 202(11): p. 1539-1547.
dcterms.referencesMeiler, F., et al., Distinct regulation of IgE, IgG4 and IgA by T regulatory cells and toll‐like receptors. Allergy, 2008. 63(11): p. 1455-1463.
dcterms.referencesTreter, S. and M. Luqman, Antigen-specific T cell tolerance down-regulates mast cell responses in vivo. Cellular immunology, 2000. 206(2): p. 116-124.
dcterms.referencesShim, J.Y., et al., Allergen‐specific conventional immunotherapy decreases immunoglobulin E‐mediated basophil histamine releasability. Clinical & Experimental Allergy, 2003. 33(1): p. 52-57.
dcterms.referencesMarshall, J.S., et al., Interleukin (IL)-10 inhibits long-term IL-6 production but not preformed mediator release from rat peritoneal mast cells. Journal of Clinical Investigation, 1996. 97(4): p. 1122.
dcterms.referencesJutel, M., A. Kosowska, and S. Smolinska, Allergen immunotherapy: past, present, and future. Allergy, asthma & immunology research, 2016. 8(3): p. 191-197.
dcterms.referencesSchandené, L., et al., B7/CD28-dependent IL-5 production by human resting T cells is inhibited by IL-10. The Journal of Immunology, 1994. 152(9): p. 4368-4374.
dcterms.referencesBohle, B., et al., Sublingual immunotherapy induces IL-10–producing T regulatory cells, allergen-specific T-cell tolerance, and immune deviation. Journal of Allergy and Clinical Immunology, 2007. 120(3): p. 707-713.
dcterms.referencesLarché, M., C.A. Akdis, and R. Valenta, Immunological mechanisms of allergen-specific immunotherapy. Nature Reviews Immunology, 2006. 6(10): p. 761-771.
dcterms.referencesJutel, M. and C. Akdis, T-cell regulatory mechanisms in specific immunotherapy, in T Cell Regulation in Allergy, Asthma and Atopic Skin Diseases. 2008, Karger Publishers. p. 158-177.
dcterms.referencesCaraballo, L., et al., Particularities of allergy in the Tropics. World Allergy Organ J, 2016. 9: p. 20
dcterms.referencesAhumada, V., et al., IgE responses to Ascaris and mite tropomyosins are risk factors for asthma. Clin Exp Allergy, 2015. 45(7): p. 1189-200.
dcterms.referencesAcevedo, N. and L. Caraballo, IgE cross-reactivity between Ascaris lumbricoides and mite allergens: possible influences on allergic sensitization and asthma. Parasite Immunol, 2011. 33(6): p. 309-21.
dcterms.referencesSantiago Hda, C., et al., Helminth infection alters IgE responses to allergens structurally related to parasite proteins. J Immunol, 2015. 194(1): p. 93-100.
dcterms.referencesPuerta, L., et al., Mite allergy in the tropics: sensitization to six domestic mite species in Cartagena, Colombia. J Investig Allergol Clin Immunol, 1993. 3(4): p. 198-204.
dcterms.referencesPuerta, L., et al., Sensitization to Chortoglyphus arcuatus and Aleuroglyphus ovatus in Dermatophagoides spp. allergic individuals. Clin Exp Allergy, 1993. 23(2): p. 117-23
dcterms.referencesFerrandiz, R., R. Casas, and S. Dreborg, Sensitization to Dermatophagoides siboney, Blomia tropicalis, and other domestic mites in asthmatic patients. Allergy, 1996. 51(7): p. 501-5.
dcterms.referencesYu, J.M., et al., Diversity of House Dust Mite Species in Xishuangbanna Dai, a Tropical Rainforest Region in Southwest China. Biomed Res Int, 2015. 2015: p. 421716.
dcterms.referencesYu, M.K., et al., Prevalence of Blomia tropicalis in wheezing children in central Taiwan. J Microbiol Immunol Infect, 2008. 41(1): p. 68-73.
dcterms.referencesCaraballo, L., et al., Sensitization to mite allergens and acute asthma in a tropical environment. J Investig Allergol Clin Immunol, 1998. 8(5): p. 281-4.
dcterms.referencesNelson, R.P., Jr., et al., Allergen-specific IgE levels and mite allergen exposure in children with acute asthma first seen in an emergency department and in nonasthmatic control subjects. J Allergy Clin Immunol, 1996. 98(2): p. 258-63.
dcterms.referencesChew, F.T., et al., Allergenic differences between the domestic mites Blomia tropicalis and Dermatophagoides pteronyssinus. Clin Exp Allergy, 1999. 29(7): p. 982-8.
dcterms.referencesPuccio, F.A., et al., Importance of including Blomia tropicalis in the routine diagnosis of Venezuelan patients with persistent allergic symptoms. Allergy, 2004. 59(7): p. 753-7.
dcterms.referencesCantillo J, P.L., From Molecular Cloning to Vaccine Development for Allergic Diseases, An Integrated View of the Molecular Recognition and Toxicology - From Analytical Procedures to Biomedical Applications. 2013.
dcterms.referencesValenta, R., et al., Vaccine development for allergen-specific immunotherapy based on recombinant allergens and synthetic allergen peptides: Lessons from the past and novel mechanisms of action for the future. J Allergy Clin Immunol, 2016. 137(2): p. 351-7
dcterms.referencesPosa, D., et al., Evolution and predictive value of IgE responses toward a comprehensive panel of house dust mite allergens during the first 2 decades of life. J Allergy Clin Immunol, 2017. 139(2): p. 541-549 e8.
dcterms.referencesResch, Y., et al., Different IgE recognition of mite allergen components in asthmatic and nonasthmatic children. J Allergy Clin Immunol, 2015. 136(4): p. 1083-91.
dcterms.references. Shen, H.D., et al., IgE and monoclonal antibody binding by the mite allergen Der p 7. Clin Exp Allergy, 1996. 26(3): p. 308-15.
dcterms.referencesCarvalho Kdos, A., et al., Blomia tropicalis Blo t 5 and Blo t 21 recombinant allergens might confer higher specificity to serodiagnostic assays than whole mite extract. BMC Immunol, 2013. 14: p. 11.
dcterms.referencesHuang, C.H., et al., Characterization of glutathione S-transferase from dust mite, Der p 8 and its immunoglobulin E cross-reactivity with cockroach glutathione S-transferase. Clin Exp Allergy, 2006. 36(3): p. 369-76.
dcterms.referencesAcevedo, N., et al., Proteomic and immunochemical characterization of glutathione transferase as a new allergen of the nematode Ascaris lumbricoides. PLoS One, 2013. 8(11): p. e78353.
dcterms.referencesResch, Y., et al., Molecular characterization of Der p 10: a diagnostic marker for broad sensitization in house dust mite allergy. Clin Exp Allergy, 2011. 41(10): p. 1468-77
dcterms.referencesYi, F.C., et al., Identification of shared and unique immunoglobulin E epitopes of the highly conserved tropomyosins in Blomia tropicalis and Dermatophagoides pteronyssinus. Clin Exp Allergy, 2002. 32(8): p. 1203-10.
dcterms.referencesAsturias, J.A., et al., Engineering of major house dust mite allergens Der p 1 and Der p 2 for allergen-specific immunotherapy. Clin Exp Allergy, 2009. 39(7): p. 1088-98.
dcterms.referencesMartinez, D., et al., Characterization of a hybrid protein designed with segments of allergens from Blomia tropicalis and Dermatophagoides pteronyssinus. Immunol Lett, 2018. 196: p. 103-112.
dcterms.referencesMartinez, D., Puerta, L., Benedetti, I., Munera, M., & Caraballo, L., Evaluation of the immune response induced by a mite derived fusion protein in BALB/c mice. World Allergy Organization Journal, 2015. 8(A): p. 18.
dcterms.referencesPuerta, L., Martinez, D., Munera, M., Cantillo, J., & Caraballo, L. , Recombinant protein assembling epitopes from different allergens of Dermatophagoides pteronyssinus. Allergy, 2012. 67: p. 149.
dcterms.referencesHuang, C.H., et al., Airway inflammation and IgE production induced by dust mite allergenspecific memory/effector Th2 cell line can be effectively attenuated by IL-35. J Immunol, 2011. 187(1): p. 462-71.
dcterms.referencesAbernathy-Carver, K.J., et al., Milk-induced eczema is associated with the expansion of T cells expressing cutaneous lymphocyte antigen. J Clin Invest, 1995. 95(2): p. 913-8.
dcterms.referencesWerfel, T., et al., Milk-responsive atopic dermatitis is associated with a casein-specific lymphocyte response in adolescent and adult patients. J Allergy Clin Immunol, 1997. 99(1 Pt 1): p. 124-33.
dcterms.referencesSilvestri, M., et al., In childhood asthma the degree of allergen-induced T-lymphocyte proliferation is related to serum IgE levels and to blood eosinophilia. Ann Allergy Asthma Immunol, 2000. 84(4): p. 426-32.
dcterms.referencesHales, B.J., H. Shen, and W.R. Thomas, Cytokine responses to Der p 1 and Der p 7: house dust mite allergens with different IgE-binding activities. Clin Exp Allergy, 2000. 30(7): p. 934-43
dcterms.referencesByron, K.A., et al., Dermatophagoides pteronyssinus II-induced interleukin-4 and interferongamma expression by freshly isolated lymphocytes of atopic individuals. Clin Exp Allergy, 1994. 24(9): p. 878-83.
dcterms.referencesClutterbuck, E.J., E.M. Hirst, and C.J. Sanderson, Human interleukin-5 (IL-5) regulates the production of eosinophils in human bone marrow cultures: comparison and interaction with IL-1, IL-3, IL-6, and GMCSF. Blood, 1989. 73(6): p. 1504-12
dcterms.referencesvan Rijt, L.S., et al., Airway eosinophils accumulate in the mediastinal lymph nodes but lack antigen-presenting potential for naive T cells. J Immunol, 2003. 171(7): p. 3372-8.
dcterms.referencesShi, H.Z., et al., Lymph node trafficking and antigen presentation by endobronchial eosinophils. J Clin Invest, 2000. 105(7): p. 945-53.
dcterms.referencesFarhan, R.K., et al., Effective antigen presentation to helper T cells by human eosinophils. Immunology, 2016. 149(4): p. 413-422.
dcterms.referencesWang, H.B., et al., Airway eosinophils: allergic inflammation recruited professional antigenpresenting cells. J Immunol, 2007. 179(11): p. 7585-92.
dcterms.referencesBharadwaj, A.S., A.K. Bewtra, and D.K. Agrawal, Dendritic cells in allergic airway inflammation. Can J Physiol Pharmacol, 2007. 85(7): p. 686-99.
dcterms.referencesRissoan, M.C., et al., Reciprocal control of T helper cell and dendritic cell differentiation. Science, 1999. 283(5405): p. 1183-6.
dcterms.referencesMaldonado-Lopez, R., et al., CD8alpha+ and CD8alpha- subclasses of dendritic cells direct the development of distinct T helper cells in vivo. J Exp Med, 1999. 189(3): p. 587-92.
dcterms.referencesLong, J.A., et al., Higher prostaglandin e2 production by dendritic cells from subjects with sthma compared with normal subjects. Am J Respir Crit Care Med, 2004. 170(5): p. 485-91.
dcterms.referencesPulendran, B., Modulating TH1/TH2 responses with microbes, dendritic cells, and pathogen recognition receptors. Immunol Res, 2004. 29(1-3): p. 187-96.
dcterms.referencesKuipers, H., et al., Dendritic cells retrovirally overexpressing IL-12 induce strong Th1 responses to inhaled antigen in the lung but fail to revert established Th2 sensitization. J Leukoc Biol, 2004. 76(5): p. 1028-38.
dcterms.referencesBatard, T., et al., Production and proteomic characterization of pharmaceutical-grade Dermatophagoides pteronyssinus and Dermatophagoides farinae extracts for allergy vaccines. Int Arch Allergy Immunol, 2006. 140(4): p. 295-305
dcterms.referencesMandhane, S.N., J.H. Shah, and R. Thennati, Allergic rhinitis: an update on disease, present treatments and future prospects. Int Immunopharmacol, 2011. 11(11): p. 1646-62.
dcterms.referencesLiu, Y.H. and J.J. Tsai, Production of salivary immunoglobulin A and suppression of Dermatophagoides pteronyssinus-induced airway inflammation by local nasal immunotherapy. Int Arch Allergy Immunol, 2005. 138(2): p. 161-8.
dcterms.referencesGarcia-Robaina, J.C., et al., Successful management of mite-allergic asthma with modified extracts of Dermatophagoides pteronyssinus and Dermatophagoides farinae in a doubleblind, placebo-controlled study. J Allergy Clin Immunol, 2006. 118(5): p. 1026-32.
dcterms.referencesValenta, R., et al., From allergen genes to allergy vaccines. Annu Rev Immunol, 2010. 28: p. 211-41.
dcterms.referencesVrtala, S., et al., Strategies for converting allergens into hypoallergenic vaccine candidates. Methods, 2004. 32(3): p. 313-20.
dcterms.referencesHoward, M., et al., Identification of a T cell-derived b cell growth factor distinct from interleukin 2. J Exp Med, 1982. 155(3): p. 914-23.
dcterms.referencesAkdis, M., et al., Interleukins (from IL-1 to IL-38), interferons, transforming growth factor beta, and TNF-alpha: Receptors, functions, and roles in diseases. J Allergy Clin Immunol, 2016. 138(4): p. 984-1010.
dcterms.referencesYang, M., et al., Eotaxin-2 and IL-5 cooperate in the lung to regulate IL-13 production and airway eosinophilia and hyperreactivity. J Allergy Clin Immunol, 2003. 112(5): p. 935-43.
dcterms.referencesLau, E.M.T., N.A. Roche, and H.K. Reddel, Therapeutic approaches to asthma-chronic obstructive pulmonary disease overlap. Expert Rev Clin Immunol, 2017. 13(5): p. 449-455.
dcterms.referencesFajt, M.L. and S.E. Wenzel, Development of New Therapies for Severe Asthma. Allergy Asthma Immunol Res, 2017. 9(1): p. 3-14.
dcterms.referencesZimmermann, M., et al., TNF-like weak inducer of apoptosis (TWEAK) and TNF-alpha cooperate in the induction of keratinocyte apoptosis. J Allergy Clin Immunol, 2011. 127(1): p. 200-7, 207 e1-10.
dcterms.referencesBrown, S.D., et al., Characterization of a high TNF-alpha phenotype in children with moderate-to-severe asthma. J Allergy Clin Immunol, 2015. 135(6): p. 1651-4
dcterms.referencesAkkoc, T., et al., Increased activation-induced cell death of high IFN-gamma-producing T(H)1 cells as a mechanism of T(H)2 predominance in atopic diseases. J Allergy Clin Immunol, 2008. 121(3): p. 652-658 e1.
dcterms.referencesRebane, A., et al., Mechanisms of IFN-gamma-induced apoptosis of human skin keratinocytes in patients with atopic dermatitis. J Allergy Clin Immunol, 2012. 129(5): p. 1297-306.
dcterms.referencesAkdis, M. and C.A. Akdis, Therapeutic manipulation of immune tolerance in allergic disease. Nat Rev Drug Discov, 2009. 8(8): p. 645-60.
dcterms.referencesMeiler, F., et al., In vivo switch to IL-10-secreting T regulatory cells in high dose allergen exposure. J Exp Med, 2008. 205(12): p. 2887-98.
dcterms.referencesStanic, B., et al., IL-10-overexpressing B cells regulate innate and adaptive immune responses. J Allergy Clin Immunol, 2015. 135(3): p. 771-80 e8.
dcterms.referencesvan de Veen, W., et al., IgG4 production is confined to human IL-10-producing regulatory B cells that suppress antigen-specific immune responses. J Allergy Clin Immunol, 2013. 131(4): p. 1204-12.
dcterms.referencesAkdis, C.A. and M. Akdis, Mechanisms of immune tolerance to allergens: role of IL-10 and Tregs. J Clin Invest, 2014. 124(11): p. 4678-80.
dcterms.referencesFigueiredo, C.A., et al., Coassociations between IL10 polymorphisms, IL-10 production, helminth infection, and asthma/wheeze in an urban tropical population in Brazil. J Allergy Clin Immunol, 2013. 131(6): p. 1683-90
dcterms.referencesPuerta Llerena, L., et al., Sensitization to Blomia tropicalis and Lepidoglyphus destructor in Dermatophagoides spp-allergic individuals. J Allergy Clin Immunol, 1991. 88(6): p. 943-50.
dcterms.referencesBuendia, E., et al., The IgE response to Ascaris molecular components is associated with clinical indicators of asthma severity. World Allergy Organ J, 2015. 8(1): p. 8.
dcterms.referencesSchwarz, H., et al., Residual endotoxin contaminations in recombinant proteins are sufficient to activate human CD1c+ dendritic cells. PLoS One, 2014. 9(12): p. e113840.
dcterms.referencesPauli, G., et al., Efficacy of recombinant birch pollen vaccine for the treatment of birchallergic rhinoconjunctivitis. J Allergy Clin Immunol, 2008. 122(5): p. 951-60.
dcterms.referencesKlimek, L., et al., Allergy immunotherapy with a hypoallergenic recombinant birch pollen allergen rBet v 1-FV in a randomized controlled trial. Clin Transl Allergy, 2015. 5: p. 28.
dcterms.referencesChen, K.W., et al., Reduction of the in vivo allergenicity of Der p 2, the major house-dust mite allergen, by genetic engineering. Mol Immunol, 2008. 45(9): p. 2486-98.
dcterms.referencesLi, N., et al., Analysis of T-cell epitopes of Der f3 in Dermatophagoides farina. Int J Clin Exp Pathol, 2015. 8(1): p. 137-45.
dcterms.referencesO'Hehir, R.E., et al., Cloned human T lymphocytes reactive with Dermatophagoides farinae (house dust mite): a comparison of T- and B-cell antigen recognition. Immunology, 1987. 62(4): p. 635-40.
dcterms.referencesGuy, C.S., et al., Distinct TCR signaling pathways drive proliferation and cytokine production in T cells. Nat Immunol, 2013. 14(3): p. 262-70.
dc.rights.accessopenAccess


Ficheros en el ítem

Thumbnail

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

Mostrar el registro sencillo del ítem

https://creativecommons.org/licenses/by-nc-nd/4.0
Excepto si se señala otra cosa, la licencia del ítem se describe como https://creativecommons.org/licenses/by-nc-nd/4.0