Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Gastroenterology ; 2024 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-38467384

RESUMO

BACKGROUND & AIMS: Histologic evaluation of gut biopsies is a cornerstone for diagnosis and management of celiac disease (CeD). Despite its wide use, the method depends on proper biopsy orientation, and it suffers from interobserver variability. Biopsy proteome measurement reporting on the tissue state can be obtained by mass spectrometry analysis of formalin-fixed paraffin-embedded tissue. Here we aimed to transform biopsy proteome data into numerical scores that give observer-independent measures of mucosal remodeling in CeD. METHODS: A pipeline using glass-mounted formalin-fixed paraffin-embedded sections for mass spectrometry-based proteome analysis was established. Proteome data were converted to numerical scores using 2 complementary approaches: a rank-based enrichment score and a score based on machine learning using logistic regression. The 2 scoring approaches were compared with each other and with histology analyzing 18 patients with CeD with biopsies collected before and after treatment with a gluten-free diet as well as biopsies from patients with CeD with varying degree of remission (n = 22). Biopsies from individuals without CeD (n = 32) were also analyzed. RESULTS: The method yielded reliable proteome scoring of both unstained and H&E-stained glass-mounted sections. The scores of the 2 approaches were highly correlated, reflecting that both approaches pick up proteome changes in the same biological pathways. The proteome scores correlated with villus height-to-crypt depth ratio. Thus, the method is able to score biopsies with poor orientation. CONCLUSIONS: Biopsy proteome scores give reliable observer and orientation-independent measures of mucosal remodeling in CeD. The proteomic method can readily be implemented by nonexpert laboratories in parallel to histology assessment and easily scaled for clinical trial settings.

2.
J Autoimmun ; 146: 103241, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38754235

RESUMO

Many antibody responses induced by infection, vaccination or autoimmunity show signs of convergence across individuals with epitope-dependent selection of particular variable region gene segments and complementarity determining region 3 properties. However, not much is known about the relationship between antigen-specific effector cells and antigen-specific precursors present in the naïve B-cell repertoire. Here, we sought to address this relationship in the context of celiac disease, where there is a stereotyped autoantibody response against the enzyme transglutaminase 2 (TG2). By generating TG2-specific monoclonal antibodies from both duodenal plasma cells and circulating naïve B cells, we demonstrate a discord between the naïve TG2-specific repertoire and the cells that are selected for autoantibody production. Hence, the naïve repertoire does not fully reflect the epitope preference and gene usage observed for memory B cells and plasma cells. Instead, distinct naïve B cells that target particular TG2 epitopes appear to be selectively activated at the expense of TG2-binding B cells targeting other epitopes.


Assuntos
Autoanticorpos , Linfócitos B , Doença Celíaca , Epitopos de Linfócito B , Proteínas de Ligação ao GTP , Ativação Linfocitária , Proteína 2 Glutamina gama-Glutamiltransferase , Transglutaminases , Doença Celíaca/imunologia , Humanos , Autoanticorpos/imunologia , Transglutaminases/imunologia , Epitopos de Linfócito B/imunologia , Proteínas de Ligação ao GTP/imunologia , Ativação Linfocitária/imunologia , Linfócitos B/imunologia , Plasmócitos/imunologia , Plasmócitos/metabolismo , Feminino , Anticorpos Monoclonais/imunologia , Epitopos/imunologia , Masculino , Adulto , Duodeno/imunologia , Duodeno/patologia
3.
Eur J Immunol ; 52(9): 1474-1481, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35715890

RESUMO

The adaptive immune response of celiac disease (CeD) involves presentation of gluten peptides to CD4+ T cells by transglutaminase 2 (TG2) specific B cells. This B-cell/T-cell crosstalk is facilitated by involvement of TG2:gluten peptide complexes that act principally in the form of enzyme-substrate intermediates. Here, we have addressed how gluten peptide affinity and complex stability in the presence of secondary substrates affect the uptake of TG2:gluten peptide complexes by TG2-specific B cells and the activation of gluten-specific T cells. We studied affinity of various gluten peptides for TG2 by biochemical assay, and monitored uptake of gluten peptides by TG2-specific B cells by flow cytometry. Crosstalk between TG2-specific B cells and gluten-specific T cells was assayed with transfectants expressing antigen receptors derived from CeD patients. We found that gluten peptides with high TG2 affinity showed better uptake by TG2-specific B cells. Uptake by B cells, and subsequent activation of T cells, was negatively affected by polyamines acting as secondary TG2 substrates. These results show that affinity between gluten peptide and TG2 governs the selection of T-cell epitopes via enhanced uptake of TG2:gluten complexes by TG2-specific B cells, and that exogenous polyamines can influence the CeD immune responses by disrupting TG2:gluten complexes.


Assuntos
Doença Celíaca , Glutens , Proteínas de Ligação ao GTP/metabolismo , Humanos , Peptídeos/metabolismo , Poliaminas , Proteína 2 Glutamina gama-Glutamiltransferase , Linfócitos T , Transglutaminases
4.
Proc Natl Acad Sci U S A ; 116(30): 15134-15139, 2019 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-31285344

RESUMO

B cells play important roles in autoimmune diseases through autoantibody production, cytokine secretion, or antigen presentation to T cells. In most cases, the contribution of B cells as antigen-presenting cells is not well understood. We have studied the autoantibody response against the enzyme transglutaminase 2 (TG2) in celiac disease patients by generating recombinant antibodies from single gut plasma cells reactive with discrete antigen domains and by undertaking proteomic analysis of anti-TG2 serum antibodies. The majority of the cells recognized epitopes in the N-terminal domain of TG2. Antibodies recognizing C-terminal epitopes interfered with TG2 cross-linking activity, and B cells specific for C-terminal epitopes were inefficient at taking up TG2-gluten complexes for presentation to gluten-specific T cells. The bias toward N-terminal epitopes hence reflects efficient T-B collaboration. Production of antibodies against N-terminal epitopes coincided with clinical onset of disease, suggesting that TG2-reactive B cells with certain epitope specificities could be the main antigen-presenting cells for pathogenic, gluten-specific T cells. The link between B cell epitopes, antigen presentation, and disease onset provides insight into the pathogenic mechanisms of a T cell-mediated autoimmune condition.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Linfócitos B/imunologia , Doença Celíaca/imunologia , Epitopos de Linfócito B/imunologia , Proteínas de Ligação ao GTP/imunologia , Linfócitos T/imunologia , Transglutaminases/imunologia , Idade de Início , Células Apresentadoras de Antígenos/patologia , Autoanticorpos/biossíntese , Autoanticorpos/genética , Autoantígenos/genética , Autoantígenos/imunologia , Linfócitos B/patologia , Doença Celíaca/genética , Doença Celíaca/patologia , Duodeno/imunologia , Duodeno/patologia , Epitopos de Linfócito B/química , Epitopos de Linfócito B/genética , Proteínas de Ligação ao GTP/química , Proteínas de Ligação ao GTP/genética , Glutens/química , Glutens/imunologia , Humanos , Soros Imunes/química , Cadeias Leves de Imunoglobulina/biossíntese , Cadeias Leves de Imunoglobulina/genética , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Proteína 2 Glutamina gama-Glutamiltransferase , Linfócitos T/patologia , Transglutaminases/química , Transglutaminases/genética
5.
Proteomics ; 21(23-24): e2100057, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34633755

RESUMO

Celiac disease (CeD) is a prevalent intestinal disorder that only develops in genetically susceptible individuals when they mount a harmful CD4+ T-cell response towards gluten peptides. Intake of gluten leads to inflammation and remodeling of the small intestine with symptoms such as nausea and diarrhea. The only current treatment is a lifelong gluten free diet. The immunological basis for CeD is well characterized but the mechanisms that drive intestinal remodeling are still poorly understood. Transcriptome or proteome analysis of intestinal biopsies gives a global snapshot of all processes that occur in the tissue, including alterations in the epithelial cell layer. This paper will introduce concepts of intestinal remodeling, recapitulate the current understanding of CeD pathogenesis and discuss findings from relevant tissue "omics" studies. On the basis of this review, I give perspectives on what tissue "omics" studies can tell us about disease pathogenesis with a particular focus on the gluten induced intestinal remodeling.


Assuntos
Doença Celíaca , Biópsia , Dieta Livre de Glúten , Glutens , Humanos , Mucosa Intestinal , Intestino Delgado
6.
Am J Pathol ; 188(7): 1563-1579, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29684362

RESUMO

Global characterization of tissue proteomes from small amounts of biopsy material has become feasible because of advances in mass spectrometry and bioinformatics tools. In celiac disease (CD), dietary gluten induces an immune response that is accompanied by pronounced remodeling of the small intestine. Removal of gluten from the diet abrogates the immune response, and the tissue architecture normalizes. In this study, differences in global protein expression of small intestinal biopsy specimens from CD patients were quantified by analyzing formalin-fixed, paraffin-embedded material using liquid chromatography-mass spectrometry and label-free protein quantitation. Protein expression was compared in biopsy specimens collected from the same patients before and after 1-year treatment with gluten-free diet (n = 10) or before and after 3-day gluten provocation (n = 4). Differential expression of proteins in particular from mature enterocytes, neutrophils, and plasma cells could distinguish untreated from treated CD mucosa, and Ig variable region IGHV5-51 expression was found to serve as a CD-specific marker of ongoing immune activation. In patients who had undergone gluten challenge, coordinated up-regulation of wound response proteins, including the CD autoantigen transglutaminase 2, was observed. Our study provides a global and unbiased assessment of antigen-driven changes in protein expression in the celiac intestinal mucosa.


Assuntos
Biomarcadores/análise , Doença Celíaca/complicações , Enteropatias/diagnóstico , Intestino Delgado/metabolismo , Espectrometria de Massas/métodos , Proteoma/análise , Adulto , Dieta Livre de Glúten , Feminino , Humanos , Enteropatias/etiologia , Enteropatias/metabolismo , Intestino Delgado/patologia , Masculino , Pessoa de Meia-Idade , Adulto Jovem
7.
J Biol Chem ; 291(49): 25542-25552, 2016 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-27784785

RESUMO

Transglutaminase 2 (TG2) is a Ca2+-dependent cross-linking enzyme involved in the pathogenesis of CD. We have previously characterized a panel of anti-TG2 mAbs generated from gut plasma cells of celiac patients and identified four epitopes (epitopes 1-4) located in the N-terminal part of TG2. Binding of the mAbs induced allosteric changes in TG2. Thus, we aimed to determine whether these mAbs could influence enzymatic activity through modulation of TG2 susceptibility to oxidative inactivation and Ca2+ affinity. All tested epitope 1 mAbs, as well as 679-14-D04, which recognizes a previously uncharacterized epitope, prevented oxidative inactivation and increased Ca2+ sensitivity of TG2. We have identified crucial residues for binding of 679-14-D04 located within a Ca2+ binding site. Epitope 1 mAbs and 679-14-D04, although recognizing separate epitopes, behaved similarly when assessing their effect on TG2 conformation, suggesting that the shared effects on TG2 function can be explained by induction of the same conformational changes. None of the mAbs targeting other epitopes showed these effects, but epitope 2 mAbs reduced the rate of TG2-catalyzed reactions. Collectively, these effects could be relevant to the pathogenesis of CD. In A20 B cells transduced with TG2-specific B-cell receptor, epitope 2-expressing cells had poorer uptake of TG2-gluten complexes and were less efficient in gluten epitope presentation to T cells than cells expressing an epitope 1 receptor. Thus, the ability of epitope 1-targeting B cells to keep TG2 active and protected from oxidation might explain why generation of epitope 1-targeting plasma cells seems to be favored in celiac patients.


Assuntos
Autoanticorpos/imunologia , Doença Celíaca/imunologia , Epitopos/imunologia , Proteínas de Ligação ao GTP/imunologia , Glutens/imunologia , Transglutaminases/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Doença Celíaca/genética , Doença Celíaca/patologia , Linhagem Celular Tumoral , Proteínas de Ligação ao GTP/genética , Antígenos HLA-DQ/genética , Antígenos HLA-DQ/imunologia , Humanos , Camundongos , Plasmócitos/imunologia , Plasmócitos/patologia , Proteína 2 Glutamina gama-Glutamiltransferase , Transglutaminases/genética
8.
Amino Acids ; 49(3): 489-500, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27394141

RESUMO

In the extracellular environment, the enzyme transglutaminase 2 (TG2) is involved in cell-matrix interactions through association with the extracellular matrix protein, fibronectin (FN). The 45 kDa gelatin-binding domain of FN (45FN) is responsible for the binding to TG2. Previous studies have demonstrated that the FN-binding site of TG2 is located in the N-terminal domain of the enzyme although with conflicting results regarding the specific residues involved. Here we have mapped the FN interaction site of human TG2 by use of hydrogen/deuterium exchange coupled with mass spectrometry, and we confirm that the FN-binding site is located in the N-terminal domain of TG2. Furthermore, by combination of site-directed mutagenesis and surface plasmon resonance analysis we have identified the TG2 residues K30, R116 and H134 as crucial to maintain the high affinity interaction with FN. Mutation of all three residues simultaneously reduced binding to 45FN by more than 2000-fold. We also identified residues in the catalytic core domain of TG2 that contributed to FN binding, hence extending the binding interface between TG2 and FN. This study provides new insights into the high affinity interaction between TG2 and FN.


Assuntos
Fibronectinas/química , Proteínas de Ligação ao GTP/química , Domínios e Motivos de Interação entre Proteínas , Transglutaminases/química , Sequência de Aminoácidos , Anticorpos/química , Anticorpos/isolamento & purificação , Domínio Catalítico , Clonagem Molecular , Medição da Troca de Deutério , Escherichia coli/genética , Escherichia coli/metabolismo , Fibronectinas/genética , Fibronectinas/metabolismo , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/metabolismo , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Humanos , Cinética , Modelos Moleculares , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Proteína 2 Glutamina gama-Glutamiltransferase , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Ressonância de Plasmônio de Superfície , Transglutaminases/genética , Transglutaminases/metabolismo
10.
J Immunol ; 193(9): 4497-506, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-25261484

RESUMO

Even though MHC class II is a dominant susceptibility factor for many diseases, culprit T cell epitopes presented by disease-associated MHC molecules remain largely elusive. T cells of celiac disease lesions recognize cereal gluten epitopes presented by the disease-associated HLA molecules DQ2.5, DQ2.2, or DQ8. Employing celiac disease and complex gluten Ag digests as a model, we tested the feasibility of using DQ2.5 and DQ2.2 as an affinity matrix for identification of disease-relevant T cell epitopes. Known gluten T cell epitope peptides were enriched by DQ2.5, whereas a different set of peptides was enriched by DQ2.2. Of 86 DQ2.2-enriched peptides, four core sequences dominated. One of these core sequences is a previously known epitope and two others are novel epitopes. The study provides insight into the selection of gluten epitopes by DQ2.2. Furthermore, the approach presented is relevant for epitope identification in other MHC class II-associated disorders.


Assuntos
Mapeamento de Epitopos , Epitopos de Linfócito T/imunologia , Glutens/imunologia , Antígenos HLA-DQ/imunologia , Linfócitos T/imunologia , Sequência de Aminoácidos , Doença Celíaca/imunologia , Linhagem Celular , Cromatografia em Gel , Mapeamento de Epitopos/métodos , Gliadina/química , Gliadina/imunologia , Glutens/química , Antígenos HLA-DQ/química , Humanos , Peptídeos/química , Peptídeos/imunologia , Ligação Proteica , Triticum/imunologia
11.
Dig Dis ; 33(2): 115-121, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25925911

RESUMO

BACKGROUND: Celiac disease is a multifactorial and polygenic disease with autoimmune features. The disease is caused by an inappropriate immune response to gluten. Elimination of gluten from the diet leads to disease remission, which is the basis for today's treatment of the disease. There is an unmet need for new alternative treatments. KEY MESSAGES: Genetic findings point to adaptive immunity playing a key role in the pathogenesis of celiac disease. MHC is by far the single most important genetic factor in the disease. In addition, a number of non-MHC genes, the majority of which have functions related to T cells and B cells, also contribute to the genetic predisposition, but each of them has modest effect. The primary MHC association is with HLA-DQ2 and HLA-DQ8. These HLA molecules present gluten epitopes to CD4+ T cells which can be considered to be the master regulators of the immune reactions that lead to the disease. The epitopes which the T cells recognize are usually deamidated, and this deamidation is mediated by the enzyme transglutaminase 2 (TG2). Celiac disease patients have disease-specific antibodies. In addition to antibodies to gluten, these include autoantibodies to TG2. Antibodies to deamidated gluten are nearly as specific for celiac disease as the anti-TG2 antibodies. Both types of antibodies appear only to be produced in subjects who are HLA-DQ2 or HLA-DQ8 when they are consuming gluten. CONCLUSION: It is hardly coincidental that TG2 is implicated in T-cell epitope formation and at the same time a target for autoantibodies. Understanding this connection is one of the major challenges for obtaining a complete understanding of how gluten causes tissue destruction and remodeling of the mucosa in the small bowel.


Assuntos
Imunidade Adaptativa , Doença Celíaca/imunologia , Doença Celíaca/patologia , Intestino Delgado/imunologia , Intestino Delgado/patologia , Epitopos de Linfócito T/imunologia , Proteínas de Ligação ao GTP/imunologia , Humanos , Imunoglobulina A/imunologia , Proteína 2 Glutamina gama-Glutamiltransferase , Transglutaminases/imunologia
12.
J Immunol ; 190(12): 5981-91, 2013 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-23690478

RESUMO

The gluten-sensitive enteropathy celiac disease is tightly associated with the production of autoantibodies specific for the enzyme transglutaminase 2 (TG2). The mechanisms underlying the activation of autoreactive B cells, however, are not well defined. To gain more insight into this autoimmune response we have characterized the binding of TG2 by a panel of human mAbs generated by expression cloning of Ig genes from single plasma cells of the celiac disease lesion. The Abs were highly specific to TG2 and bound preferentially to the open, Ca(2+)-activated enzyme conformation. Epitope mapping revealed that they recognize few distinct conformational epitopes that cluster in the N-terminal half of the enzyme. Two of the epitopes were overlapping with the fibronectin binding site in TG2, and none of the epitopes was accessible when TG2 was in a cell surface-bound form. Based on our findings, we propose that the autoantibodies are generated against the soluble, catalytically active enzyme, whereas Abs reactive with cell surface-associated TG2 are absent from the response due to negative selection of B cells recognizing membrane-bound self-Ag. The findings give insight into the mechanisms controlling the formation of anti-TG2 autoantibodies in celiac disease.


Assuntos
Autoanticorpos/imunologia , Doença Celíaca/imunologia , Epitopos de Linfócito B/imunologia , Transglutaminases/imunologia , Animais , Especificidade de Anticorpos , Autoantígenos/química , Autoantígenos/imunologia , Autoimunidade/imunologia , Ensaio de Imunoadsorção Enzimática , Mapeamento de Epitopos , Epitopos de Linfócito B/química , Citometria de Fluxo , Imunofluorescência , Proteínas de Ligação ao GTP , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 2 Glutamina gama-Glutamiltransferase , Transglutaminases/química
13.
PLoS One ; 18(6): e0287662, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37368893

RESUMO

BACKGROUND: Formation of complexes between transglutaminase 2 (TG2) and gluten can mechanistically explain why TG2 serves both as B-cell autoantigen and as an enzyme that creates deamidated gluten epitopes in coeliac disease (CeD). A model has been proposed where TG2 released from shed epithelial cells encounters high concentrations of dietary gluten peptides to form these TG2:gluten complexes. In this work we have characterised TG2 protein expression in gut epithelial cells in humans. METHODS: Western blot analysis, immunofluorescence staining and mass spectrometry in combination with laser capture microdissection to gain spatial resolution were used to characterise TG2 expression in the epithelial cell layer of healthy and coeliac disease affected duodenum. FINDINGS: TG2 is expressed in human duodenal epithelial cells, including cells in the apical region that are shed into the gut lumen. In untreated CeD the apical expression of TG2 is doubled. Enzymatically active TG2 is readily released from isolated human intestinal epithelial cells. CONCLUSION: Shed epithelial cells are a plausible source of pathogenic TG2 enzyme in CeD. Increased epithelial TG2 expression and increased epithelial shedding in active CeD may reinforce action of luminal TG2 in this condition.


Assuntos
Doença Celíaca , Proteína 2 Glutamina gama-Glutamiltransferase , Humanos , Autoanticorpos , Células Epiteliais/metabolismo , Glutens/metabolismo , Transglutaminases/metabolismo
14.
Nat Commun ; 14(1): 6216, 2023 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-37798283

RESUMO

Transglutaminase 3 (TG3), the autoantigen of dermatitis herpetiformis (DH), is a calcium dependent enzyme that targets glutamine residues in polypeptides for either transamidation or deamidation modifications. To become catalytically active TG3 requires proteolytic cleavage between the core domain and two C-terminal ß-barrels (C1C2). Here, we report four X-ray crystal structures representing inactive and active conformations of human TG3 in complex with a TG3-specific Fab fragment of a DH patient derived antibody. We demonstrate that cleaved TG3, upon binding of a substrate-mimicking inhibitor, undergoes a large conformational change as a ß-sheet in the catalytic core domain moves and C1C2 detaches. The unique enzyme-substrate conformation of TG3 without C1C2 is recognized by DH autoantibodies. The findings support a model where B-cell receptors of TG3-specific B cells bind and internalize TG3-gluten enzyme-substrate complexes thereby facilitating gluten-antigen presentation, T-cell help and autoantibody production.


Assuntos
Doença Celíaca , Dermatite Herpetiforme , Humanos , Autoanticorpos , Transglutaminases , Imunoglobulina A/metabolismo , Glutens
15.
Adv Sci (Weinh) ; 10(25): e2300401, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37424036

RESUMO

Dermatitis herpetiformis (DH) is an inflammatory skin disorder often considered as an extra intestinal manifestation of celiac disease (CeD). Hallmarks of CeD and DH are auto-antibodies to transglutaminase 2 (TG2) and transglutaminase 3 (TG3), respectively. DH patients have auto-antibodies reactive with both transglutaminase enzymes. Here it is reported that in DH both gut plasma cells and serum auto-antibodies are specific for either TG2 or TG3 with no TG2-TG3 cross reactivity. By generating monoclonal antibodies from TG3-specific duodenal plasma cells of DH patients, three conformational epitope groups are defined. Both TG2-specific and TG3-specific gut plasma cells have few immunoglobulin (Ig) mutations, and the two transglutaminase-reactive populations show distinct selection of certain heavy and light chain V-genes. Mass spectrometry analysis of TG3-specific serum IgA corroborates preferential usage of IGHV2-5 in combination with IGKV4-1. Collectively, these results demonstrate parallel induction of anti-TG2 and anti-TG3 auto-antibody responses involving separate B-cell populations in DH patients.


Assuntos
Doença Celíaca , Dermatite Herpetiforme , Humanos , Imunoglobulina A , Plasmócitos , Proteína 2 Glutamina gama-Glutamiltransferase , Transglutaminases
16.
J Biol Chem ; 286(43): 37866-73, 2011 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-21908620

RESUMO

The mechanism of activation of transglutaminase 2 (TG2) in the extracellular matrix remains a fundamental mystery in our understanding of the biology of this multifunctional mammalian enzyme. Earlier investigations have highlighted the role of a disulfide bond formed by vicinal Cys residues in maintaining calcium-bound TG2 in an inactive state. Here, we have shown that the redox potential of this disulfide bond is approximately -190 mV, a high value for a disulfide bond in proteins. Consistent with this observation, TG2 activity in a freshly wounded fibroblast culture depends upon the redox potential of the environment. We sought to identify a physiological mechanism for the activation of oxidized TG2. With a k(cat)/K(m) of 1.6 µm(-1) min(-1), human thioredoxin (Trx) was a highly specific activator of oxidized human TG2. Trx-mediated activation of TG2 was blocked by PX-12, a small molecule Trx inhibitor that is undergoing clinical trials as a cancer chemotherapeutic agent. In a mixed culture containing fibroblasts and monocytic cells, interferon-γ stimulated Trx release from monocytes, which in turn activated TG2 around the fibroblasts. Recombinant human Trx could also activate extracellular TG2 in cryosections of human and mouse small intestinal biopsies. In addition to explaining how TG2 can be activated by dietary gluten in the small intestinal mucosa of celiac sprue patients, our findings reveal a new strategy for inhibiting the undesirable consequences of TG2 activity in this widespread, lifelong disease.


Assuntos
Fibroblastos/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Monócitos/metabolismo , Tiorredoxinas/metabolismo , Transglutaminases/metabolismo , Animais , Antivirais/farmacologia , Biópsia , Doença Celíaca/metabolismo , Doença Celíaca/patologia , Linhagem Celular , Dissulfetos/farmacologia , Ativação Enzimática/efeitos dos fármacos , Fibroblastos/patologia , Proteínas de Ligação ao GTP/genética , Humanos , Imidazóis/farmacologia , Interferon gama/farmacologia , Intestino Delgado/metabolismo , Intestino Delgado/patologia , Camundongos , Camundongos Knockout , Monócitos/patologia , Oxirredução/efeitos dos fármacos , Proteína 2 Glutamina gama-Glutamiltransferase , Tiorredoxinas/genética , Transglutaminases/genética
17.
PLoS One ; 17(4): e0266543, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35385534

RESUMO

BACKGROUND: Celiac disease is an autoimmune enteropathy driven by dietary intake of gluten proteins. Typical histopathologic features are villous flattening, crypt hyperplasia and infiltration of inflammatory cells in the intestinal epithelium and lamina propria. The disease is hallmarked by the gluten-dependent production of autoantibodies targeting the enzyme transglutaminase 2 (TG2). While these antibodies are specific and sensitive diagnostic markers of the disease, a role in the development of the enteropathy has never been established. METHODS: We addressed this question by injecting murine antibodies harboring the variable domains of a prototypic celiac anti-TG2 immunoglobulin into TG2-sufficient and TG2-deficient mice evaluating for celiac enteropathy. RESULTS: We found no histopathologic abnormalities nor clinical signs of disease related to the injection of anti-TG2 IgG or IgA. CONCLUSIONS: Our findings do not support a direct role for secreted anti-TG2 antibodies in the development of the celiac enteropathy.


Assuntos
Doença Celíaca , Transglutaminases , Animais , Autoanticorpos , Doença Celíaca/patologia , Proteínas de Ligação ao GTP/metabolismo , Glutens/metabolismo , Imunoglobulina A , Mucosa Intestinal/metabolismo , Camundongos , Proteína 2 Glutamina gama-Glutamiltransferase , Transglutaminases/metabolismo
18.
J Biol Chem ; 285(33): 25402-9, 2010 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-20547769

RESUMO

Transglutaminase 2 (TG2) in the extracellular matrix is largely inactive but is transiently activated upon certain types of inflammation and cell injury. The enzymatic activity of extracellular TG2 thus appears to be tightly regulated. As TG2 is known to be sensitive to changes in the redox environment, inactivation through oxidation presents a plausible mechanism. Using mass spectrometry, we have identified a redox-sensitive cysteine triad consisting of Cys(230), Cys(370), and Cys(371) that is involved in oxidative inactivation of TG2. Within this triad, Cys(370) was found to participate in disulfide bonds with both Cys(230) and its neighbor, Cys(371). Notably, Ca(2+) was found to protect against formation of these disulfide bonds. To investigate the role of each cysteine residue, we created alanine mutants and found that Cys(230) appears to promote oxidation and inactivation of TG2 by facilitating formation of Cys(370)-Cys(371) through formation of the Cys(230)-Cys(370) disulfide bond. Although vicinal disulfide pairs are found in several transglutaminase isoforms, Cys(230) is unique for TG2, suggesting that this residue acts as an isoform-specific redox sensor. Our findings suggest that oxidation is likely to influence the amount of active TG2 present in the extracellular environment.


Assuntos
Proteínas de Ligação ao GTP/química , Proteínas de Ligação ao GTP/metabolismo , Transglutaminases/química , Transglutaminases/metabolismo , Cisteína/química , Cisteína/metabolismo , Proteínas de Ligação ao GTP/genética , Humanos , Espectrometria de Massas , Oxirredução , Proteína 2 Glutamina gama-Glutamiltransferase , Estrutura Secundária de Proteína , Transglutaminases/genética
19.
PLoS One ; 16(11): e0259082, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34731200

RESUMO

A hallmark of celiac disease is the gluten-dependent production of antibodies specific for deamidated gluten peptides (DGP) and the enzyme transglutaminase 2 (TG2). Both types of antibodies are believed to result from B cells receiving help from gluten-specific CD4+ T cells and differentiating into antibody-producing plasma cells. We have here studied the collaboration between DGP- and TG2-specific B cells with gluten-specific CD4+ T cells using transgenic mice expressing celiac patient-derived T-cell and B-cell receptors, as well as between B-cell transfectants and patient-derived gluten-specific T-cell clones. We show that multivalent TG2-gluten complexes are efficient antigens for both TG2-specific and DGP-specific B cells and allow both types of B cells to receive help from gluten-specific T cells of many different specificities.


Assuntos
Doença Celíaca/genética , Glutens/genética , Proteína 2 Glutamina gama-Glutamiltransferase/genética , Receptores de Antígenos de Linfócitos B/genética , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linfócitos B/patologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Doença Celíaca/imunologia , Doença Celíaca/patologia , Gliadina/genética , Gliadina/imunologia , Glutens/imunologia , Humanos , Camundongos , Camundongos Transgênicos , Proteína 2 Glutamina gama-Glutamiltransferase/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/patologia
20.
Adv Sci (Weinh) ; 8(4): 2003526, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33643806

RESUMO

In celiac disease (CeD), gluten activates adaptive immune cells that cause damage to the small intestinal mucosa. Histological evaluation of intestinal biopsies allows for grading of disease severity. CeD can effectively be treated with a life-long gluten-free diet. Gluten challenge of treated CeD patients is used to confirm diagnosis and to test drug efficacy in clinical trials, but patients respond with different magnitudes to the same gluten challenge. In this study of 19 well-treated CeD patients, proteome analysis of total tissue or isolated epithelial cell compartment from formalin-fixed paraffin embedded biopsies collected before and after 14-day gluten challenge demonstrates that patients with strong mucosal response to challenge have signs of ongoing tissue inflammation already before challenge. This low-level tissue inflammation at baseline is paralleled by increased gluten specific CD4+ T-cell frequencies in the gut and presence of a low-level blood inflammatory profile. Thus, apparently well-treated CeD is frequently not entirely quiescent, with presence of low-grade inflammation and antigluten immunity in the gut mucosa. Histology assessment alone appears insufficient to judge full recovery and gut mucosal healing of CeD patients. The findings raise a concern whether a seemingly proper gluten-free diet is able to curb gut inflammation in all CeD patients.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA