Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.240
Filter
1.
PLoS One ; 19(5): e0298864, 2024.
Article in English | MEDLINE | ID: mdl-38753630

ABSTRACT

Fibrotic remodeling is the primary driver of functional loss in chronic kidney disease, with no specific anti-fibrotic agent available for clinical use. Transglutaminase 2 (TG2), a wound response enzyme that irreversibly crosslinks extracellular matrix proteins causing dysregulation of extracellular matrix turnover, is a well-characterized anti-fibrotic target in the kidney. We describe the humanization and characterization of two anti-TG2 monoclonal antibodies (zampilimab [hDC1/UCB7858] and BB7) that inhibit crosslinking by TG2 in human in vitro and rabbit/cynomolgus monkey in vivo models of chronic kidney disease. Determination of zampilimab half-maximal inhibitory concentration (IC50) against recombinant human TG2 was undertaken using the KxD assay and determination of dissociation constant (Kd) by surface plasmon resonance. Efficacy in vitro was established using a primary human renal epithelial cell model of tubulointerstitial fibrosis, to assess mature deposited extracellular matrix proteins. Proof of concept in vivo used a cynomolgus monkey unilateral ureteral obstruction model of chronic kidney disease. Zampilimab inhibited TG2 crosslinking transamidation activity with an IC50 of 0.25 nM and Kd of <50 pM. In cell culture, zampilimab inhibited extracellular TG2 activity (IC50 119 nM) and dramatically reduced transforming growth factor-ß1-driven accumulation of multiple extracellular matrix proteins including collagens I, III, IV, V, and fibronectin. Intravenous administration of BB7 in rabbits resulted in a 68% reduction in fibrotic index at Day 25 post-unilateral ureteral obstruction. Weekly intravenous administration of zampilimab in cynomolgus monkeys with unilateral ureteral obstruction reduced fibrosis at 4 weeks by >50%, with no safety signals. Our data support the clinical investigation of zampilimab for the treatment of kidney fibrosis.


Subject(s)
Disease Models, Animal , Fibrosis , GTP-Binding Proteins , Macaca fascicularis , Protein Glutamine gamma Glutamyltransferase 2 , Renal Insufficiency, Chronic , Transglutaminases , Animals , Humans , Fibrosis/drug therapy , Rabbits , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/pathology , Transglutaminases/antagonists & inhibitors , Transglutaminases/metabolism , GTP-Binding Proteins/antagonists & inhibitors , GTP-Binding Proteins/metabolism , GTP-Binding Proteins/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized/pharmacology , Male , Kidney/pathology , Kidney/drug effects , Kidney/metabolism
2.
BMJ Case Rep ; 17(5)2024 May 06.
Article in English | MEDLINE | ID: mdl-38719261

ABSTRACT

Olmesartan is an angiotensin II receptor blocker licensed for the treatment of hypertension. It can cause a sprue-like enteropathy (SLE), characterised by chronic diarrhoea, weight loss and villous atrophy. Transiently raised anti-tissue transglutaminase (ATTG) antibody has also been rarely reported in the literature.We describe the case of a woman in her mid-50s, who presented with a history of intermittent loose stools over 1 year, associated with significant weight loss. She had two marginally raised serum ATTG antibody tests during her work-up.After extensive investigations, she was diagnosed with olmesartan-induced enteropathy. On subsequent follow-up, her symptoms had resolved with cessation of her olmesartan therapy.This case adds to existing literature, highlighting the importance of considering olmesartan as a possible differential diagnosis for SLE. It also reports the presence of a raised ATTG antibody which is infrequently reported in this context.


Subject(s)
Diarrhea , Imidazoles , Tetrazoles , Transglutaminases , Weight Loss , Humans , Female , Imidazoles/adverse effects , Diarrhea/chemically induced , Tetrazoles/adverse effects , Middle Aged , Transglutaminases/immunology , Diagnosis, Differential , Angiotensin II Type 1 Receptor Blockers/adverse effects , Autoantibodies/blood , Protein Glutamine gamma Glutamyltransferase 2 , Chronic Disease , Celiac Disease/diagnosis , GTP-Binding Proteins/immunology , GTP-Binding Proteins/antagonists & inhibitors
3.
Clin Exp Med ; 24(1): 102, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38758367

ABSTRACT

Immunotherapy is widely used in cancer treatment; however, only a subset of patients responds well to it. Significant efforts have been made to identify patients who will benefit from immunotherapy. Successful anti-tumor immunity depends on an intact cancer-immunity cycle, especially long-lasting CD8+ T-cell responses. Interferon (IFN)-α/ß/IFN-γ/interleukin (IL)-15 pathways have been reported to be involved in the development of CD8+ T cells. And these pathways may predict responses to immunotherapy. Herein, we aimed to analyze multiple public databases to investigate whether IFN-α/ß/IFN-γ/IL-15 pathways could be used to predict the response to immunotherapy. Results showed that IFN-α/ß/IFN-γ/IL-15 pathways could efficiently predict immunotherapy response, and guanylate-binding protein 1 (GBP1) could represent the IFN-α/ß/IFN-γ/IL-15 pathways. In public and private cohorts, we further demonstrated that GBP1 could efficiently predict the response to immunotherapy. Functionally, GBP1 was mainly expressed in macrophages and strongly correlated with chemokines involved in T-cell migration. Therefore, our study comprehensively investigated the potential role of GBP1 in immunotherapy, which could serve as a novel biomarker for immunotherapy and a target for drug development.


Subject(s)
GTP-Binding Proteins , Immunotherapy , Interferon-alpha , Interferon-gamma , Interleukin-15 , Neoplasms , Humans , Interleukin-15/genetics , GTP-Binding Proteins/genetics , GTP-Binding Proteins/immunology , Interferon-gamma/metabolism , Neoplasms/immunology , Neoplasms/therapy , Immunotherapy/methods , Interferon-beta , CD8-Positive T-Lymphocytes/immunology , Signal Transduction
4.
Cell Rep ; 43(4): 114045, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38578826

ABSTRACT

Autoantibodies against the enzyme transglutaminase 2 (TG2) are characteristic of celiac disease (CeD), and TG2-specific immunoglobulin (Ig) A plasma cells are abundant in gut biopsies of patients. Here, we describe the corresponding population of autoreactive B cells in blood. Circulating TG2-specific IgA cells are present in untreated patients on a gluten-containing diet but not in controls. They are clonally related to TG2-specific small intestinal plasma cells, and they express gut-homing molecules, indicating that they are plasma cell precursors. Unlike other IgA-switched cells, the TG2-specific cells are negative for CD27, placing them in the double-negative (IgD-CD27-) category. They have a plasmablast or activated memory B cell phenotype, and they harbor fewer variable region mutations than other IgA cells. Based on their similarity to naive B cells, we propose that autoreactive IgA cells in CeD are generated mainly through chronic recruitment of naive B cells via an extrafollicular response involving gluten-specific CD4+ T cells.


Subject(s)
B-Lymphocytes , Celiac Disease , GTP-Binding Proteins , Immunoglobulin A , Plasma Cells , Protein Glutamine gamma Glutamyltransferase 2 , Transglutaminases , Celiac Disease/immunology , Celiac Disease/pathology , Humans , Transglutaminases/immunology , Transglutaminases/metabolism , Immunoglobulin A/immunology , Immunoglobulin A/metabolism , Immunoglobulin A/blood , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Plasma Cells/immunology , Plasma Cells/metabolism , GTP-Binding Proteins/immunology , GTP-Binding Proteins/metabolism , Autoantibodies/immunology , Autoantibodies/blood , Adult , Male , Female , Middle Aged , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism , Glutens/immunology
5.
J Pediatr Gastroenterol Nutr ; 78(5): 1143-1148, 2024 May.
Article in English | MEDLINE | ID: mdl-38477348

ABSTRACT

OBJECTIVES: Autoantibodies against tissue transglutaminase (tTG) are serological markers of celiac disease. The aim was to study the applicability of human leukocyte antigen (HLA)-genotyping and tTG autoantibodies in the screening of celiac disease in a longitudinal birth cohort followed to age 15 years. METHODS: Included were 13,860 HLA-DQ-genotyped children at birth and previously invited to a screening at age 3 and 9 years, respectively. HLA-DQB1*02 and/or DQB1*03:02 (HLA-risk) children were compared with non-HLA-DQB1*02 and non-DQB1*03:02 (HLA-nonrisk) children. The present study reinvited 12,948/13,860 (93.4%) children at age 15 years of whom 1056/2374 (44.5%) participated in screening at both age 3 and 9 years. Both immunoglobulin A (IgA) and G (IgG) autoantibodies against tTG were analyzed separately in radiobinding assays. Persistently tTG autoantibody-positive children were examined with intestinal biopsy to confirm the diagnosis of celiac disease. RESULTS: At age 3 years, celiac disease was diagnosed in 56/1635 (3.4%) HLA-risk children compared with 0/1824 HLA-nonrisk children (p < 0.001). By age 9 years, celiac disease was diagnosed in 72/1910 (3.8%) HLA-risk children compared with 0/2167 HLA-nonrisk children (p < 0.001). Screening at age 15 years detected 14/1071 (1.3%) HLA-risk children positive for IgA-tTG and/or IgG-tTG of whom 12/1071 (1.1%) remained persistently positive. Among those, 10/1071 (0.9%, 95% confidence interval: 0.4%-1.7%) HLA-risk children were diagnosed with celiac disease compared with 0/1303 HLA-nonrisk children (p < 0.001) and 5/491 (1.0%) were negative in screenings at both 3 and 9 years of age. CONCLUSIONS: Screening for celiac disease needs to be performed at multiple timepoints to detect all cases but can be restricted to children at HLA-risk.


Subject(s)
Autoantibodies , Celiac Disease , GTP-Binding Proteins , Immunoglobulin A , Transglutaminases , Humans , Celiac Disease/diagnosis , Celiac Disease/immunology , Celiac Disease/genetics , Child , Child, Preschool , Transglutaminases/immunology , Longitudinal Studies , Autoantibodies/blood , Adolescent , Female , Male , Immunoglobulin A/blood , GTP-Binding Proteins/immunology , Immunoglobulin G/blood , Protein Glutamine gamma Glutamyltransferase 2 , HLA-DQ Antigens/genetics , Mass Screening/methods , Genotype , HLA-DQ beta-Chains/genetics , Risk Factors , Genetic Predisposition to Disease
6.
J Neurol ; 271(5): 2596-2604, 2024 May.
Article in English | MEDLINE | ID: mdl-38337123

ABSTRACT

BACKGROUND: A prior small-scale single center study suggested an association between celiac disease (CD)-type immunity and refractory temporal lobe epilepsy (TLE) with hippocampal sclerosis (HS). The present study addresses this putative association in a large, well-characterized group of drug-resistant epilepsy (DRE) patients. These patients were grouped based on the spectrum of CD and gluten sensitivity-associated antibodies. METHODS: In this cross-sectional study, 253 consecutive adult epilepsy patients (135 females, 118 males; age 16-76 years) were categorized into three groups: (i) CD-positive group with either prior diagnosis of CD or CD-specific TG2/EmA antibodies, (ii) AGA-positive group with antigliadin antibodies (AGA) but without CD, and (iii) CD/AGA-negative group without any gluten sensitivity-associated antibodies or CD. Clinical and immunological findings were then compared among the groups. RESULTS: TLE with HS was more common in the CD-positive group compared to CD/AGA-negative group (31.8% versus 11.9%, P = 0.019). Autoimmune disorders were more common in the AGA-positive group than in the CD/AGA-negative group (P = 0.025). Considering HS lateralization; left lateralization was more common in CD-positive group compared to CD/AGA-negative group (71.4% versus 25%, P = 0.030). TG6 seropositivity did not differ among the groups (P > 0.05). CONCLUSIONS: This study provides further evidence linking TLE with HS and CD-type autoimmunity suggesting that CD-type immune response to gluten can be one potential mechanism as a disease modifier leading to DRE and HS. Understanding these immunological factors is imperative for developing immunomodulatory or dietary treatments for DRE potentially preventing HS progression.


Subject(s)
Celiac Disease , Drug Resistant Epilepsy , Epilepsy, Temporal Lobe , Hippocampus , Sclerosis , Humans , Female , Male , Adult , Middle Aged , Celiac Disease/complications , Celiac Disease/immunology , Epilepsy, Temporal Lobe/immunology , Epilepsy, Temporal Lobe/complications , Drug Resistant Epilepsy/immunology , Drug Resistant Epilepsy/etiology , Sclerosis/immunology , Young Adult , Adolescent , Cross-Sectional Studies , Aged , Hippocampus/pathology , Hippocampus/immunology , Autoantibodies/blood , Gliadin/immunology , Transglutaminases/immunology , GTP-Binding Proteins/immunology , Protein Glutamine gamma Glutamyltransferase 2 , Hippocampal Sclerosis
7.
JCI Insight ; 8(5)2023 03 08.
Article in English | MEDLINE | ID: mdl-36883568

ABSTRACT

WHIM syndrome is an inherited immune disorder caused by an autosomal dominant heterozygous mutation in CXCR4. The disease is characterized by neutropenia/leukopenia (secondary to retention of mature neutrophils in bone marrow), recurrent bacterial infections, treatment-refractory warts, and hypogammaglobulinemia. All mutations reported in WHIM patients lead to the truncations in the C-terminal domain of CXCR4, R334X being the most frequent. This defect prevents receptor internalization and enhances both calcium mobilization and ERK phosphorylation, resulting in increased chemotaxis in response to the unique ligand CXCL12. Here, we describe 3 patients presenting neutropenia and myelokathexis, but normal lymphocyte count and immunoglobulin levels, carrying what we believe to be a novel Leu317fsX3 mutation in CXCR4, leading to a complete truncation of its intracellular tail. The analysis of the L317fsX3 mutation in cells derived from patients and in vitro cellular models reveals unique signaling features in comparison with R334X mutation. The L317fsX3 mutation impairs CXCR4 downregulation and ß-arrestin recruitment in response to CXCL12 and reduces other signaling events - including ERK1/2 phosphorylation, calcium mobilization, and chemotaxis - all processes that are typically enhanced in cells carrying the R334X mutation. Our findings suggest that, overall, the L317fsX3 mutation may be causative of a form of WHIM syndrome not associated with an augmented CXCR4 response to CXCL12.


Subject(s)
GTP-Binding Proteins , Primary Immunodeficiency Diseases , beta-Arrestins , Humans , beta-Arrestin 1/genetics , beta-Arrestin 1/immunology , beta-Arrestins/genetics , beta-Arrestins/immunology , Calcium/metabolism , GTP-Binding Proteins/genetics , GTP-Binding Proteins/immunology , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/physiology , Mutation , Neutropenia/genetics , Neutropenia/immunology , Primary Immunodeficiency Diseases/genetics , Primary Immunodeficiency Diseases/immunology , Signal Transduction/genetics , Signal Transduction/physiology , Warts/genetics , Warts/immunology
8.
Yi Chuan ; 45(11): 1007-1017, 2023 Nov 20.
Article in English | MEDLINE | ID: mdl-38764266

ABSTRACT

Guanylate-binding proteins (GBPs) are a subfamily of interferon-inducible proteins that undertake distinct roles in the the context of bacteria, virus, chlamydia and parasites infections. These proteins exert a notable influence on the progression and outcomes of infectious diseases. Within the realm of host cell-autonomous immunity against pathogens, GBPs have been identified as the regulators of pyroptosis through canonical and noncanonical inflammasome activation pathways. In this review, we summarize the structure and evolution of GBP family members, the canonical and noncanonical inflammasome activation pathways, the roles of GBPs in regulating inflammasome activation, and the mechanisms of GBPs affecting infections induced by different pathogens. We hope to provide new basic research clues for the pathogenesis and diagnosis and treatment of infectious diseases.


Subject(s)
GTP-Binding Proteins , Inflammasomes , Inflammasomes/immunology , Humans , Animals , GTP-Binding Proteins/genetics , GTP-Binding Proteins/immunology , Communicable Diseases/immunology , Communicable Diseases/genetics
9.
BMC Immunol ; 23(1): 44, 2022 09 17.
Article in English | MEDLINE | ID: mdl-36115937

ABSTRACT

Lupus nephritis (LN) is a common and serious clinical manifestation of systemic lupus erythematosus. However, the pathogenesis of LN is not fully understood. The currently available treatments do not cure the disease and appear to have a variety of side effects in the long term. The purpose of this study was to search for key molecules involved in the LN immune response through bioinformatics techniques to provide a reference for LN-specific targeted therapy. The GSE112943 dataset was downloaded from the Gene Expression Omnibus database, and 20 of the samples were selected for analysis. In total, 2330 differentially expressed genes were screened. These genes were intersected with a list of immune genes obtained from the IMMPORT immune database to obtain 128 differentially expressed immune-related genes. Enrichment analysis showed that most of these genes were enriched in the interferon signalling pathway. Gene set enrichment analysis revealed that the sample was significantly enriched for expression of the interferon signalling pathway. Further analysis of the core gene cluster showed that nine genes, GBP2, VCAM1, ADAR, IFITM1, BST2, MX2, IRF5, OAS1 and TRIM22, were involved in the interferon signalling pathway. According to our analysis, the guanylate binding protein 2 (GBP2), interferon regulatory factor 5 and 2'-5'-oligoadenylate synthetase 1 (OAS1) genes are involved in three interferon signalling pathways. At present, we do not know whether GBP2 is associated with LN. Therefore, this study focused on the relationship between GBP2 and LN pathogenesis. We speculate that GBP2 may play a role in the pathogenesis of LN as a member of the interferon signalling pathway. Further immunohistochemical results showed that the expression of GBP2 was increased in the renal tissues of LN patients compared with the control group, confirming this conjecture. In conclusion, GBP2 is a member of the interferon signalling pathway that may have implications for the pathogenesis of LN and serves as a potential biomarker for LN.


Subject(s)
GTP-Binding Proteins , Interferons , Lupus Nephritis , Antiviral Agents , Biomarkers , GTP-Binding Proteins/genetics , GTP-Binding Proteins/immunology , Humans , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/immunology , Interferons/genetics , Interferons/immunology , Ligases , Lupus Nephritis/genetics , Lupus Nephritis/immunology , Signal Transduction/genetics , Signal Transduction/immunology
10.
Sci Rep ; 12(1): 11097, 2022 06 30.
Article in English | MEDLINE | ID: mdl-35773466

ABSTRACT

Tuberculosis (TB) is a global health problem of major concern. Identification of immune biomarkers may facilitate the early diagnosis and targeted treatment of TB. We used public RNA-sequencing datasets of patients with TB and healthy controls to identify differentially expressed genes and their associated functional networks. GBP1 expression was consistently significantly upregulated in TB, and 4492 differentially expressed genes were simultaneously associated with TB and high GBP1 expression. Weighted gene correlation analysis identified 12 functional modules. Modules positively correlated with TB and high GBP1 expression were associated with the innate immune response, neutrophil activation, neutrophil-mediated immunity, and NOD receptor signaling pathway. Eleven hub genes (GBP1, HLA-B, ELF4, HLA-E, IFITM2, TNFRSF14, CD274, AIM2, CFB, RHOG, and HORMAD1) were identified. The least absolute shrinkage and selection operator model based on hub genes accurately predicted the occurrence of TB (area under the receiver operating characteristic curve = 0.97). The GBP1-module-pathway network based on the STRING database showed that GBP1 expression correlated with the expression of interferon-stimulated genes (GBP5, BATF2, EPSTI1, RSAD2, IFI44L, IFIT3, and OAS3). Our study suggests GBP1 as an optimal diagnostic biomarker for TB, further indicating an association of the AIM2 inflammasome signaling pathway in TB pathology.


Subject(s)
Tuberculosis , Biomarkers , GTP-Binding Proteins/genetics , GTP-Binding Proteins/immunology , Gene Expression Profiling , Humans , Immunity, Innate/genetics , Membrane Proteins/metabolism , ROC Curve , Signal Transduction/genetics , Tuberculosis/diagnosis , Tuberculosis/genetics , Tuberculosis/immunology
11.
Cell Rep ; 37(8): 110040, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34818546

ABSTRACT

Tissue damage induces immediate-early signals, activating Rho small GTPases to trigger actin polymerization essential for later wound repair. However, how tissue damage is sensed to activate Rho small GTPases locally remains elusive. Here, we found that wounding the C. elegans epidermis induces rapid relocalization of CDC-42 into plasma membrane-associated clusters, which subsequently recruits WASP/WSP-1 to trigger actin polymerization to close the wound. In addition, wounding induces a local transient increase and subsequent reduction of H2O2, which negatively regulates the clustering of CDC-42 and wound closure. CDC-42 CAAX motif-mediated prenylation and polybasic region-mediated cation-phospholipid interaction are both required for its clustering. Cysteine residues participate in intermolecular disulfide bonds to reduce membrane association and are required for negative regulation of CDC-42 clustering by H2O2. Collectively, our findings suggest that H2O2-regulated fine-tuning of CDC-42 localization can create a distinct biomolecular cluster that facilitates rapid epithelial wound repair after injury.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Cell Cycle Proteins/metabolism , GTP-Binding Proteins/metabolism , Wound Healing/physiology , Actins , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/immunology , Cell Cycle Proteins/immunology , Cell Membrane/metabolism , Epidermal Cells/metabolism , Epidermis/metabolism , GTP-Binding Proteins/immunology , Hydrogen Peroxide/metabolism , Membrane Proteins/metabolism , Oxidation-Reduction , Polymerization , Signal Transduction , Wiskott-Aldrich Syndrome Protein Family/immunology , Wiskott-Aldrich Syndrome Protein Family/metabolism , Wound Healing/immunology , rho GTP-Binding Proteins/metabolism
12.
J Immunol ; 207(11): 2673-2680, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34732469

ABSTRACT

Stimulator of IFN genes (STING) is a key molecule that binds to cyclic dinucleotides produced by the cyclic GMP-AMP synthase to activate IFN expression and autophagy in the fight against microbial infection. The regulation of STING in the activation of IFN expression has been extensively reported, whereas the regulation of STING in the initiation of autophagy is still insufficiently determined. IFN-inducible guanylate-binding proteins (GBPs) are central to the cell-autonomous immunity in defending a host against viral, bacterial, and protozoan infections. In this study using the Chinese tree shrew (Tupaia belangeri chinensis), which is genetically close to primates, we found that Tupaia GBP1 (tGBP1) combines with Tupaia STING (tSTING), promotes autophagy, and moderately inhibits HSV type 1 (HSV-1) infection. The antiviral effects of tGBP1 are IFN independent. Mechanistically, tGBP1 interacted with tSTING, Tupaia sequestosome 1, and Tupaia microtubule associated protein 1 L chain 3, forming a complex which promotes autophagy in response to HSV-1 infection. This function of tGBP1 against HSV-1 infection was lost in tSTING knockout cells. Overexpression of either tSTING or its mutant tSTING-ΔCTT that can only activate autophagy rescued the anti-HSV-1 activity of tGBP1 in tSTING knockout cells. Our study not only elucidated the underlying mechanism of tGBP1 antiviral activity against HSV-1 infection, but also uncovered the regulation of tSTING in the initiation of autophagy in response to HSV-1 infection.


Subject(s)
Autophagy/immunology , GTP-Binding Proteins/immunology , Herpes Simplex/immunology , Herpesvirus 1, Human/immunology , Animals , HEK293 Cells , Humans , Tupaia
13.
Cells ; 10(7)2021 07 20.
Article in English | MEDLINE | ID: mdl-34360011

ABSTRACT

Transglutaminase 2 (TG2) is a ubiquitously expressed enzyme catalyzing the crosslinking between Gln and Lys residues and involved in various pathophysiological events. Besides this crosslinking activity, TG2 functions as a deamidase, GTPase, isopeptidase, adapter/scaffold, protein disulfide isomerase, and kinase. It also plays a role in the regulation of hypusination and serotonylation. Through these activities, TG2 is involved in cell growth, differentiation, cell death, inflammation, tissue repair, and fibrosis. Depending on the cell type and stimulus, TG2 changes its subcellular localization and biological activity, leading to cell death or survival. In normal unstressed cells, intracellular TG2 exhibits a GTP-bound closed conformation, exerting prosurvival functions. However, upon cell stimulation with Ca2+ or other factors, TG2 adopts a Ca2+-bound open conformation, demonstrating a transamidase activity involved in cell death or survival. These functional discrepancies of TG2 open form might be caused by its multifunctional nature, the existence of splicing variants, the cell type and stimulus, and the genetic backgrounds and variations of the mouse models used. TG2 is also involved in the phagocytosis of dead cells by macrophages and in fibrosis during tissue repair. Here, we summarize and discuss the multifunctional and controversial roles of TG2, focusing on cell death/survival and fibrosis.


Subject(s)
Aminoacyltransferases/genetics , Carbon-Nitrogen Lyases/genetics , Fibrosis/enzymology , GTP-Binding Proteins/genetics , Inflammation/enzymology , Protein Disulfide-Isomerases/genetics , Transglutaminases/genetics , Alternative Splicing , Aminoacyltransferases/immunology , Animals , Calcium/immunology , Calcium/metabolism , Carbon-Nitrogen Lyases/immunology , Cell Death , Cell Survival , Fibrosis/genetics , Fibrosis/immunology , Fibrosis/pathology , GTP-Binding Proteins/immunology , Gene Expression , Guanosine Triphosphate/immunology , Guanosine Triphosphate/metabolism , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Isoenzymes/genetics , Isoenzymes/immunology , Macrophages/enzymology , Macrophages/immunology , Phagocytosis/genetics , Protein Disulfide-Isomerases/immunology , Protein Glutamine gamma Glutamyltransferase 2 , Transglutaminases/immunology
14.
Front Immunol ; 12: 674313, 2021.
Article in English | MEDLINE | ID: mdl-34149709

ABSTRACT

Several environmental, genetic, and immune factors create a "perfect storm" for the development of coeliac disease: the antigen gluten, the strong association of coeliac disease with HLA, the deamidation of gluten peptides by the enzyme transglutaminase 2 (TG2) generating peptides that bind strongly to the predisposing HLA-DQ2 or HLA-DQ8 molecules, and the ensuing unrestrained T cell response. T cell immunity is at the center of the disease contributing to the inflammatory process through the loss of tolerance to gluten and the differentiation of HLA-DQ2 or HLA-DQ8-restricted anti-gluten inflammatory CD4+ T cells secreting pro-inflammatory cytokines and to the killing of intestinal epithelial cells by cytotoxic intraepithelial CD8+ lymphocytes. However, recent studies emphasize that the individual contribution of each of these cell subsets is not sufficient and that interactions between these different populations of T cells and the simultaneous activation of innate and adaptive immune pathways in distinct gut compartments are required to promote disease immunopathology. In this review, we will discuss how tissue destruction in the context of coeliac disease results from the complex interactions between gluten, HLA molecules, TG2, and multiple innate and adaptive immune components.


Subject(s)
Adaptive Immunity/immunology , Celiac Disease/immunology , Glutens/immunology , HLA Antigens/immunology , Immunity, Innate/immunology , Animals , Celiac Disease/pathology , GTP-Binding Proteins/immunology , Humans , Protein Glutamine gamma Glutamyltransferase 2 , Transglutaminases/immunology
15.
Nutrients ; 13(5)2021 May 11.
Article in English | MEDLINE | ID: mdl-34064541

ABSTRACT

An association between celiac disease and IgA nephropathy (IgAN) has been suggested. In celiac disease, in addition to circulating in serum, IgA-class tissue transglutaminase (tTG) autoantibodies are deposited in the small bowel mucosa and extraintestinal organs. In this case series of IgAN patients with or without celiac disease, we studied whether celiac disease-type IgA-tTG deposits occur in kidney biopsies. The study included nine IgAN patients, four of them with celiac disease. At the time of the diagnostic kidney biopsy serum tTG autoantibodies were measured and colocalization of IgA and tTG was investigated in the frozen kidney biopsies. Three IgAN patients with celiac disease had IgA-tTG deposits in the kidney even though in two of these the celiac disease diagnosis had been set years later. These deposits were not found in a patient with already diagnosed celiac disease following a gluten-free diet. Of the five non-celiac IgAN patients, three had IgA-tTG deposits in the kidney. We conclude that tTG-targeted IgA deposits can be found in the kidney biopsies of gluten-consuming IgAN patients but their specificity to celiac disease seems limited.


Subject(s)
Autoantibodies/blood , Biopsy , Celiac Disease/diagnosis , Celiac Disease/pathology , GTP-Binding Proteins/immunology , Glomerulonephritis, IGA/pathology , Kidney/pathology , Transglutaminases/immunology , Adult , Diet, Gluten-Free , Female , Glutens , Humans , Immunoglobulin A/blood , Intestinal Mucosa/pathology , Male , Middle Aged , Protein Glutamine gamma Glutamyltransferase 2 , Retrospective Studies , Young Adult
16.
Sci Rep ; 11(1): 11946, 2021 06 07.
Article in English | MEDLINE | ID: mdl-34099759

ABSTRACT

The detailed pathogenesis of eosinophilic bronchitis (EB) remains unclear. Transglutaminase 2 (TG2) has been implicated in many respiratory diseases including asthma. Herein, we aim to assess preliminarily the relationship of TG2 with EB in the context of the development of an appropriate EB model through ovalbumin (OVA) sensitization and challenge in the C57BL/6 mouse strain. Our data lead us to propose a 50 µg dose of OVA challenge as appropriate to establish an EB model in C57BL/6 mice, whereas a challenge with a 400 µg dose of OVA significantly induced asthma. Compared to controls, TG2 is up-regulated in the airway epithelium of EB mice and EB patients. When TG2 activity was inhibited by cystamine treatment, there were no effects on airway responsiveness; in contrast, the lung pathology score and eosinophil counts in bronchoalveolar lavage fluid were significantly increased whereas the cough frequency was significantly decreased. The expression levels of interleukin (IL)-4, IL-13, IL-6, mast cell protease7 and the transient receptor potential (TRP) ankyrin 1 (TRPA1), TRP vanilloid 1 (TRPV1) were significantly decreased. These data open the possibility of an involvement of TG2 in mediating the increased cough frequency in EB through the regulation of TRPA1 and TRPV1 expression. The establishment of an EB model in C57BL/6 mice opens the way for a genetic investigation of the involvement of TG2 and other molecules in this disease using KO mice, which are often generated in the C57BL/6 genetic background.


Subject(s)
Bronchitis/immunology , Disease Models, Animal , Eosinophils/immunology , GTP-Binding Proteins/immunology , Transglutaminases/immunology , Animals , Asthma/chemically induced , Asthma/immunology , Bronchitis/chemically induced , Bronchitis/metabolism , Cystamine/pharmacology , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Eosinophils/drug effects , Eosinophils/metabolism , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Gene Expression Regulation/drug effects , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Lung/drug effects , Lung/immunology , Lung/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Ovalbumin , Protein Glutamine gamma Glutamyltransferase 2 , TRPA1 Cation Channel/genetics , TRPA1 Cation Channel/immunology , TRPA1 Cation Channel/metabolism , Transglutaminases/genetics , Transglutaminases/metabolism
17.
J Immunol ; 206(10): 2420-2429, 2021 05 15.
Article in English | MEDLINE | ID: mdl-33941660

ABSTRACT

We have recently shown that type 2 transglutaminase (TG2) plays a key role in the host's inflammatory response during bacterial infections. In this study, we investigated whether the enzyme is involved in the regulation of the STING pathway, which is the main signaling activated in the presence of both self- and pathogen DNA in the cytoplasm, leading to type I IFN (IFN I) production. In this study, we demonstrated that TG2 negatively regulates STING signaling by impairing IRF3 phosphorylation in bone marrow-derived macrophages, isolated from wild-type and TG2 knockout mice. In the absence of TG2, we found an increase in the IFN-ß production and in the downstream JAK/STAT pathway activation. Interestingly, proteomic analysis revealed that TG2 interacts with TBK1, affecting its interactome composition. Indeed, TG2 ablation facilitates the TBK1-IRF3 interaction, thus indicating that the enzyme plays a negative regulatory effect on IRF3 recruitment in the STING/TBK1 complex. In keeping with these findings, we observed an increase in the IFNß production in bronchoalveolar lavage fluids from COVID-19-positive dead patients paralleled by a dramatic decrease of the TG2 expression in the lung pneumocytes. Taken together, these results suggest that TG2 plays a negative regulation on the IFN-ß production associated with the innate immunity response to the cytosolic presence of both self- and pathogen DNA.


Subject(s)
COVID-19/immunology , GTP-Binding Proteins/immunology , Immunity, Innate , Interferon Regulatory Factor-3/immunology , Membrane Proteins/immunology , Protein Serine-Threonine Kinases/immunology , SARS-CoV-2/immunology , Signal Transduction/immunology , Transglutaminases/immunology , Animals , COVID-19/genetics , COVID-19/pathology , GTP-Binding Proteins/genetics , Humans , Interferon Regulatory Factor-3/genetics , Interferon-beta/genetics , Interferon-beta/immunology , Membrane Proteins/genetics , Mice , Mice, Knockout , Protein Glutamine gamma Glutamyltransferase 2 , Protein Serine-Threonine Kinases/genetics , Signal Transduction/genetics , Transglutaminases/genetics
18.
Front Immunol ; 12: 645143, 2021.
Article in English | MEDLINE | ID: mdl-33959126

ABSTRACT

The reason why only few coeliac patients develop the cutaneous manifestation of the disease, named dermatitis herpetiformis (DH), is still unknown. Epidermal transglutaminase (TG3) has been described as the main autoantigen of humoral immunity in DH but the mechanisms leading to this autoimmune response remain obscure. Here we characterized T cells from skin, gut and peripheral blood of DH and coeliac disease (CD) patients, evaluated the impact of the gluten-free diet on circulating T lymphocytes' phenotype and investigated antigen specific T cell response toward epidermal and tissue transglutaminase (TG2). DH patients showed an increased frequency of skin-derived T cells producing TNFα when compared to CD patients. Moreover, circulating T cells producing TNFα and IL-17A positively correlated with clinical score of skin disease activity and decreased after gluten-free diet. Finally, TG2 and TG3-specific T cells resulted more reactive to antigens stimulation in DH patients and showed cross reactivity toward the two autoantigens in both the group of patients. Our data suggest a role of TNFα and IL-17A producing cells in the development of DH and, for the first time, show the existence of a crossed T cell response toward the two transglutaminases isoforms, thus suggesting new insights on T cells role in skin damage.


Subject(s)
Celiac Disease/immunology , Dermatitis Herpetiformis/immunology , GTP-Binding Proteins/immunology , T-Lymphocytes/immunology , Transglutaminases/immunology , Adolescent , Adult , Aged , Child , Female , Humans , Interleukin-17/immunology , Male , Middle Aged , Protein Glutamine gamma Glutamyltransferase 2 , Tumor Necrosis Factor-alpha/immunology
19.
J Clin Pathol ; 74(12): 766-773, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33789921

ABSTRACT

AIMS: Despite clinical evidence of liver involvement in patients with coeliac disease (CeD), there is a lack of a method to prove this association. METHODS: Of 146 treatment-naive patients with CeD, 26 had liver dysfunction. Liver biopsies and corresponding small intestinal biopsies were obtained from these 26 patients. Multicolour immunohistochemical and immunofluorescence confocal microscopic studies were performed on paraffin-embedded tissue to detect the IgA/anti-TG2 deposits. Follow-up liver biopsies were taken after a gluten-free diet. RESULTS: Twenty-six out of the 146 patients (17.8%) with suspected coeliac-associated liver disease on histological examination revealed irregular sinusoidal dilatation in 15 (57.6%), steatohepatitis in 4 (15.3%), non-specific chronic hepatitis in 3 (11.5%), autoimmune hepatitis in 2 (7.6%) biopsies, including cirrhosis in one of them, irregular perisinusoidal fibrosis and changes of non-cirrhotic portal fibrosis in one biopsy each (3.8%). IgA/anti-tTG deposits were observed in 22 (84.6%) liver biopsies by dual immunohistochemistry technique, and in 24 (92.3%) by confocal immunofluorescence technique and in all corresponding duodenal biopsies (100%). Overall, IgA/anti-tTG deposits showed 100% sensitivity, 77% specificity and 85% positive predictive value for establishing an association of extraintestinal pathology and CeD using archived tissues. Follow-up liver biopsies could be obtained in five patients; four of them showed not only resolution of the histological lesions but disappearance of IgA/anti-tTG co-localisation. CONCLUSIONS: Data of the present study adds to the body of evidence that liver lesions in patients with CeD are disease related and may have been caused by a similar pathogenic mechanism that causes intestinal changes.


Subject(s)
Autoantibodies/analysis , Celiac Disease/immunology , GTP-Binding Proteins/immunology , Immunohistochemistry , Intestinal Mucosa/immunology , Intestine, Small/immunology , Liver/immunology , Transglutaminases/immunology , Adolescent , Adult , Biopsy , Case-Control Studies , Celiac Disease/diagnosis , Celiac Disease/diet therapy , Diet, Gluten-Free , Female , Fluorescent Antibody Technique , Humans , Intestinal Mucosa/pathology , Intestine, Small/pathology , Liver/pathology , Male , Microscopy, Confocal , Predictive Value of Tests , Protein Glutamine gamma Glutamyltransferase 2 , Time Factors , Treatment Outcome , Young Adult
20.
Pathog Dis ; 79(5)2021 04 24.
Article in English | MEDLINE | ID: mdl-33885766

ABSTRACT

Many cytosolic bacterial pathogens hijack the host actin polymerization machinery to form actin tails that promote direct cell-to-cell spread, enabling these pathogens to avoid extracellular immune defenses. However, these pathogens are still susceptible to intracellular cell-autonomous immune responses that restrict bacterial actin-based motility. Two classes of cytosolic antimotility factors, septins and guanylate-binding proteins (GBPs), have recently been established to block actin tail formation by the human-adapted bacterial pathogen Shigella flexneri. Both septin cages and GBP1 microcapsules restrict S. flexneri cell-to-cell spread by blocking S. flexneri actin-based motility. While septins assemble into cage-like structures around immobile S. flexneri, GBP1 forms microcapsules around both motile and immobile bacteria. The interplay between these two defense programs remains elusive. Here, we demonstrate that GBP1 microcapsules block septin cage assembly, likely by interfering with the function of S. flexneri IcsA, the outer membrane protein that promotes actin-based motility, as this protein is required for septin cage formation. However, S. flexneri that escape from GBP1 microcapsules via the activity of IpaH9.8, a type III secreted effector that promotes the degradation of GBPs, are often captured within septin cages. Thus, our studies reveal how septin cages and GBP1 microcapsules represent complementary host cell antimotility strategies.


Subject(s)
Actins/metabolism , Bacterial Proteins/metabolism , DNA-Binding Proteins/metabolism , GTP-Binding Proteins , Septins/metabolism , Shigella flexneri , Transcription Factors/metabolism , GTP-Binding Proteins/genetics , GTP-Binding Proteins/immunology , GTP-Binding Proteins/metabolism , HeLa Cells , Host-Pathogen Interactions/immunology , Humans , Shigella flexneri/immunology , Shigella flexneri/metabolism , Shigella flexneri/pathogenicity
SELECTION OF CITATIONS
SEARCH DETAIL
...