• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 161
  • 39
  • 16
  • 11
  • 5
  • 4
  • 3
  • 2
  • 2
  • 1
  • 1
  • 1
  • 1
  • 1
  • 1
  • Tagged with
  • 306
  • 188
  • 182
  • 115
  • 75
  • 51
  • 49
  • 46
  • 43
  • 40
  • 39
  • 39
  • 37
  • 37
  • 36
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
301

Imunogenetické a hormonální predispoziční markery systémových revmatických onemocnění,zejména systémového lupus erythematodu / Immunogenetic and hormonal markers of predisposition to systemic rheumatic diseases particularly systemic lupus erythematosus

Fojtíková, Markéta January 2011 (has links)
Fojtikova 2011 INTRODUCTION: Several factors like genetic susceptibility is required for systemic rheumatic diseases development. Immunomodulatory PRL effect supports autoimmunity. AIMS: 1. To detect the immunogenetic background (alleles HLA class I, II and microsatellite polymorphism of the transmembrane part exon 5 of MIC-A gene) of SLE and PsA. 2. To detect PRL serum and synovial fluid with regard to clinical and laboratory RA activity. 3. To find the role of the functional polymorphism -1149G/T SNP PRL of extrapituitary promoter of PRL gene in SLE, RA, PsA, SSc and inflammatory myopathies development. METHODS: Genetic analyses of pateints with SLE (n=156), RA (n=173), PsA (n=100), SSc (n=75), PM (n=47) a DM (n=68) and 123 healthy individuals: PCR-SSP (HLA clase I and II), PCR-fragment analysis (MIC-A) a PCR-RFLP (-1149 G/T SNP PRL). In 29 RA a 26 OA PRL serum and synovial fluid concentrations were detected using immunoradiometric assay. RESULTS: 1. The allele HLA-DRB1*03 (pc=0.008; OR 2.5) and haplotype HLA-DRB1*03-DQB1*0201 (pc <0.001; OR 4.54) were determined as risk immunogenetic markers for SLE in Czech population. In SLE versus controls allele MIC-A5.1 was increased (pc =0.005; OR 1.88). MIC-A5.1 together with HLA-DRB1*03 increases the risk for SLE development, pc <0.000001; OR 9.71....
302

The role of SHP2 in metastatic breast cancer

Hao Chen (12447552) 22 April 2022 (has links)
<p>  </p> <p>Metastatic breast cancer (MBC) is an extremely recalcitrant disease capable of overcoming targeted therapies and evading immune surveillance via the engagement of complicated signaling networks. Resistance to targeted therapies and therapeutic failure of immune checkpoint blockade (ICB) are two major challenges in treating MBC. To survive in the dynamic tumor microenvironment (TME) during metastatic progression, shared signaling nodes are required for MBC cells to regulate the signaling networks efficiently, which are potential multifunctional therapeutic targets. SH2 containing protein tyrosine phosphatase-2 (SHP2) is a druggable oncogenic phosphatase that is a key shared node in both tumor cells and immune cells. How tumor-cell autonomous SHP2 manages its signaling inputs and outputs to facilitate the growth of tumor cells, drug resistance, immunosuppression, and the limited response of ICB in MBC is not fully understood. Herein, we used inducible genetic depletion and two distinct types of pharmacological inhibitors to investigate anti-tumor effects with immune reprogramming during SHP2 targeting. </p> <p>We first focus on the signaling inputs and outputs of SHP2. We find that phosphorylation of SHP2 at Y542 predicts the survival rates of breast cancer patients and their immune profiles. Phosphorylation of SHP2 at Y542 is elevated with differential activation mechanisms under a growth-factor-induced and extracellular matrix (ECM)-rich culture environment. Phosphorylation of SHP2 at Y542 is also elevated in HER2 positive MBC cells upon acquired resistance to the HER2 kinase inhibitor, neratinib. The resistant cells can be targeted by SHP2 inhibitors. SHP2 inhibitors block ERK1/2 and AKT signaling and readily prevented MBC cell growth induced by multiple growth factors. Inhibition of SHP2 also blocks these signaling events generated from the ECM signaling. In fact, the inhibitory effects of SHP2 blockade are actually enhanced in the ECM-rich culture environment. We utilize the <em>in vitro</em> T-cell killing assays and demonstrate that pretreatment of tumor cells with FGF2 and PDGF reduces the cytotoxicity of CD8+ T cells in a SHP2-dependent manner. Both growth factors and ECM-rich culture environment transcriptionally induce PD-L1 via SHP2. SHP2 inhibition balances MAPK signaling and STAT1 signaling, which prevents growth factor-mediated suppression of INF-γ-induced expression of MHC class I. </p> <p>Next, we evaluate the efficacy of SHP2 inhibitors. Blockade of SHP2 in the adjuvant setting decreased pulmonary metastasis <em>in vivo</em> and extended the survival of systemic tumor-bearing mice. Tumor-cell autonomous depletion of SHP2 reduces pulmonary metastasis and relieves exhaustion markers on CD8+ and CD4+ cells. Meanwhile, both systemic SHP2 inhibition and tumor-cell autonomous SHP2 depletion reduce tumor-infiltrated CD4+ T cells and M2-polarized tumor associated macrophages. </p> <p>Finally, we investigate potential combination therapies with SHP2 inhibitors. The combination of SHP2 inhibitors and FGFR-targeted kinase inhibitors synergistically blocks the growth of MBC cells. Pharmacological inhibition SHP2 sensitizes MBC cells growing in the lung to α-PD-L1 antibody treatment via relieving T cell exhaustion induced by ICB. </p> <p>Overall, our findings support the conclusion that MBC cells are capable of simultaneously engaging several survival pathways and immune-suppressive mechanisms via SHP2 in response to multiple growth factors and ECM signaling. Inhibition of SHP2, potentially in combination with other targeted agents and ICB, holds promise for the therapeutic management of MBC.</p>
303

Développement d’approches de contrôle de qualité pour la caractérisation de l’immunopeptidome de cellules infectées par les coronavirus

Despault-Duquette, Jérôme 12 1900 (has links)
La présentation de l’antigène est un mécanisme par lequel les cellules nucléées présentent un court peptide sur la molécule de classe 1 du Complexe Majeur d’Histocompatibilité (CMH-1) codée par les gènes « antigènes d’histocompatibilité humains ». Le terme “immunopeptidomique” est utilisé pour décrire l’ensemble des peptides associés aux molécules du CMH-1. Les cellules T CD8+ patrouillent l’organisme, s’attachent à la molécule CMH-1 par leur récepteur T et détruisent les cellules affichant un peptide atypique. Ce domaine présente un grand intérêt au niveau du traitement des infections virales et dans la conception de vaccins. Compte tenu que les coronavirus ont été à l’origine de trois épidémies durant les 20 dernières années, et que de multiples souches circulent chez l’humain ainsi que dans le règne animal, il est impératif de développer des vaccins universels qui pourrait prévenir de futurs événement épidémiologiques mondiaux reliés aux coronavirus. L’immunopeptidomique souffre d'un manque de protocoles normalisés et de contrôle et d’assurance de la qualité des échantillons afin de libérer tout son potentiel dans la recherche biomédicale. Dans le cadre de cette étude, la spectrométrie de masse, la cytométrie de flux et des approches bio-informatiques ont été utilisées pour développer des protocoles de contrôle de qualité pour la caractérisation de l’immunopeptidome de cellules infectées par les coronavirus. Nous avons isolé et analysé l’immunopeptidome de cellules MRC-5 avant et après infection par le coronavirus humain OC-43. En plus d’observer une forte baisse de l’abondance des molécules HLA et de la variété des peptides présentés après l’infection, 9 peptides viraux ont été isolés à partir des molécules du CMH-1. Ces peptides pourraient être utilisés afin de contribuer à formuler un vaccin pan-coronavirus qui élicite une réponse balancée entre la réponse humorale et la réponse cytotoxique. / Antigen presentation is a mechanism by which nucleated cells present a short peptide on the Major Histocompatibility Complex (MHC-1) class 1 molecule encoded by the “human histocompatibility antigen” genes. The term "immunopeptidomics" is used to describe the set of peptides associated with MHC-1 molecules. CD8+ T cells patrol the body, attach to the MHC-1 molecule through their T receptor and destroy cells displaying an atypical peptide. This field is of great interest in the treatment of viral infections and in vaccine design. Given that coronaviruses have been responsible for three epidemics in the last 20 years, and that multiple strains circulate in humans and animals, it is imperative to develop universal vaccines that could prevent future global epidemiological events related to coronaviruses. Immunopeptidomics suffers from a lack of standardized protocols and sample quality control and assurance to unleash its full potential in biomedical research. In this study, mass spectrometry, flow cytometry, and bioinformatics approaches were used to develop quality control protocols for characterizing the immunopeptidome of coronavirusinfected cells. We isolated and analyzed the immunopeptidome of MRC-5 cells before and after infection with human coronavirus OC-43. In addition to observing a strong decrease in the abundance of HLA molecules and in the variety of peptides presented after infection, 9 viral peptides were isolated from MHC-1 molecules. These peptides could be used to help formulate a pan-coronavirus vaccine that elicits a balanced response between humoral and cytotoxic responses.
304

Exploring the landscape of actionable HLA I-associated tumor antigens across cancers

Apavaloaei, Anca 08 1900 (has links)
Presque toutes les cellules nucléées expriment des peptides associés au CMH I (HLA I chez l’humain)(MAP) qui sont échantillonnés à partir du protéome cellulaire et transportés vers la surface cellulaire pour inspection par les lymphocytes T CD8. En tant que tel, la collection de MAP à la surface des cellules, ou immunopeptidome, informe les lymphocytes T CD8 de l’état cellulaire interne. L’immunosurveillance du cancer repose sur la capacité des lymphocytes T CD8 à reconnaître les MAP anormaux sur les cellules tumorales et à les éliminer tout en épargnant les cellules saines. Par conséquent, l’existence du cancer indique que bien souvent, les lymphocytes T CD8 spécifiques à la tumeur sont impuissants, dysfonctionnels ou incapables d’exercer leur fonction. Les vaccins anticancéreux peuvent actionner la destruction des tumeurs en stimulant la reconnaissance des MAP anormaux. Toutefois, le développement de vaccins anticancéreux efficaces est entravé par le manque de MAP exploitables, ou antigènes tumoraux (TA), exprimés exclusivement sur les cellules tumorales. La recherche et l’identification de TA ont été largement limitées aux MAP dérivés de mutations non synonymes situées dans des exons canoniques codant pour des protéines. Ces régions génomiques ne représentent que 2% du génome humain. Le fait que les MAP puissent potentiellement dériver de la traduction non canonique de toutes les régions génomiques n’a été pleinement compris que récemment. Ici, nous avons utilisé la protéogénomique pour découvrir des TA exploitables dérivés de produits de traduction canoniques et non canoniques partagés au sein ou entre divers types de cancers humains. Premièrement, nous avons utilisé des cellules souches pluripotentes induites (iPSC) pour identifier les MAP associés à la pluripotence (paMAP) étant partagés par les cellules cancéreuses. Les antigènes pluripotents sont exprimés dans les tissus embryonnaires et absents des tissus adultes sains, mais anormalement réexprimés par les cellules cancéreuses. Ainsi, bien qu'ils ne soient pas mutés, les paMAP constituent des cibles idéales et spécifiques au cancer. Nous avons identifié un ensemble de 48 paMAP dérivés de transcrits codants et non codants (48 %) impliqués dans le maintien de la pluripotence et exprimés de manière aberrante dans plusieurs types de cancer. Ainsi, bien qu’elles proviennent de différents types de cellules et de tissus, des tumeurs 4 distinctes convergent vers un programme transcriptionnel associé à la pluripotence. En effet, l’expression des paMAP dans les cancers est corrélée à l’hypométhylation récurrente de leurs gènes sources, la présence d’aberrations génomiques courantes et l’adoption par les tumeurs de stratégies d’évasion immunitaire communes. Enfin, comme plusieurs paMAP sont immunogènes, leur utilisation comme cibles dans des vaccins anticancéreux pourrait entrer en synergie avec les inhibiteurs disponibles des voies d'évasion immunitaire et améliorer le traitement de plusieurs cancers agressifs. Ensuite, nous avons évalué l’ensemble des TA ayant un potentiel thérapeutique dans deux types de tumeurs présentant une charge mutationnelle particulièrement élevée, le mélanome et le cancer du poumon non à petites cellules (NSCLC). Nous avons constaté que les TA mutés (mTSAs) représentent une minorité (1 %) des TA exploitables dans ces deux types de cancer. Cela peut s'expliquer par une faible expression d'ARN de la plupart des mutations non synonymes ainsi que par leur localisation en dehors des régions génomiques les plus efficaces pour la génération de MAP. En revanche, 99 % des TA dérivent de séquences génomiques non mutées spécifiques au cancer (aeTSA), surexprimées dans le cancer (TAA) ou spécifiques à la lignée cellulaire d'origine (LSA, exprimés par les mélanocytes ou par les cellules épithéliales pulmonaires, pour le mélanome et le NSCLC, respectivement). Tout comme les paMAP, environ 50 % des aeTSA identifiés dans le mélanome et le NSCLC proviennent de séquences non canoniques et sont régulés de manière épigénétique. Alors que les mTSA sont exclusivement spécifiques à chaque patient patient, les aeTSA sont partagés entre les échantillons tumoraux. De plus, leur absence dans les tissus normaux, leur abondance et leur capacité à activer les lymphocytes T CD8 en font des cibles idéales pour traiter les mélanomes et les NSCLC. En conclusion, cette thèse fournit un aperçu de la biogenèse de différents types de TA dans diverses cohortes de patients et ouvre la voie au développement d’immunothérapies ciblées et efficaces contre une grande variété de cancers. / Nearly all nucleated cells express MHC I (HLA I in humans)-associated peptides (MAPs) which are sampled from the cellular proteome and transported to the cell surface for inspection by CD8 T cells. As such, the collection of cell-surface MAPs, or the immunopeptidome, informs CD8 T cells on the inner cell state. Cancer immunosurveillance relies on the capacity of CD8 T cells to recognize abnormal MAPs on tumor cells and eliminate them while sparing healthy cells. Hence, the existence of cancer indicates that tumor-specific CD8 T cells are underpowered, dysfunctional or inhibited from exerting their function. Anti-cancer vaccines can boost tumor killing by stimulating the recognition of abnormal MAPs. The development of effective anti-cancer vaccines is limited by the identification of actionable MAPs, or tumor antigens (TAs), expressed exclusively on tumor cells. The TA search space has been largely limited to MAPs derived from non-synonymous mutations in canonical protein-coding exons which represent a mere 2% of the human genome. That MAPs can derive from the non-canonical translation of potentially all genomic regions has only recently been fully appreciated. Herein, we used proteogenomics to discover actionable TAs derived from canonical and non-canonical translation products shared within or across different types of human cancer. First, we used induced pluripotent stem cells (iPSCs) to identify pluripotency-associated MAPs (paMAPs) shared by cancer cells. Pluripotency antigens are restricted to embryonic tissues and absent from healthy adult tissues but abnormally re-expressed by cancer cells, which makes them ideal tumor-specific targets despite being unmutated. We identified a set of 46 paMAPs derived from coding and allegedly non-coding (48%) transcripts involved in pluripotency maintenance and aberrantly expressed in multiple cancer types. Thus, despite originating from different cell types and tissues, distinct tumor types converged towards a pluripotency-associated transcriptional program. Indeed, the expression of paMAPs across cancers correlated with recurrent source gene hypomethylation, genomic aberrations, and immune evasion properties. Several paMAPs were immunogenic, thus their targeting could synergize with available inhibitors of immune evasion pathways to improve the outcome of multiple aggressive cancers. 7 Next, we evaluated the actionable TA landscape of two tumor types with particularly high mutational load, melanoma and non-small cell lung cancer (NSCLC). We found that mutated TAs (mTSAs) represent a minority (1%) of actionable TAs in both cancer types, which can be explained by a low RNA expression of most non-synonymous mutations and their localization outside genomic regions proficient for MAP generation. By contrast, 99% of TAs derived from unmutated genomic sequences specific to cancer (aeTSAs), overexpressed in cancer (TAAs), or specific to the cell lineage of origin (LSAs, expressed by melanocytes or by lung epithelial cells, for melanoma and NSCLC LSAs, respectively). As for paMAPs, around 50% of aeTSAs in melanoma and NSCLC were non-canonical and were epigenetically regulated. Whereas mTSAs were exclusively patient-specific, aeTSAs were shared among tumor samples and exhibited all characteristics of targetable TAs, including tumor-specificity, high abundance, and immunogenicity. Altogether, this thesis provides insights into the biogenesis of different TA types in various patient cohorts and paves the way for the development of effective TA-based immunotherapies against a large variety of cancers.
305

Estudo caso-controle da região HLA de pacientes com Granulomatose com poliangeíte / Case-control study of HLA region in Brazilian carriers of Granulomatosis with Polyangiitis (Wegener\'s)

Tavares, Marcos Soares 19 December 2016 (has links)
Os alelos HLA-DPB1*04 e HLA-DRB1*15 estão fortemente associados à Granulomatose com poliangeíte (GPA). Neste estudo, analisamos se os pacientes brasileiros com diagnóstico de GPA apresentam uma base genética na região HLA. Conduzimos um estudo caso-controle, em que analisamos os alelos da região HLA classe I e II em 55 pacientes com diagnóstico de GPA, atendidos no ambulatório de Vasculites Pulmonares do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, e comparamos com os resultados de 110 controles saudáveis. Comparamos também quatro diferentes apresentações clínicas da GPA e a positividade do anticorpo anticitoplasma de neutrófilos (ANCA) com os alelos da região HLA classe I e II. Foi também construída uma árvore de decisões, usando o algoritmo de CART, para a verificação da associação entre os alelos HLA e GPA. Como resultados, observamos que a GPA esteve fortemente associada à presença dos alelos DPB1*04 e DRB1*15 (p = 0,007, odds ratio [OR]: 2,9, 95% intervalo de confiança [IC]: 1,09-3,8; p = 0,006, OR: 2,87, 95% IC: 1,44-4,75, respectivamente) e não à presença do alelo DRB1*04. O alelo DRB1*13 esteve associado com proteção contra GPA (p = 0,042, OR: 0,42, 95% CI: 0,21-0,99). O alelo DPB1*04 esteve significativamente associado a GPA e ANCA-C positivo (OR: 5,47) e à presença de insuficiência renal aguda (p = 0,01037). Concluímos que houve uma interdependência significativa entre os alelos DPB1*0401, DPB1*0402, DRB1*13, C*2 e GPA. Na população estudada, quando o alelo DPB1*04 esteve presente em homozigose, o risco de GPA foi de 81%. Quando o alelo DPB1*0401 esteve ausente ou em heterozigose com o DPB1*0402, como o outro alelo, ou DPB1*0402 esteve em homozigose, o risco da GPA foi de 52,9%. No caso de ausência dos alelos DPB1*0401, DPB1*0402 e DRB1*13, a presença do alelo C*2 aumentou o risco da GPA para 62,5%. Finalmente, na ausência do alelo DPB1*0401 e DPB1*0402 e na presença do alelo DRB1*13, o risco de GPA diminuiu para 0% / The alleles HLA-DPB1*04 and HLA-DRB1*15 are strongly associated with granulomatosis with polyangiitis (GPA). In this study, we examined whether Brazilian patients with GPA had an HLA region genetic background. We conducted a case-control study, in which we analysed alleles of HLA region class I and II from 55 patients with GPA (at the Pulmonary Vasculitis Clinic of the University of São Paulo) and compared the results with those from 110 healthy controls. Comparisons were also performed for 4 different clinical presentations of GPA and anti-neutrophil cytoplasmic antibody (ANCA) positivity and the HLA class I and II region alleles. A tree model decision analysis was conducted using CART algorithm. Our results showed that GPA was strongly associated with alleles DPB1*04 and DRB1*15 (p = 0.007, odds ratio [OR]: 2.9, 95% confidence interval [CI]: 1.09-3.8; p = 0.006, OR: 2.87, 95% CI: 1.44-4.75, respectively) and not with the allele DRB1*04. DRB1*13 allele was associated with protection against GPA (p = 0.042, OR: 0.42, 95% CI: 0.21-0.99). DPB1*04 was significantly associated with GPA plus positive C-ANCA (OR: 5.47) and acute renal failure (p = 0.01037). We concluded that there was a significant interdependence among alleles and GPA. In our population, when allele DPB1*04 was presented in homozygous, the risk of GPA was 81%. When DPB1*0401 allele was absent or heterozygous with DPB1*0402 as the other allele, or DPB1*0402 was homozygous, the risk of disease was 52.9%. If DPB1*0401, DPB1*0402, and DRB1*13 were absent, the presence of C*2 increased the risk of GPA to 62.5%. Finally, in the absence of DPB1*0401 and DPB1*0402 and the presence of DRB1*13, the risk of GPA decreased to 0%
306

Estudo caso-controle da região HLA de pacientes com Granulomatose com poliangeíte / Case-control study of HLA region in Brazilian carriers of Granulomatosis with Polyangiitis (Wegener\'s)

Marcos Soares Tavares 19 December 2016 (has links)
Os alelos HLA-DPB1*04 e HLA-DRB1*15 estão fortemente associados à Granulomatose com poliangeíte (GPA). Neste estudo, analisamos se os pacientes brasileiros com diagnóstico de GPA apresentam uma base genética na região HLA. Conduzimos um estudo caso-controle, em que analisamos os alelos da região HLA classe I e II em 55 pacientes com diagnóstico de GPA, atendidos no ambulatório de Vasculites Pulmonares do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, e comparamos com os resultados de 110 controles saudáveis. Comparamos também quatro diferentes apresentações clínicas da GPA e a positividade do anticorpo anticitoplasma de neutrófilos (ANCA) com os alelos da região HLA classe I e II. Foi também construída uma árvore de decisões, usando o algoritmo de CART, para a verificação da associação entre os alelos HLA e GPA. Como resultados, observamos que a GPA esteve fortemente associada à presença dos alelos DPB1*04 e DRB1*15 (p = 0,007, odds ratio [OR]: 2,9, 95% intervalo de confiança [IC]: 1,09-3,8; p = 0,006, OR: 2,87, 95% IC: 1,44-4,75, respectivamente) e não à presença do alelo DRB1*04. O alelo DRB1*13 esteve associado com proteção contra GPA (p = 0,042, OR: 0,42, 95% CI: 0,21-0,99). O alelo DPB1*04 esteve significativamente associado a GPA e ANCA-C positivo (OR: 5,47) e à presença de insuficiência renal aguda (p = 0,01037). Concluímos que houve uma interdependência significativa entre os alelos DPB1*0401, DPB1*0402, DRB1*13, C*2 e GPA. Na população estudada, quando o alelo DPB1*04 esteve presente em homozigose, o risco de GPA foi de 81%. Quando o alelo DPB1*0401 esteve ausente ou em heterozigose com o DPB1*0402, como o outro alelo, ou DPB1*0402 esteve em homozigose, o risco da GPA foi de 52,9%. No caso de ausência dos alelos DPB1*0401, DPB1*0402 e DRB1*13, a presença do alelo C*2 aumentou o risco da GPA para 62,5%. Finalmente, na ausência do alelo DPB1*0401 e DPB1*0402 e na presença do alelo DRB1*13, o risco de GPA diminuiu para 0% / The alleles HLA-DPB1*04 and HLA-DRB1*15 are strongly associated with granulomatosis with polyangiitis (GPA). In this study, we examined whether Brazilian patients with GPA had an HLA region genetic background. We conducted a case-control study, in which we analysed alleles of HLA region class I and II from 55 patients with GPA (at the Pulmonary Vasculitis Clinic of the University of São Paulo) and compared the results with those from 110 healthy controls. Comparisons were also performed for 4 different clinical presentations of GPA and anti-neutrophil cytoplasmic antibody (ANCA) positivity and the HLA class I and II region alleles. A tree model decision analysis was conducted using CART algorithm. Our results showed that GPA was strongly associated with alleles DPB1*04 and DRB1*15 (p = 0.007, odds ratio [OR]: 2.9, 95% confidence interval [CI]: 1.09-3.8; p = 0.006, OR: 2.87, 95% CI: 1.44-4.75, respectively) and not with the allele DRB1*04. DRB1*13 allele was associated with protection against GPA (p = 0.042, OR: 0.42, 95% CI: 0.21-0.99). DPB1*04 was significantly associated with GPA plus positive C-ANCA (OR: 5.47) and acute renal failure (p = 0.01037). We concluded that there was a significant interdependence among alleles and GPA. In our population, when allele DPB1*04 was presented in homozygous, the risk of GPA was 81%. When DPB1*0401 allele was absent or heterozygous with DPB1*0402 as the other allele, or DPB1*0402 was homozygous, the risk of disease was 52.9%. If DPB1*0401, DPB1*0402, and DRB1*13 were absent, the presence of C*2 increased the risk of GPA to 62.5%. Finally, in the absence of DPB1*0401 and DPB1*0402 and the presence of DRB1*13, the risk of GPA decreased to 0%

Page generated in 0.0882 seconds