Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.564
Filter
1.
Int J Mol Sci ; 25(11)2024 May 21.
Article in English | MEDLINE | ID: mdl-38891798

ABSTRACT

Systemic lupus erythematosus (SLE) is a multifactorial autoimmune disease characterized by self-immune tolerance breakdown and the production of autoantibodies, causing the deposition of immune complexes and triggering inflammation and immune-mediated damage. SLE pathogenesis involves genetic predisposition and a combination of environmental factors. Clinical manifestations are variable, making an early diagnosis challenging. Heat shock proteins (Hsps), belonging to the chaperone system, interact with the immune system, acting as pro-inflammatory factors, autoantigens, as well as immune tolerance promoters. Increased levels of some Hsps and the production of autoantibodies against them are correlated with SLE onset and progression. The production of these autoantibodies has been attributed to molecular mimicry, occurring upon viral and bacterial infections, since they are evolutionary highly conserved. Gut microbiota dysbiosis has been associated with the occurrence and severity of SLE. Numerous findings suggest that proteins and metabolites of commensal bacteria can mimic autoantigens, inducing autoimmunity, because of molecular mimicry. Here, we propose that shared epitopes between human Hsps and those of gut commensal bacteria cause the production of anti-Hsp autoantibodies that cross-react with human molecules, contributing to SLE pathogenesis. Thus, the involvement of the chaperone system, gut microbiota dysbiosis, and molecular mimicry in SLE ought to be coordinately studied.


Subject(s)
Dysbiosis , Gastrointestinal Microbiome , Lupus Erythematosus, Systemic , Molecular Mimicry , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/microbiology , Lupus Erythematosus, Systemic/metabolism , Humans , Molecular Mimicry/immunology , Dysbiosis/immunology , Gastrointestinal Microbiome/immunology , Molecular Chaperones/metabolism , Molecular Chaperones/immunology , Heat-Shock Proteins/immunology , Heat-Shock Proteins/metabolism , Autoantibodies/immunology , Animals , Autoantigens/immunology , Autoantigens/metabolism , Autoimmunity
2.
J Transl Med ; 22(1): 549, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38849852

ABSTRACT

Cellular communication (CC) influences tumor development by mediating intercellular junctions between cells. However, the role and underlying mechanisms of CC in malignant transformation remain unknown. Here, we investigated the spatiotemporal heterogeneity of CC molecular expression during malignant transformation. It was found that although both tight junctions (TJs) and gap junctions (GJs) were involved in maintaining the tumor microenvironment (TME), they exhibited opposite characteristics. Mechanistically, for epithelial cells (parenchymal component), the expression of TJ molecules consistently decreased during normal-cancer transformation and is a potential oncogenic factor. For fibroblasts (mesenchymal component), the expression of GJs consistently increased during normal-cancer transformation and is a potential oncogenic factor. In addition, the molecular profiles of TJs and GJs were used to stratify colorectal cancer (CRC) patients, where subtypes characterized by high GJ levels and low TJ levels exhibited enhanced mesenchymal signals. Importantly, we propose that leiomodin 1 (LMOD1) is biphasic, with features of both TJs and GJs. LMOD1 not only promotes the activation of cancer-associated fibroblasts (CAFs) but also inhibits the Epithelial-mesenchymal transition (EMT) program in cancer cells. In conclusion, these findings demonstrate the molecular heterogeneity of CC and provide new insights into further understanding of TME heterogeneity.


Subject(s)
Cancer-Associated Fibroblasts , Cell Communication , Colorectal Neoplasms , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Tumor Microenvironment , Animals , Humans , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Cell Line, Tumor , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/genetics , Epithelial-Mesenchymal Transition/genetics , Gap Junctions/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Spatio-Temporal Analysis , Tight Junctions/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Autoantigens/genetics , Autoantigens/metabolism
3.
Biochim Biophys Acta Mol Basis Dis ; 1870(6): 167228, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38734318

ABSTRACT

BACKGROUND: Early embryonic arrest and fragmentation (EEAF) is a common cause of female infertility, but the genetic causes remain to be largely unknown. CIP2A encodes the cellular inhibitor of PP2A, playing a crucial role in mitosis and mouse oocyte meiosis. METHODS: Exome sequencing and Sanger sequencing were performed to identify candidate causative genes in patients with EEAF. The pathogenicity of the CIP2A variant was assessed and confirmed in cultured cell lines and human oocytes through Western blotting, semi-quantitative RT-PCR, TUNEL staining, and fluorescence localization analysis. FINDINGS: We identified CIP2A (c.1510C > T, p.L504F) as a novel disease-causing gene in human EEAF from a consanguineous family. L504 is highly conserved throughout evolution. The CIP2A variant (c.1510C > T, p.L504F) reduced the expression level of the mutant CIP2A protein, leading to the abnormal aggregation of mutant CIP2A protein and cell apoptosis. Abnormal aggregation of CIP2A protein and chromosomal dispersion occurred in the patient's oocytes and early embryos. We further replicated the patient phenotype by knockdown CIP2A in human oocytes. Additionally, CIP2A deficiency resulted in decreased levels of phosphorylated ERK1/2. INTERPRETATION: We first found that the CIP2A loss-of-function variant associate with female infertility characterized by EEAF. Our findings suggest the uniqueness and importance of CIP2A gene in human oocyte and early embryo development. FUNDING: This work was supported by National Key Research and Development Program of China (2023YFC2706302), the National Natural Science Foundation of China (81000079, 81170165, and 81870959), the HUST Academic Frontier Youth Team (2016QYTD02), and the Key Research of Huazhong University of Science and Technology, Tongji Hospital (2022A20).


Subject(s)
Autoantigens , Infertility, Female , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Oocytes , Humans , Female , Autoantigens/genetics , Autoantigens/metabolism , Infertility, Female/genetics , Infertility, Female/pathology , Infertility, Female/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Oocytes/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Apoptosis/genetics , Loss of Function Mutation , Adult , Exome Sequencing , Animals , Pedigree , Mice
4.
Cell Signal ; 120: 111234, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38795810

ABSTRACT

Tumor dormancy is the underpinning for cancer relapse and chemoresistance, leading to massive cancer-related death in colorectal cancer (CRC). However, our comprehension of the mechanisms dictating tumor dormancy and strategies for eliminating dormant tumor cells remains restricted. In this study, we identified that collagen XVII (COL17A1), a hemidesmosomal transmembrane protein, can promote the dormancy of CRC cells. The upregulation of COL17A1 was observed to prolong quiescence periods and diminish drug susceptibility of CRC cells. Mechanistically, COL17A1 acts as a scaffold, enhancing the crosstalk between mTORC2 and Akt, thereby instigating the mTORC2-mediated dormant signaling. Notably, the activation of mTORC2 is contingent upon the intracellular domain of COL17A1, regardless of its ectodomain shedding. Our findings underscore a pivotal role of the COL17A1-mTORC2 axis in CRC dormancy, suggesting that mTORC2-specific inhibitors may hold therapeutic prospects for the eradication of dormant tumor cells.


Subject(s)
Collagen Type XVII , Colorectal Neoplasms , Mechanistic Target of Rapamycin Complex 2 , Non-Fibrillar Collagens , Signal Transduction , Humans , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/genetics , Mechanistic Target of Rapamycin Complex 2/metabolism , Non-Fibrillar Collagens/metabolism , Non-Fibrillar Collagens/genetics , Cell Line, Tumor , Animals , Proto-Oncogene Proteins c-akt/metabolism , Autoantigens/metabolism , Mice , Mice, Nude , Cell Proliferation , Mice, Inbred BALB C
5.
Biomol NMR Assign ; 18(1): 111-118, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38691336

ABSTRACT

Human La-related protein 1 (HsLARP1) is involved in post-transcriptional regulation of certain 5' terminal oligopyrimidine (5'TOP) mRNAs as well as other mRNAs and binds to both the 5'TOP motif and the 3'-poly(A) tail of certain mRNAs. HsLARP1 is heavily involved in cell proliferation, cell cycle defects, and cancer, where HsLARP1 is significantly upregulated in malignant cells and tissues. Like all LARPs, HsLARP1 contains a folded RNA binding domain, the La motif (LaM). Our current understanding of post-transcriptional regulation that emanates from the intricate molecular framework of HsLARP1 is currently limited to small snapshots, obfuscating our understanding of the full picture on HsLARP1 functionality in post-transcriptional events. Here, we present the nearly complete resonance assignment of the LaM of HsLARP1, providing a significant platform for future NMR spectroscopic studies.


Subject(s)
Amino Acid Motifs , Nuclear Magnetic Resonance, Biomolecular , Humans , Amino Acid Sequence , Autoantigens/chemistry , Autoantigens/metabolism , Nitrogen Isotopes , Ribonucleoproteins/chemistry , Ribonucleoproteins/metabolism , RNA-Binding Proteins
6.
PLoS One ; 19(5): e0287877, 2024.
Article in English | MEDLINE | ID: mdl-38787820

ABSTRACT

Type 1 diabetes (T1D) is characterized by HLA class I-mediated presentation of autoantigens on the surface of pancreatic ß-cells. Recognition of these autoantigens by CD8+ T cells results in the destruction of pancreatic ß-cells and, consequently, insulin deficiency. Most epitopes presented at the surface of ß-cells derive from the insulin precursor molecule proinsulin. The intracellular processing pathway(s) involved in the generation of these peptides are poorly defined. In this study, we show that a proinsulin B-chain antigen (PPIB5-14) originates from proinsulin molecules that are processed by ER-associated protein degradation (ERAD) and thus originate from ER-resident proteins. Furthermore, screening genes encoding for E2 ubiquitin conjugating enzymes, we identified UBE2G2 to be involved in proinsulin degradation and subsequent presentation of the PPIB10-18 autoantigen. These insights into the pathway involved in the generation of insulin-derived peptides emphasize the importance of proinsulin processing in the ER to T1D pathogenesis and identify novel targets for future T1D therapies.


Subject(s)
Autoantigens , Endoplasmic Reticulum-Associated Degradation , Proinsulin , Proteolysis , Ubiquitin-Conjugating Enzymes , Proinsulin/metabolism , Proinsulin/immunology , Proinsulin/genetics , Autoantigens/metabolism , Autoantigens/immunology , Humans , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Antigen Presentation/immunology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/immunology
7.
J Biol Chem ; 300(5): 107286, 2024 May.
Article in English | MEDLINE | ID: mdl-38636657

ABSTRACT

Hepatitis C virus (HCV) infection is tightly connected to the lipid metabolism with lipid droplets (LDs) serving as assembly sites for progeny virions. A previous LD proteome analysis identified annexin A3 (ANXA3) as an important HCV host factor that is enriched at LDs in infected cells and required for HCV morphogenesis. To further characterize ANXA3 function in HCV, we performed proximity labeling using ANXA3-BioID2 as bait in HCV-infected cells. Two of the top proteins identified proximal to ANXA3 during HCV infection were the La-related protein 1 (LARP1) and the ADP ribosylation factor-like protein 8B (ARL8B), both of which have been previously described to act in HCV particle production. In follow-up experiments, ARL8B functioned as a pro-viral HCV host factor without localizing to LDs and thus likely independent of ANXA3. In contrast, LARP1 interacts with HCV core protein in an RNA-dependent manner and is translocated to LDs by core protein. Knockdown of LARP1 decreased HCV spreading without altering HCV RNA replication or viral titers. Unexpectedly, entry of HCV particles and E1/E2-pseudotyped lentiviral particles was reduced by LARP1 depletion, whereas particle production was not altered. Using a recombinant vesicular stomatitis virus (VSV)ΔG entry assay, we showed that LARP1 depletion also decreased entry of VSV with VSV, MERS, and CHIKV glycoproteins. Therefore, our data expand the role of LARP1 as an HCV host factor that is most prominently involved in the early steps of infection, likely contributing to endocytosis of viral particles through the pleiotropic effect LARP1 has on the cellular translatome.


Subject(s)
Annexin A3 , Hepacivirus , Hepatitis C , SS-B Antigen , Virus Internalization , Humans , Annexin A3/metabolism , Annexin A3/genetics , Autoantigens/metabolism , Autoantigens/genetics , HEK293 Cells , Hepacivirus/metabolism , Hepacivirus/physiology , Hepatitis C/metabolism , Hepatitis C/virology , Hepatitis C/genetics , Host-Pathogen Interactions , Lipid Droplets/metabolism , Lipid Droplets/virology , Ribonucleoproteins/metabolism , Ribonucleoproteins/genetics , Viral Core Proteins/metabolism , Viral Core Proteins/genetics , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/genetics
8.
Sci Rep ; 14(1): 9571, 2024 04 26.
Article in English | MEDLINE | ID: mdl-38671086

ABSTRACT

Primary vitreoretinal lymphoma (PVRL) is a rare subtype of DLBCL and can progress into primary central nervous system lymphoma (PCNSL). To investigate the role of chronic antigenic stimulation in PVRL, we cloned and expressed B-cell receptors (BCR) from PVRL patients and tested for binding against human auto-antigens. SEL1L3, a protein with multiple glycosylation sites, was identified as the BCR target in 3/20 PVRL cases. SEL1L3 induces proliferation and BCR pathway activation in aggressive lymphoma cell lines. Moreover, SEL1L3 conjugated to a toxin killed exclusively lymphoma cells with respective BCR-reactivity. Western Blot analysis indicates the occurrence of hyper-N-glycosylation of SEL1L3 at aa 527 in PVRL patients with SEL1L3-reactive BCRs. The BCR of a PVRL patient with serum antibodies against SEL1L3 was cloned from a vitreous body biopsy at diagnosis and of a systemic manifestation at relapse. VH4-04*07 was used in both lymphoma manifestations with highly conserved CDR3 regions. Both BCRs showed binding to SEL1L3, suggesting continued dependence of lymphoma cells on antigen stimulation. These results indicate an important role of antigenic stimulation by post-translationally modified auto-antigens in the genesis of PVRL. They also provide the basis for a new treatment approach targeting unique lymphoma BCRs with ultimate specificity.


Subject(s)
Receptors, Antigen, B-Cell , Humans , Receptors, Antigen, B-Cell/metabolism , Glycosylation , Cell Line, Tumor , Retinal Neoplasms/genetics , Retinal Neoplasms/metabolism , Retinal Neoplasms/pathology , Retinal Neoplasms/immunology , Autoantigens/immunology , Autoantigens/metabolism , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/immunology , Lymphoma, Large B-Cell, Diffuse/pathology , Lymphoma, Large B-Cell, Diffuse/metabolism , Female , Male , Vitreous Body/metabolism , Vitreous Body/pathology , Middle Aged , Aged
9.
Cell Mol Biol (Noisy-le-grand) ; 70(4): 176-180, 2024 Apr 28.
Article in English | MEDLINE | ID: mdl-38678610

ABSTRACT

Recently, the progression of gastric cancer (GC), as one of the most ordinary malignant tumors, has been reported to be associated with circular RNAs. This study aimed to identify the role of circular RNA_LARP4 in GC. We performed real-time quantitative polymerase chain reaction (RT-qPCR) in 46 paired GC patients and GC cell lines to detect the expression of circular RNA_LARP4. Moreover, the role of circular RNA_LARP4 in GC proliferation was identified through proliferation assay and colony formation assay, while the role of circular RNA_LARP4 in GC metastasis was measured through scratch wound assay and transwell assay. Furthermore, the potential targets of circular RNA_LARP4 were predicted through bioinformatics methods and further identified by western blot assay and RT-qPCR. Circular RNA_LARP4 expression was remarkably lower in GC tissues compared with that in adjacent samples. Besides, cell proliferation of GC was inhibited after overexpression of circular RNA_LARP4, while cell migration and invasion of GC was inhibited after overexpression of circular RNA_LARP4. Furthermore, Upstream frameshift 1 (UPF1) was predicted as the potential target of circular RNA_LARP4 and was upregulated via overexpression of circular RNA_LARP4 in GC. Circular RNA_LARP4 inhibits GC cell proliferation and metastasis via targeting UPF1 in vitro, which might provide a new tumor suppressor in GC development.


Subject(s)
Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , RNA, Circular , Stomach Neoplasms , Female , Humans , Male , Middle Aged , Autoantigens/genetics , Autoantigens/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Neoplasm Invasiveness/genetics , Neoplasm Metastasis , RNA/genetics , RNA/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism , SS-B Antigen , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Up-Regulation/genetics
10.
Nat Commun ; 15(1): 899, 2024 Feb 06.
Article in English | MEDLINE | ID: mdl-38321013

ABSTRACT

Antigen-specific regulatory T cells (Tregs) suppress pathogenic autoreactivity and are potential therapeutic candidates for autoimmune diseases such as systemic lupus erythematosus (SLE). Lupus nephritis is associated with autoreactivity to the Smith (Sm) autoantigen and the human leucocyte antigen (HLA)-DR15 haplotype; hence, we investigated the potential of Sm-specific Tregs (Sm-Tregs) to suppress disease. Here we identify a HLA-DR15 restricted immunodominant Sm T cell epitope using biophysical affinity binding assays, then identify high-affinity Sm-specific T cell receptors (TCRs) using high-throughput single-cell sequencing. Using lentiviral vectors, we transduce our lead Sm-specific TCR into Tregs derived from patients with SLE who are anti-Sm and HLA-DR15 positive. Compared with polyclonal mock-transduced Tregs, Sm-Tregs potently suppress Sm-specific pro-inflammatory responses in vitro and suppress disease progression in a humanized mouse model of lupus nephritis. These results show that Sm-Tregs are a promising therapy for SLE.


Subject(s)
Lupus Erythematosus, Systemic , Lupus Nephritis , Mice , Animals , Humans , T-Lymphocytes, Regulatory , Autoantigens/metabolism
11.
J Autoimmun ; 144: 103174, 2024 04.
Article in English | MEDLINE | ID: mdl-38377868

ABSTRACT

In many autoimmune diseases, autoantigen-specific Th17 cells play a pivotal role in disease pathogenesis. Th17 cells can transdifferentiate into other T cell subsets in inflammatory conditions, however, there have been no attempts to target Th17 cell plasticity using vaccines. We investigated if autoantigen-specific Th17 cells could be specifically targeted using a therapeutic vaccine approach, where antigen was formulated in all-trans retinoic acid (ATRA)-containing liposomes, permitting co-delivery of antigen and ATRA to the same target cell. Whilst ATRA was previously found to broadly reduce Th17 responses, we found that antigen formulated in ATRA-containing cationic liposomes only inhibited Th17 cells in an antigen-specific manner and not when combined with an irrelevant antigen. Furthermore, this approach shifted existing Th17 cells away from IL-17A expression and transcriptomic analysis of sorted Th17 lineage cells from IL-17 fate reporter mice revealed a shift of antigen-specific Th17 cells to exTh17 cells, expressing functional markers associated with T cell regulation and tolerance. In the experimental autoimmune encephalomyelitis (EAE) mouse model of MS, vaccination with myelin-specific (MOG) antigen in ATRA-containing liposomes reduced Th17 responses and alleviated disease. This highlights the potential of therapeutic vaccination for changing the phenotype of existing Th17 cells in the context of immune mediated diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Th17 Cells , Mice , Animals , Liposomes/metabolism , Tretinoin/pharmacology , Tretinoin/metabolism , Autoantigens/metabolism , Adjuvants, Immunologic , Immunization , Vaccination , Phenotype , Mice, Inbred C57BL , Th1 Cells
12.
J Transl Med ; 22(1): 183, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38378668

ABSTRACT

BACKGROUND: Myasthenia gravis (MG) and the experimental autoimmune MG (EAMG) animal model are characterized by T-cell-induced and B-cell-dominated autoimmune diseases that affect the neuromuscular junction. Several subtypes of CD4+ T cells, including T helper (Th) 17 cells, follicular Th cells, and regulatory T cells (Tregs), contribute to the pathogenesis of MG. However, increasing evidence suggests that CD8+ T cells also play a critical role in the pathogenesis and treatment of MG. MAIN BODY: Herein, we review the literature on CD8+ T cells in MG, focusing on their potential effector and regulatory roles, as well as on relevant evidence (peripheral, in situ, cerebrospinal fluid, and under different treatments), T-cell receptor usage, cytokine and chemokine expression, cell marker expression, and Treg, Tc17, CD3+CD8+CD20+ T, and CXCR5+ CD8+ T cells. CONCLUSIONS: Further studies on CD8+ T cells in MG are necessary to determine, among others, the real pattern of the Vß gene usage of autoantigen-specific CD8+ cells in patients with MG, real images of the physiology and function of autoantigen-specific CD8+ cells from MG/EAMG, and the subset of autoantigen-specific CD8+ cells (Tc1, Tc17, and IL-17+IFN-γ+CD8+ T cells). There are many reports of CD20-expressing T (or CD20 + T) and CXCR5+ CD8 T cells on autoimmune diseases, especially on multiple sclerosis and rheumatoid arthritis. Unfortunately, up to now, there has been no report on these T cells on MG, which might be a good direction for future studies.


Subject(s)
CD8-Positive T-Lymphocytes , Myasthenia Gravis, Autoimmune, Experimental , Animals , Humans , T-Lymphocytes, Helper-Inducer/metabolism , Myasthenia Gravis, Autoimmune, Experimental/metabolism , T-Lymphocytes, Regulatory , Autoantigens/metabolism
13.
Cell Res ; 34(4): 295-308, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38326478

ABSTRACT

Autoreactive B cells are silenced through receptor editing, clonal deletion and anergy induction. Additional autoreactive B cells are ignorant because of physical segregation from their cognate autoantigen. Unexpectedly, we find that follicular B cell-derived autoantigen, including cell surface molecules such as FcγRIIB, is a class of homeostatic autoantigen that can induce spontaneous germinal centers (GCs) and B cell-reactive autoantibodies in non-autoimmune animals with intact T and B cell repertoires. These B cell-reactive B cells form GCs in a manner dependent on spontaneous follicular helper T (TFH) cells, which preferentially recognize B cell-derived autoantigen, and in a manner constrained by spontaneous follicular regulatory T (TFR) cells, which also carry specificities for B cell-derived autoantigen. B cell-reactive GC cells are continuously generated and, following immunization or infection, become intermixed with foreign antigen-induced GCs. Production of plasma cells and antibodies derived from B cell-reactive GC cells are markedly enhanced by viral infection, potentially increasing the chance for autoimmunity. Consequently, immune homeostasis in healthy animals not only involves classical tolerance of silencing and ignoring autoreactive B cells but also entails a reactive equilibrium attained by a spontaneous B cell-reactive triad of B cells, TFH cells and TFR cells.


Subject(s)
T-Lymphocytes, Helper-Inducer , T-Lymphocytes, Regulatory , Animals , T-Lymphocytes, Helper-Inducer/metabolism , B-Lymphocytes , Germinal Center/metabolism , Autoantigens/metabolism
14.
Arthritis Rheumatol ; 76(1): 92-99, 2024 01.
Article in English | MEDLINE | ID: mdl-37530745

ABSTRACT

OBJECTIVE: Autoantibodies are clinically useful in phenotyping patients with systemic sclerosis (SSc). Gastrointestinal (GI) function is regulated by the enteric nervous system (ENS) and commonly impaired in SSc, suggesting that the SSc autoimmune response may target ENS antigens. We sought to identify novel anti-ENS autoantibodies with an aim to clinically phenotype SSc GI dysfunction. METHODS: Serum from a patient with SSc with GI dysfunction but without defined SSc-associated autoantibodies was used for autoantibody discovery. Immunoprecipitations performed with murine myenteric plexus lysates were on-bead digested, and autoantigens were identified by mass spectrometry. Prevalence was determined, and clinical features associated with novel autoantibodies were evaluated in a SSc cohort using regression analyses. The expression of gephyrin in human GI tract tissue was examined by immunohistochemistry. RESULTS: We identified gephyrin as a novel SSc autoantigen. Anti-gephyrin antibodies were present in 9% of patients with SSc (16/188) and absent in healthy controls (0/46). Anti-gephyrin antibody-positive patients had higher constipation scores (1.00 vs 0.50, P = 0.02) and were more likely to have severe constipation and severe distention/bloating (46% vs 15%, P = 0.005; 54% vs 25%, P = 0.023, respectively). Anti-gephyrin antibody levels were significantly higher among patients with severe constipation (0.04 vs 0.00; P = 0.001) and severe distention and bloating (0.03 vs 0.004; P = 0.010). Severe constipation was associated with anti-gephyrin antibodies even in the adjusted model. Importantly, gephyrin was expressed in the ENS, which regulates gut motility. CONCLUSION: Gephyrin is a novel ENS autoantigen that is expressed in human myenteric ganglia. Anti-gephyrin autoantibodies are associated with the presence and severity of constipation in patients with SSc.


Subject(s)
Autoantibodies , Membrane Proteins , Scleroderma, Systemic , Membrane Proteins/metabolism , Autoantigens/metabolism , Scleroderma, Systemic/immunology , Scleroderma, Systemic/metabolism , Scleroderma, Systemic/pathology , Scleroderma, Systemic/physiopathology , Autoantibodies/analysis , Gastrointestinal Tract/innervation , Gastrointestinal Tract/physiopathology , Humans , Animals , Mice , Neurons/metabolism , Enteric Nervous System/metabolism , Enteric Nervous System/physiopathology
15.
Semin Arthritis Rheum ; 64S: 152315, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38008707

ABSTRACT

BACKGROUND: The generation and persistence of autoreactive B and plasma cells is crucial to the pathogenesis of many human autoimmune diseases. Secreted autoantibodies frequently serve as biomarkers in clinical practice and, in some cases, function as pathogenic effector molecules. Nonetheless, the primary break of B cell tolerance against autoantigens, the triggers that maintain autoreactive B cell memory, and the phenotype that autoreactive B cells adopt during the disease course are poorly understood. OBJECTIVES: To study phenotype and functional characteristics of human autoreactive B cells in the course of human disease using rheumatoid arthritis and the B cell response against posttranslationally modified antigens as prototype. METHODS: Combinatorial, antigen-specific identification and multiparameter phenotyping of autoreactive B cells by conventional and spectral flow cytometry in cohorts with well-defined clinical phenotypes, including patients in the phase preceding disease and in those reaching long-term, drugfree remission. RESULTS: Autoreactive B cells against post-translationally modified proteins operate as remarkably activated effector memory cells in patients with established disease and maintain this state throughout the disease course. The activation generates cytokine-secreting germinal center emigrants that resist conventional therapy, and migratory plasmablasts expressing homing markers that can direct the cells to sites of inflammation. In the pre-clinical at-risk phase, the degree of activation is lower, and migratory plasmablasts are less frequent. The cells are cross-reactive to different posttranslational modifications and express B cell receptors that are extensively glycosylated in the variable domain. CONCLUSIONS: Immune phenotyping of disease-specific, autoreactive B cells reveals heterogeneous features of human autoimmunity that reflect disease stage and course and that are only revealed upon antigen-specific cellular analysis. In rheumatoid arthritis, the picture of germinal center-derived B cell autoreactivity against post-translationally modified antigens emerges that displays extensive cross-reactivity and a likely dependence on T cell help. Such features may be different for other human autoimmune diseases with different disease kinetics, which each may require different strategies for (autoreactive) B cell targeting.


Subject(s)
Arthritis, Rheumatoid , Autoimmune Diseases , Humans , Autoimmunity , Autoantibodies , Autoantigens/metabolism , Disease Progression
16.
Curr Opin Biotechnol ; 85: 103056, 2024 02.
Article in English | MEDLINE | ID: mdl-38141322

ABSTRACT

Autoantibodies (AAb) are an immunological resource ripe for exploitation in cancer detection and treatment. Key to this translation is a better understanding of the self-epitope that AAb target in tumor tissue, but do not bind to in normal tissue. Posttranslational modifications (PTMs) on self-proteins are known to break tolerance in many autoimmune diseases and have also recently been described in cancer. This scope of possible autoantigens is quite broad and new high-dimensional and -throughput technologies to probe this repertoire will be necessary to fully exploit their potential. Here, we discuss the strengths and weaknesses of existing high-throughput platforms to detect AAb, review the current methods for characterizing immunogenic PTMs, describe the main challenges to identifying disease-relevant antigens and suggest the properties of future technologies that may be able to address these challenges. We conclude that exploiting the evolutionary power of the immune system to distinguish between self and nonself has great potential to be translated into antibody-based clinical applications.


Subject(s)
Autoimmune Diseases , Neoplasms , Humans , Autoantibodies/metabolism , Autoantigens/metabolism , Proteins/metabolism , Protein Processing, Post-Translational
17.
Nucleic Acids Res ; 52(5): 2273-2289, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38118002

ABSTRACT

Albeit N1-Methyladenosine (m1A) RNA modification represents an important regulator of RNA metabolism, the role of m1A modification in carcinogenesis remains enigmatic. Herein, we found that histone lactylation enhances ALKBH3 expression and simultaneously attenuates the formation of tumor-suppressive promyelocytic leukemia protein (PML) condensates by removing the m1A methylation of SP100A, promoting the malignant transformation of cancers. First, ALKBH3 is specifically upregulated in high-risk ocular melanoma due to excessive histone lactylation levels, referring to m1A hypomethylation status. Moreover, the multiomics analysis subsequently identified that SP100A, a core component for PML bodies, serves as a downstream candidate target for ALKBH3. Therapeutically, the silencing of ALKBH3 exhibits efficient therapeutic efficacy in melanoma both in vitro and in vivo, which could be reversed by the depletion of SP100A. Mechanistically, we found that YTHDF1 is responsible for recognition of the m1A methylated SP100A transcript, which increases its RNA stability and translational efficacy. Conclusively, we initially demonstrated that m1A modification is necessary for tumor suppressor gene expression, expanding the current understandings of dynamic m1A function during tumor progression. In addition, our results indicate that lactylation-driven ALKBH3 is essential for the formation of PML nuclear condensates, which bridges our knowledge of m1A modification, metabolic reprogramming, and phase-separation events.


Subject(s)
AlkB Homolog 3, Alpha-Ketoglutarate-Dependent Dioxygenase , Antigens, Nuclear , Autoantigens , Eye Neoplasms , Histones , Melanoma , Humans , AlkB Homolog 3, Alpha-Ketoglutarate-Dependent Dioxygenase/metabolism , Demethylation , DNA Methylation , Histones/genetics , Histones/metabolism , Promyelocytic Leukemia Protein/genetics , Promyelocytic Leukemia Protein/metabolism , RNA/metabolism , Transcription Factors/metabolism , Antigens, Nuclear/metabolism , Autoantigens/metabolism , Eye Neoplasms/metabolism
18.
Front Immunol ; 14: 1242860, 2023.
Article in English | MEDLINE | ID: mdl-38094290

ABSTRACT

The regulatory T (Treg) cells constitute a functionally defined subpopulation of T cells that modulate the immune system and maintain immune tolerance through suppression of the development of autoimmune responses to self-antigens and allergic reactions to external antigens. Reduction in the number or function of Treg cells has been suggested as a key immune abnormality underlying the development of autoimmune and allergic diseases. In vitro studies have demonstrated that purified polyvalent immunoglobulin G (IgG) from multiple healthy blood donors can exert immunomodulatory effects on Treg cells. Incubation of polyvalent human IgG with purified CD4+CD25high T cells increased the intracellular expression of interleukin (IL)-10. Intravenous administration of polyvalent human IgG induced significant expansions of CD4+ Foxp3+ Treg cells and clinical improvements in patients with autoimmune diseases. In human clinical trials, intramuscular administration of autologous total IgG significantly increased the percentage of IL-10-producing CD4+ Treg cells in the peripheral blood of healthy subjects and provided significant clinical improvements in patients with atopic dermatitis. These results suggest a clinical usefulness of polyvalent IgG-induced activation of Treg cells in human subjects. This review proposes a new hypothesis for immune tolerance mechanism by integrating the pre-existing "idiotypic network theory" and "Treg cell theory" into an "anti-idiotypic Treg cell theory." Based on this hypothesis, an "active anti-idiotypic therapy" for allergic and autoimmune diseases using autologous polyvalent IgG (as immunizing antigens) is suggested as follows: (1) Intramuscular or subcutaneous administration of autologous polyvalent IgG produces numerous immunogenic peptides derived from idiotypes of autologous IgG through processing of dendritic cells, and these peptides activate anti-idiotypic Treg cells in the same subject. (2) Activated anti-idiotypic Treg cells secrete IL-10 and suppress Th2 cell response to allergens and autoimmune T cell response to self-antigens. (3) These events can induce a long-term clinical improvements in patients with allergic and autoimmune diseases. Further studies are needed to evaluate the detailed molecular mechanism underlying polyvalent IgG-induced Treg cell activation and the clinical usefulness of this immunomodulatory therapy for autoimmune and allergic diseases.


Subject(s)
Autoimmune Diseases , Hypersensitivity , Humans , T-Lymphocytes, Regulatory , Interleukin-10/metabolism , Immunoglobulin G/metabolism , Immune Tolerance , Allergens , Hypersensitivity/metabolism , Autoimmune Diseases/therapy , Autoimmune Diseases/metabolism , Autoantigens/metabolism
19.
Nat Commun ; 14(1): 8227, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-38086807

ABSTRACT

Centromeres are epigenetically defined via the presence of the histone H3 variant CENP-A. Contacting CENP-A nucleosomes, the constitutive centromere associated network (CCAN) and the kinetochore assemble, connecting the centromere to spindle microtubules during cell division. The DNA-binding centromeric protein CENP-B is involved in maintaining centromere stability and, together with CENP-A, shapes the centromeric chromatin state. The nanoscale organization of centromeric chromatin is not well understood. Here, we use single-molecule fluorescence and cryoelectron microscopy (cryoEM) to show that CENP-A incorporation establishes a dynamic and open chromatin state. The increased dynamics of CENP-A chromatin create an opening for CENP-B DNA access. In turn, bound CENP-B further opens the chromatin fiber structure and induces nucleosomal DNA unwrapping. Finally, removal of CENP-A increases CENP-B mobility in cells. Together, our studies show that the two centromere-specific proteins collaborate to reshape chromatin structure, enabling the binding of centromeric factors and establishing a centromeric chromatin state.


Subject(s)
Chromatin , Chromosomal Proteins, Non-Histone , Centromere Protein A/metabolism , Cryoelectron Microscopy , Chromosomal Proteins, Non-Histone/metabolism , Centromere/metabolism , Nucleosomes , DNA/metabolism , Autoantigens/metabolism
20.
Aging (Albany NY) ; 15(21): 12570-12587, 2023 11 09.
Article in English | MEDLINE | ID: mdl-37950733

ABSTRACT

BACKGROUND: Mycobacterium tuberculosis (Mtb) is the bacterial pathogen responsible for causing tuberculosis (TB), a severe public health concern that results in numerous deaths worldwide. Ubiquitination (Ub) is an essential physiological process that aids in maintaining homeostasis and contributes to the development of TB. Therefore, the main objective of our study was to investigate the potential role of Ub-related genes in TB. METHODS: Our research entailed utilizing single sample gene set enrichment analysis (ssGSEA) in combination with several machine learning techniques to discern the Ub-related signature of TB and identify potential diagnostic markers that distinguish TB from healthy controls (HC). RESULTS: In summary, we used the ssGSEA algorithm to determine the score of Ub families (E1, E2, E3, DUB, UBD, and ULD). Notably, the score of E1, E3, and UBD were lower in TB patients than in HC individuals, and we identified 96 Ub-related differentially expressed genes (UbDEGs). Employing machine learning algorithms, we identified 11 Ub-related hub genes and defined two distinct Ub-related subclusters. Notably, through GSVA and functional analysis, it was determined that these subclusters were implicated in numerous immune-related processes. We further investigated these Ub-related hub genes in four TB-related diseases and found that TRIM68 exhibited higher correlations with various immune cells in different conditions, indicating that it may play a crucial role in the immune process of these diseases. CONCLUSION: The observed enrichment of Ub-related gene expression in TB patients emphasizes the potential involvement of ubiquitination in the progression of TB. These significant findings establish a basis for future investigations to elucidate the molecular mechanisms associated with TB, select suitable diagnostic biomarkers, and design innovative therapeutic interventions for combating this fatal infectious disease.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Humans , Tuberculosis/genetics , Tuberculosis/microbiology , Ubiquitination , Algorithms , Tripartite Motif Proteins/genetics , Autoantigens/metabolism , Ubiquitin-Protein Ligases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...