Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.757
Filter
1.
Nat Commun ; 15(1): 6069, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39025846

ABSTRACT

Whole genome duplication is frequently observed in cancer, and its prevalence in our prior analysis of end-stage, homologous recombination deficient high grade serous ovarian cancer (almost 80% of samples) supports the notion that whole genome duplication provides a fitness advantage under the selection pressure of therapy. Here, we therefore aim to identify potential therapeutic vulnerabilities in primary high grade serous ovarian cancer with whole genome duplication by assessing differentially expressed genes and pathways in 79 samples. We observe that MHC-II expression is lowest in tumors which have acquired whole genome duplication early in tumor evolution, and further demonstrate that reduced MHC-II expression occurs in subsets of tumor cells rather than in canonical antigen-presenting cells. Early whole genome duplication is also associated with worse patient survival outcomes. Our results suggest an association between the timing of whole genome duplication, MHC-II expression and clinical outcome in high grade serous ovarian cancer that warrants further investigation for therapeutic targeting.


Subject(s)
Cystadenocarcinoma, Serous , Ovarian Neoplasms , Female , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Humans , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Gene Expression Regulation, Neoplastic , Gene Duplication , Genome, Human , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism
2.
Oncotarget ; 15: 507-520, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39028303

ABSTRACT

Macrophage Migration Inhibitory Factor (MIF) and its homolog D-dopachrome Tautomerase (DDT) have been implicated as drivers of tumor progression across a variety of cancers. Recent evidence suggests MIF as a therapeutic target in immune checkpoint inhibition (ICI) resistant melanomas, however clinical evidence of MIF and particularly of DDT remain limited. This retrospective study analyzed 97 patients treated at Yale for melanoma between 2002-2020. Bulk-RNA sequencing of patient tumor samples from the Skin Cancer SPORE Biorepository was used to evaluate for differential gene expression of MIF, DDT, CD74, and selected inflammatory markers, and gene expression was correlated with patient survival outcomes. Our findings revealed a strong correlation between MIF and DDT levels, with no statistically significant difference across common melanoma mutations and subtypes. Improved survival was associated with lower MIF and DDT levels and higher CD74:MIF and CD74:DDT levels. High CD74:DDT and CD74:MIF levels were also associated with enrichment of infiltrating inflammatory cell markers. These data suggest DDT as a novel target in immune therapy. Dual MIF and DDT blockade may provide synergistic responses in patients with melanoma, irrespective of common mutations, and may overcome ICI resistance. These markers may also provide prognostic value for further biomarker development.


Subject(s)
Antigens, Differentiation, B-Lymphocyte , Biomarkers, Tumor , Histocompatibility Antigens Class II , Intramolecular Oxidoreductases , Macrophage Migration-Inhibitory Factors , Melanoma , Humans , Macrophage Migration-Inhibitory Factors/metabolism , Macrophage Migration-Inhibitory Factors/genetics , Melanoma/drug therapy , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Melanoma/mortality , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Antigens, Differentiation, B-Lymphocyte/metabolism , Antigens, Differentiation, B-Lymphocyte/genetics , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Prognosis , Male , Female , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Middle Aged , Retrospective Studies , Aged , Adult , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Skin Neoplasms/metabolism , Skin Neoplasms/mortality , Mutation , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Aged, 80 and over
3.
Int J Mol Sci ; 25(13)2024 Jul 04.
Article in English | MEDLINE | ID: mdl-39000461

ABSTRACT

Antigen presentation is a crucial mechanism that drives the T cell-mediated immune response and the development of Multiple Sclerosis (MS). Genetic alterations within the highly variable Major Histocompatibility Complex Class II (MHC II) have been proven to result in significant changes in the molecular basis of antigen presentation and the clinical course of patients with both Adult-Onset MS (AOMS) and Pediatric-Onset MS (POMS). Among the numerous polymorphisms of the Human Leucocyte Antigens (HLA), within MHC II complex, HLA-DRB1*15:01 has been labeled, in Caucasian ethnic groups, as a high-risk allele for MS due to the ability of its structure to increase affinity to Myelin Basic Protein (MBP) epitopes. This characteristic, among others, in the context of the trimolecular complex or immunological synapsis, provides the foundation for autoimmunity triggered by environmental or endogenous factors. As with all professional antigen presenting cells, macrophages are characterized by the expression of MHC II and are often implicated in the formation of MS lesions. Increased presence of M1 macrophages in MS patients has been associated both with progression and onset of the disease, each involving separate but similar mechanisms. In this critical narrative review, we focus on macrophages, discussing how HLA genetic alterations can promote dysregulation of this population's homeostasis in the periphery and the Central Nervous System (CNS). We also explore the potential interconnection in observed pathological macrophage mechanisms and the function of the diverse structure of HLA alleles in neurodegenerative CNS, seen in MS, by comparing available clinical with molecular data through the prism of HLA-immunogenetics. Finally, we discuss available and experimental pharmacological approaches for MS targeting the trimolecular complex that are based on cell phenotype modulation and HLA genotype involvement and try to reveal fertile ground for the potential development of novel drugs.


Subject(s)
Alleles , Macrophages , Multiple Sclerosis , Humans , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Macrophages/immunology , Macrophages/metabolism , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Antigen Presentation/genetics , Genetic Predisposition to Disease , Animals , Polymorphism, Genetic
4.
Int J Mol Sci ; 25(13)2024 Jul 04.
Article in English | MEDLINE | ID: mdl-39000468

ABSTRACT

Previously, we found that a greater dissimilarity in swine leukocyte antigen (SLA) class I and class II alleles between mating partners resulted in increased farrowing rates in a highly inbred population of Microminipigs (MMPs). In this follow-up study, we have analyzed the effects of dissimilarity in SLA alleles between mating partners for seven different reproductive traits, including litter size and the number of stillborn and live or dead weaned piglets. We determined the relationships among reproductive traits within each mating event and the amino acid distances of SLA alleles as markers of diversity between mating partners. Our results indicate that mating partners with greater amino acid pairwise genetic distances in the SLA-1 class I gene or DQB1 class II gene alleles were associated with significantly larger litter sizes and higher numbers of live piglets at birth and weaning. Also, partners with greater pairwise distances in the SLA-2 class I gene alleles exhibited fewer pre-weaning deaths. These findings suggest that the dissimilarity in SLA class I and class II alleles between mating partners may affect not only farrowing rates but also other key reproductive traits such as litter size and improved piglet survival rates. Consequently, SLA alleles could serve as valuable genetic markers for selecting mating partners in breeding programs and for conducting epistatic studies on various reproductive traits in MMPs.


Subject(s)
Alleles , Histocompatibility Antigens Class I , Reproduction , Animals , Swine/genetics , Histocompatibility Antigens Class I/genetics , Reproduction/genetics , Female , Litter Size/genetics , Swine, Miniature/genetics , Male , Histocompatibility Antigens Class II/genetics , Amino Acids/genetics
5.
Nat Commun ; 15(1): 5895, 2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39003267

ABSTRACT

Autoimmune thyroid diseases (AITD) such as Graves' disease (GD) or Hashimoto's thyroiditis (HT) are organ-specific diseases that involve complex interactions between distinct components of thyroid tissue. Here, we use spatial transcriptomics to explore the molecular architecture, heterogeneity and location of different cells present in the thyroid tissue, including thyroid follicular cells (TFCs), stromal cells such as fibroblasts, endothelial cells, and thyroid infiltrating lymphocytes. We identify damaged antigen-presenting TFCs with upregulated CD74 and MIF expression in thyroid samples from AITD patients. Furthermore, we discern two main fibroblast subpopulations in the connective tissue including ADIRF+ myofibroblasts, mainly enriched in GD, and inflammatory fibroblasts, enriched in HT patients. We also demonstrate an increase of fenestrated PLVAP+ vessels in AITD, especially in GD. Our data unveil stromal and thyroid epithelial cell subpopulations that could play a role in the pathogenesis of AITD.


Subject(s)
Antigens, Differentiation, B-Lymphocyte , Graves Disease , Hashimoto Disease , Thyroid Gland , Humans , Graves Disease/pathology , Graves Disease/immunology , Graves Disease/genetics , Graves Disease/metabolism , Thyroid Gland/pathology , Thyroid Gland/metabolism , Hashimoto Disease/pathology , Hashimoto Disease/immunology , Hashimoto Disease/metabolism , Hashimoto Disease/genetics , Antigens, Differentiation, B-Lymphocyte/metabolism , Antigens, Differentiation, B-Lymphocyte/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Histocompatibility Antigens Class II/metabolism , Histocompatibility Antigens Class II/genetics , Thyroid Epithelial Cells/metabolism , Thyroid Epithelial Cells/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Transcriptome , Myofibroblasts/metabolism , Myofibroblasts/pathology , Stromal Cells/metabolism , Stromal Cells/pathology , Female , Macrophage Migration-Inhibitory Factors , Intramolecular Oxidoreductases
6.
Sci Adv ; 10(28): eadk2091, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38996030

ABSTRACT

The mechanism by which interferon regulatory factor 8 (IRF8) mutation contributes to lymphomagenesis is unknown. We modeled IRF8 variants in B cell lymphomas and found that they affected the expression of regulators of antigen presentation. Expression of IRF8 mutants in murine B cell lymphomas suppressed CD4, but not CD8, activation elicited by antigen presentation and downmodulated CD74 and human leukocyte antigen (HLA) DM, intracellular regulators of antigen peptide processing/loading in the major histocompatibility complex (MHC) II. Concordantly, mutant IRF8 bound less efficiently to the promoters of these genes. Mice harboring IRF8 mutant lymphomas displayed higher tumor burden and remodeling of the tumor microenvironment, typified by depletion of CD4, CD8, and natural killer cells, increase in regulatory T cells and T follicular helper cells. Deconvolution of bulk RNA sequencing data from IRF8-mutant human diffuse large B cell lymphoma (DLBCL) recapitulated part of the immune remodeling detected in mice. We concluded that IRF8 mutations contribute to DLBCL biology by facilitating immune escape.


Subject(s)
Antigen Presentation , Antigens, Differentiation, B-Lymphocyte , Histocompatibility Antigens Class II , Interferon Regulatory Factors , Mutation , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Animals , Antigen Presentation/immunology , Antigen Presentation/genetics , Humans , Mice , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Differentiation, B-Lymphocyte/metabolism , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/immunology , Tumor Microenvironment/immunology , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/immunology , Lymphoma, Large B-Cell, Diffuse/pathology , Cell Line, Tumor , Tumor Escape/genetics , Gene Expression Regulation, Neoplastic
7.
Front Immunol ; 15: 1404668, 2024.
Article in English | MEDLINE | ID: mdl-38903492

ABSTRACT

Heart transplantation is associated with major hurdles, including the limited number of available organs for transplantation, the risk of rejection due to genetic discrepancies, and the burden of immunosuppression. In this study, we demonstrated the feasibility of permanent genetic engineering of the heart during ex vivo perfusion. Lentiviral vectors encoding for short hairpin RNAs targeting beta2-microglobulin (shß2m) and class II transactivator (shCIITA) were delivered to the graft during two hours of normothermic EVHP. Highly efficient genetic engineering was indicated by stable reporter gene expression in endothelial cells and cardiomyocytes. Remarkably, swine leucocyte antigen (SLA) class I and SLA class II expression levels were decreased by 66% and 76%, respectively, in the vascular endothelium. Evaluation of lactate, troponin T, and LDH levels in the perfusate and histological analysis showed no additional cell injury or tissue damage caused by lentiviral vectors. Moreover, cytokine secretion profiles (IL-6, IL-8, and TNF-α) of non-transduced and lentiviral vector-transduced hearts were comparable. This study demonstrated the ex vivo generation of genetically engineered hearts without compromising tissue integrity. Downregulation of SLA expression may contribute to reduce the immunogenicity of the heart and support graft survival after allogeneic or xenogeneic transplantation.


Subject(s)
Genetic Vectors , Heart Transplantation , Histocompatibility Antigens Class I , Lentivirus , Animals , Lentivirus/genetics , Heart Transplantation/methods , Genetic Vectors/genetics , Swine , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Perfusion/methods , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Histocompatibility Antigens Class II/immunology , beta 2-Microglobulin/genetics , Cytokines/metabolism , Genetic Engineering , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/immunology , Humans , RNA, Small Interfering/genetics , Graft Survival/immunology , Graft Survival/genetics , Endothelial Cells/metabolism , Endothelial Cells/immunology , Nuclear Proteins , Trans-Activators
8.
Cell Host Microbe ; 32(7): 1089-1102.e10, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38889725

ABSTRACT

Avian influenza A virus (IAV) surveillance in Northern California, USA, revealed unique IAV hemagglutinin (HA) genome sequences in cloacal swabs from lesser scaups. We found two closely related HA sequences in the same duck species in 2010 and 2013. Phylogenetic analyses suggest that both sequences belong to the recently discovered H19 subtype, which thus far has remained uncharacterized. We demonstrate that H19 does not bind the canonical IAV receptor sialic acid (Sia). Instead, H19 binds to the major histocompatibility complex class II (MHC class II), which facilitates viral entry. Unlike the broad MHC class II specificity of H17 and H18 from bat IAV, H19 exhibits a species-specific MHC class II usage that suggests a limited host range and zoonotic potential. Using cell lines overexpressing MHC class II, we rescued recombinant H19 IAV. We solved the H19 crystal structure and identified residues within the putative Sia receptor binding site (RBS) that impede Sia-dependent entry.


Subject(s)
Ducks , Hemagglutinin Glycoproteins, Influenza Virus , Histocompatibility Antigens Class II , Influenza A virus , Phylogeny , Receptors, Virus , Animals , Influenza A virus/genetics , Influenza A virus/immunology , Receptors, Virus/metabolism , Receptors, Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Histocompatibility Antigens Class II/metabolism , Histocompatibility Antigens Class II/genetics , Ducks/virology , Humans , Virus Internalization , Influenza in Birds/virology , Binding Sites , Protein Binding , Crystallography, X-Ray , Cell Line , N-Acetylneuraminic Acid/metabolism , Host Specificity , Species Specificity
9.
Methods Mol Biol ; 2809: 215-235, 2024.
Article in English | MEDLINE | ID: mdl-38907900

ABSTRACT

MHC-II molecules are key mediators of antigen presentation in vertebrate species and bind to their ligands with high specificity. The very high polymorphism of MHC-II genes within species and the fast-evolving nature of these genes across species has resulted in tens of thousands of different alleles, with hundreds of new alleles being discovered yearly through large sequencing projects in different species. Here we describe how to use MixMHC2pred to predict the binding specificity of any MHC-II allele directly from its amino acid sequence. We then show how both MHC-II ligands and CD4+ T cell epitopes can be predicted in different species with our approach. MixMHC2pred is available at http://mixmhc2pred.gfellerlab.org/ .


Subject(s)
Alleles , Epitopes, T-Lymphocyte , Histocompatibility Antigens Class II , Ligands , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Animals , Humans , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/metabolism , Protein Binding , Software , Computational Biology/methods , Antigen Presentation/genetics , Amino Acid Sequence
10.
PLoS Genet ; 20(6): e1011309, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38885192

ABSTRACT

PLK1 (Polo-like kinase 1) plays a critical role in the progression of lung adenocarcinoma (LUAD). Recent studies have unveiled that targeting PLK1 improves the efficacy of immunotherapy, highlighting its important role in the regulation of tumor immunity. Nevertheless, our understanding of the intricate interplay between PLK1 and the tumor microenvironment (TME) remains incomplete. Here, using genetically engineered mouse model and single-cell RNA-seq analysis, we report that PLK1 promotes an immunosuppressive TME in LUAD, characterized with enhanced M2 polarization of tumor associated macrophages (TAM) and dampened antigen presentation process. Mechanistically, elevated PLK1 coincides with increased secretion of CXCL2 cytokine, which promotes M2 polarization of TAM and diminishes expression of class II major histocompatibility complex (MHC-II) in professional antigen-presenting cells. Furthermore, PLK1 negatively regulates MHC-II expression in cancer cells, which has been shown to be associated with compromised tumor immunity and unfavorable patient outcomes. Taken together, our results reveal PLK1 as a novel modulator of TME in LUAD and provide possible therapeutic interventions.


Subject(s)
Adenocarcinoma of Lung , Cell Cycle Proteins , Lung Neoplasms , Polo-Like Kinase 1 , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins , Single-Cell Analysis , Tumor Microenvironment , Animals , Humans , Mice , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/immunology , Adenocarcinoma of Lung/pathology , Antigen Presentation/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Chemokine CXCL2/genetics , Chemokine CXCL2/metabolism , Gene Expression Regulation, Neoplastic , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism
11.
Inflamm Res ; 73(7): 1157-1172, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38713235

ABSTRACT

BACKGROUND: Lymphatic abnormalities are essential for pathophysiologic changes of creeping fat (CrF) in Crohn's disease (CD). Anti-tumor necrosis factor (TNF) therapy has been proved to alleviate CrF lesions, however, whether it achieves these by remodeling lymphatics is unknown. METHODS: CD74 expression was detected in CrF and uninvolved mesentery of CD patients. Lymphatic functions in vitro were evaluated and lymphatic endothelium barrier were checked by transendothelial electrical resistance (TEER) and FITC-Dextran permeability. Protein level of tight junction and signaling pathways were detected by western blotting. RESULTS: CD74 was upregulated in LECs of CrF and positively correlated with TNF-α synthesis. This was suppressed by IFX administration. In vitro, TNF-α stimulated LECs to express CD74 through NF-κB signaling pathway, and this was rescued by IFX. CD74 downregulation suppressed the abilities of LECs in proliferation, migration and tube formation. Interaction of CD74-MIF impaired LECs' barrier via reducing tight junction proteins in an ERK1/2-dependent manner, which was reversed by CD74 downregulation. Consistently, the CD patients receiving IFX therapy displayed decreased lymphangiogenesis and improved mesenteric lymphatic endothelium barrier, companied with reduced adipocyte size and adipokine levels in CrF. CONCLUSIONS: Anti-TNF therapy could modify pathological changes in CrF by alleviating CD74-mediated lymphatic abnormalities.


Subject(s)
Adipose Tissue , Antigens, Differentiation, B-Lymphocyte , Crohn Disease , Histocompatibility Antigens Class II , Infliximab , Tumor Necrosis Factor-alpha , Crohn Disease/drug therapy , Crohn Disease/metabolism , Humans , Antigens, Differentiation, B-Lymphocyte/genetics , Infliximab/therapeutic use , Infliximab/pharmacology , Adipose Tissue/drug effects , Adipose Tissue/pathology , Adipose Tissue/metabolism , Tumor Necrosis Factor-alpha/metabolism , Male , Female , Adult , Histocompatibility Antigens Class II/genetics , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Cells, Cultured , Young Adult , Middle Aged , Gastrointestinal Agents/therapeutic use , Gastrointestinal Agents/pharmacology , NF-kappa B/metabolism , Lymphangiogenesis/drug effects , Cell Movement/drug effects
12.
Nat Commun ; 15(1): 4418, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38806459

ABSTRACT

The mechanisms by which the number of memory CD8 T cells is stably maintained remains incompletely understood. It has been postulated that maintaining them requires help from CD4 T cells, because adoptively transferred memory CD8 T cells persist poorly in MHC class II (MHCII)-deficient mice. Here we show that chronic interferon-γ signals, not CD4 T cell-deficiency, are responsible for their attrition in MHCII-deficient environments. Excess IFN-γ is produced primarily by endogenous colonic CD8 T cells in MHCII-deficient mice. IFN-γ neutralization restores the number of memory CD8 T cells in MHCII-deficient mice, whereas repeated IFN-γ administration or transduction of a gain-of-function STAT1 mutant reduces their number in wild-type mice. CD127high memory cells proliferate actively in response to IFN-γ signals, but are more susceptible to attrition than CD127low terminally differentiated effector memory cells. Furthermore, single-cell RNA-sequencing of memory CD8 T cells reveals proliferating cells that resemble short-lived, terminal effector cells and documents global downregulation of gene signatures of long-lived memory cells in MHCII-deficient environments. We propose that chronic IFN-γ signals deplete memory CD8 T cells by compromising their long-term survival and by diverting self-renewing CD127high cells toward terminal differentiation.


Subject(s)
CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Immunologic Memory , Interferon-gamma , STAT1 Transcription Factor , Animals , CD8-Positive T-Lymphocytes/immunology , Interferon-gamma/metabolism , Interferon-gamma/immunology , CD4-Positive T-Lymphocytes/immunology , Mice , STAT1 Transcription Factor/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/deficiency , Mice, Inbred C57BL , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Signal Transduction , Mice, Knockout , Memory T Cells/immunology , Memory T Cells/metabolism , Interleukin-7 Receptor alpha Subunit/metabolism , Cell Proliferation , Adoptive Transfer
13.
Animal Model Exp Med ; 7(3): 377-387, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38720469

ABSTRACT

BACKGROUND: Atherosclerosis is a chronic cardiovascular disease of great concern. However, it is difficult to establish a direct connection between conventional small animal models and clinical practice. The pig's genome, physiology, and anatomy reflect human biology better than other laboratory animals, which is crucial for studying the pathogenesis of atherosclerosis. METHODS: We used whole-genome sequencing data from nine Bama minipigs to perform a genome-wide linkage analysis, and further used bioinformatic tools to filter and identify underlying candidate genes. Candidate gene function prediction was performed using the online prediction tool STRING 12.0. Immunohistochemistry and immunofluorescence were used to detect the expression of proteins encoded by candidate genes. RESULTS: We mapped differential single nucleotide polymorphisms (SNPs) to genes and obtained a total of 102 differential genes, then we used GO and KEGG pathway enrichment analysis to identify four candidate genes, including SLA-1, SLA-2, SLA-3, and TAP2. nsSNPs cause changes in the primary and tertiary structures of SLA-I and TAP2 proteins, the primary structures of these two proteins have undergone amino acid changes, and the tertiary structures also show slight changes. In addition, immunohistochemistry and immunofluorescence results showed that the expression changes of TAP2 protein in coronary arteries showed a trend of increasing from the middle layer to the inner layer. CONCLUSIONS: We have identified SLA-I and TAP2 as potential susceptibility genes of atherosclerosis, highlighting the importance of antigen processing and immune response in atherogenesis.


Subject(s)
Atherosclerosis , Polymorphism, Single Nucleotide , Swine, Miniature , Animals , Swine , Atherosclerosis/genetics , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Swine Diseases/genetics , Swine Diseases/pathology , Genetic Predisposition to Disease , Male , Antigens, CD/genetics , Antigens, CD/metabolism
14.
Tohoku J Exp Med ; 263(2): 133-139, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38692862

ABSTRACT

Genetic factors, particularly human leukocyte antigen (HLA) class II genes, are known to significantly influence the onset of type 1 diabetes (T1D). Additionally, patients with T1D often develop autoimmune thyroid diseases (AITD). Despite this association, comprehensive research on individuals with both AITD and T1D in Japan, especially regarding the influence of specific HLA alleles, remains insufficient. In this retrospective study, we analyzed 44 inpatients diagnosed with T1D. These patients were predominantly female, with an average onset age of 35 years, poor blood sugar control, and approximately 43.2% had concurrent AITD. We observed significant associations of HLA-DRB1*04:05, HLA-DRB1*09:01 and HLA-DRB1*15:02 alleles with T1D regardless of AITD presence, which had been previously established for T1D in Japanese. In this context, comparing Japanese patients with AITD alone, we noted AITD comorbidity with T1D results in alterations in the frequencies of HLA-DRB1*09:01, HLA-DRB1*04:03, and HLA-DRB1*15:02. Furthermore, HLA-DRB1*04:05, HLA-DRB1*09:01, HLA-DRB1*13:02, and HLA-DRB1*15:01 alleles may be alleles whose susceptibility varies for both conditions. These findings underscore the importance of understanding the relationship between T1D, AITD, and HLA genetics, which may inform personalized treatment strategies and facilitate the development of targeted therapies. Future research endeavors should aim to elucidate underlying mechanisms and validate these findings in larger cohorts.


Subject(s)
Alleles , Diabetes Mellitus, Type 1 , Genetic Predisposition to Disease , Humans , Diabetes Mellitus, Type 1/genetics , Female , Male , Adult , HLA-DRB1 Chains/genetics , Japan , Asian People/genetics , Thyroiditis, Autoimmune/genetics , Middle Aged , Gene Frequency/genetics , Genes, MHC Class II/genetics , Histocompatibility Antigens Class II/genetics , Young Adult , Adolescent , East Asian People
15.
Oncogene ; 43(28): 2215-2227, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38802647

ABSTRACT

Approximately 40% of patients with lung adenocarcinoma (LUAD) often develop bone metastases during the course of their disease. However, scarcely any in vivo model of LUAD bone metastasis has been established, leading to a poor understanding of the mechanisms underlying LUAD bone metastasis. Here, we established a multiorgan metastasis model via the left ventricular injection of luciferase-labeled LUAD cells into nude mice and then screened out lung metastasis (LuM) and bone metastasis (BoM) cell subpopulations. BoM cells exhibited greater stemness and epithelial-mesenchymal transition (EMT) plasticity than LuM cells and initially colonized the bone and subsequently disseminated to distant organs after being reinjected into mice. Moreover, a CD74-ROS1 fusion mutation (C6; R34) was detected in BoM cells but not in LuM cells. Mechanistically, BoM cells bearing the CD74-ROS1 fusion highly secrete the C-C motif chemokine ligand 5 (CCL5) protein by activating STAT3 signaling, recruiting macrophages in tumor microenvironment and strongly inducing M2 polarization of macrophages. BoM cell-activated macrophages produce a high level of TGF-ß1, thereby facilitating EMT and invasion of LUAD cells via TGF-ß/SMAD2/3 signaling. Targeting the CD74-ROS1/CCL5 axis with Crizotinib (a ROS1 inhibitor) and Maraviroc (a CCL5 receptor inhibitor) in vivo strongly impeded bone metastasis and secondary metastasis of BoM cells. Our findings reveal the critical role of the CD74-ROS1/STAT3/CCL5 axis in the interaction between LUAD bone metastasis cells and macrophages for controlling LUAD cell dissemination, highlighting the significance of the bone microenvironment in LUAD bone metastasis and multiorgan secondary metastasis, and suggesting that targeting CD74-ROS1 and CCL5 is a promising therapeutic strategy for LUAD bone metastasis.


Subject(s)
Adenocarcinoma of Lung , Bone Neoplasms , Epithelial-Mesenchymal Transition , Lung Neoplasms , Macrophages , Protein-Tyrosine Kinases , Proto-Oncogene Proteins , Animals , Humans , Mice , Bone Neoplasms/secondary , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Macrophages/metabolism , Macrophages/pathology , Lung Neoplasms/secondary , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/secondary , Adenocarcinoma of Lung/metabolism , Epithelial-Mesenchymal Transition/genetics , Protein-Tyrosine Kinases/metabolism , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Antigens, Differentiation, B-Lymphocyte/metabolism , Antigens, Differentiation, B-Lymphocyte/genetics , Chemokine CCL5/metabolism , Chemokine CCL5/genetics , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Mice, Nude , Cell Line, Tumor , Tumor Microenvironment , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Signal Transduction
16.
Virology ; 595: 110083, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38696887

ABSTRACT

Porcine reproductive and respiratory syndrome virus (PRRSV) infection inhibits swine leukocyte antigen class I (SLA-I) expression in pigs, resulting in inefficient antigen presentation and subsequent low levels of cellular PRRSV-specific immunity as well as persistent viremia. We previously observed that the non-structural protein 4 (nsp4) of PRRSV contributed to inhibition of the ß2-microglobulin (ß2M) and SLA-I expression in cells. Here, we constructed a series of nsp4 mutants with different combination of amino acid mutations to attenuate the inhibitory effect of nsp4 on ß2M and SLA-I expression. Almost all nsp4 mutants exogenously expressed in cells showed an attenuated effect on inhibition of ß2M and SLA-I expression, but the recombinant PRRSV harboring these nsp4 mutants failed to be rescued with exception of the rPRRSV-nsp4-mut10 harboring three amino acid mutations. However, infection of rPRRSV-nsp4-mut10 not only enhanced ß2M and SLA-I expression in both cells and pigs but also promoted the DCs to active the CD3+CD8+T lymphocytes more efficiently, as compared with its parental PRRSV (rPRRVS-nsp4-wt). These data suggested that the inhibition of nsp4-mediated ß2M downregulation improved ß2M/SLA-I expression in pigs.


Subject(s)
Down-Regulation , Histocompatibility Antigens Class I , Porcine Reproductive and Respiratory Syndrome , Porcine respiratory and reproductive syndrome virus , Viral Nonstructural Proteins , beta 2-Microglobulin , Porcine respiratory and reproductive syndrome virus/genetics , Porcine respiratory and reproductive syndrome virus/physiology , Porcine respiratory and reproductive syndrome virus/immunology , Animals , Swine , Porcine Reproductive and Respiratory Syndrome/virology , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine Reproductive and Respiratory Syndrome/genetics , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Viral Nonstructural Proteins/immunology , beta 2-Microglobulin/genetics , beta 2-Microglobulin/metabolism , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Histocompatibility Antigens Class II/immunology , Cell Line , CD8-Positive T-Lymphocytes/immunology , Mutation
17.
Nat Commun ; 15(1): 3749, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702311

ABSTRACT

Regulatory T cells (Tregs) are plastic cells playing a pivotal role in the maintenance of immune homeostasis. Tregs actively adapt to the microenvironment where they reside; as a consequence, their molecular and functional profiles differ among tissues and pathologies. In tumors, the features acquired by Tregs remains poorly characterized. Here, we observe that human tumor-infiltrating Tregs selectively overexpress CD74, the MHC class II invariant chain. CD74 has been previously described as a regulator of antigen-presenting cell biology, however its function in Tregs remains unknown. CD74 genetic deletion in human primary Tregs reveals that CD74KO Tregs exhibit major defects in the organization of their actin cytoskeleton and intracellular organelles. Additionally, intratumoral CD74KO Tregs show a decreased activation, a drop in Foxp3 expression, a low accumulation in the tumor, and consistently, they are associated with accelerated tumor rejection in preclinical models in female mice. These observations are unique to tumor conditions as, at steady state, CD74KO-Treg phenotype, survival, and suppressive capacity are unaffected in vitro and in vivo. CD74 therefore emerges as a specific regulator of tumor-infiltrating Tregs and as a target to interfere with Treg anti-tumor activity.


Subject(s)
Antigens, Differentiation, B-Lymphocyte , Histocompatibility Antigens Class II , T-Lymphocytes, Regulatory , T-Lymphocytes, Regulatory/immunology , Animals , Antigens, Differentiation, B-Lymphocyte/metabolism , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Differentiation, B-Lymphocyte/immunology , Histocompatibility Antigens Class II/metabolism , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/genetics , Humans , Female , Mice , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Tumor Microenvironment/immunology , Neoplasms/immunology , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice, Inbred C57BL , Mice, Knockout
18.
J Cell Sci ; 137(10)2024 May 15.
Article in English | MEDLINE | ID: mdl-38682259

ABSTRACT

SARS-CoV-2 interferes with antigen presentation by downregulating major histocompatibility complex (MHC) II on antigen-presenting cells, but the mechanism mediating this process is unelucidated. Herein, analysis of protein and gene expression in human antigen-presenting cells reveals that MHC II is downregulated by the SARS-CoV-2 main protease, NSP5. This suppression of MHC II expression occurs via decreased expression of the MHC II regulatory protein CIITA. CIITA downregulation is independent of the proteolytic activity of NSP5, and rather, NSP5 delivers HDAC2 to the transcription factor IRF3 at an IRF-binding site within the CIITA promoter. Here, HDAC2 deacetylates and inactivates the CIITA promoter. This loss of CIITA expression prevents further expression of MHC II, with this suppression alleviated by ectopic expression of CIITA or knockdown of HDAC2. These results identify a mechanism by which SARS-CoV-2 limits MHC II expression, thereby delaying or weakening the subsequent adaptive immune response.


Subject(s)
Histocompatibility Antigens Class II , Histone Deacetylase 2 , Nuclear Proteins , Promoter Regions, Genetic , SARS-CoV-2 , Trans-Activators , Humans , Antigen Presentation/genetics , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/immunology , COVID-19/virology , COVID-19/immunology , COVID-19/genetics , COVID-19/metabolism , Cysteine Endopeptidases/metabolism , Cysteine Endopeptidases/genetics , Down-Regulation/genetics , HEK293 Cells , Histocompatibility Antigens Class II/metabolism , Histocompatibility Antigens Class II/genetics , Histone Deacetylase 2/metabolism , Histone Deacetylase 2/genetics , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Promoter Regions, Genetic/genetics , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , SARS-CoV-2/immunology , Trans-Activators/metabolism , Trans-Activators/genetics , Viral Nonstructural Proteins/metabolism , Viral Nonstructural Proteins/genetics
19.
Adv Sci (Weinh) ; 11(23): e2401513, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38602454

ABSTRACT

Transgenic mice expressing human major histocompatibility complex class II (MHCII) risk alleles are widely used in autoimmune disease research, but limitations arise due to non-physiologic expression. To address this, physiologically relevant mouse models are established via knock-in technology to explore the role of MHCII in diseases like rheumatoid arthritis. The gene sequences encoding the ectodomains are replaced with the human DRB1*04:01 and 04:02 alleles, DRA, and CD74 (invariant chain) in C57BL/6N mice. The collagen type II (Col2a1) gene is modified to mimic human COL2. Importantly, DRB1*04:01 knock-in mice display physiologic expression of human MHCII also on thymic epithelial cells, in contrast to DRB1*04:01 transgenic mice. Humanization of the invariant chain enhances MHCII expression on thymic epithelial cells, increases mature B cell numbers in spleen, and improves antigen presentation. To validate its functionality, the collagen-induced arthritis (CIA) model is used, where DRB1*04:01 expression led to a higher susceptibility to arthritis, as compared with mice expressing DRB1*04:02. In addition, the humanized T cell epitope on COL2 allows autoreactive T cell-mediated arthritis development. In conclusion, the humanized knock-in mouse faithfully expresses MHCII, confirming the DRB1*04:01 alleles role in rheumatoid arthritis and being also useful for studying MHCII-associated diseases.


Subject(s)
Alleles , Antigens, Differentiation, B-Lymphocyte , Arthritis, Rheumatoid , Disease Models, Animal , Gene Knock-In Techniques , Histocompatibility Antigens Class II , Mice, Inbred C57BL , Mice, Transgenic , Animals , Mice , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , Antigens, Differentiation, B-Lymphocyte/genetics , Antigens, Differentiation, B-Lymphocyte/immunology , Humans , Gene Knock-In Techniques/methods , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Arthritis, Experimental/genetics , Arthritis, Experimental/immunology , HLA-DRB1 Chains/genetics , HLA-DRB1 Chains/immunology , Collagen Type II/genetics , Collagen Type II/immunology
20.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 32(2): 603-609, 2024 Apr.
Article in Chinese | MEDLINE | ID: mdl-38660873

ABSTRACT

OBJECTIVE: To investigate the accuracy of next-generation sequencing technology (NGS) in detecting the polymorphisms of HLA-DRB1, DQB1, DQA1, DRB3, DRB4, DRB5, DPA1 and DPB1 alleles in randomly-selected unrelated healthy individuals from Shenzhen Han population, investigate the potential reason for HLA-DRB1 allele dropout in routine NGS, and establish an internal quality control system. METHODS: NGS-based HLA class II genotyping was performed on 1 012 samples using the MiSeqDxTM platform. The suspected missed alleles indicated by the quality control software and HLA-DRB1 homozygotes were confirmed by PCR-SSOP or PCR-SBT methods. RESULTS: A total of 139 alleles were detected, including HLA-DRB1(45), DRB3(7), DRB4(5), DRB5(7), DQA1(17), DQB1(21), DPA1(10) and DPB1(27). HLA-DRB1*09:01(17.09%),15:01(10.72%); DRB3*02:02(25.99%),03:01(10.18%); DRB4*01:03(36.46%); DRB5*01:01(15.42%); DQA1*01:02(20.01%),03:02(17.19%); DQB1*03:01(19.47%),03:03(17.98%), 05:02(11.66%), 06:01(10.67%); DPA1*02:02(54.45%), 01:03(31.18%) and DPB1*05:01(39.13%), 02:01(16.90%) alleles were the most common alleles in Shenzhen Han population (frequencies >10%). There was no statistical difference between the gene frequencies of HLA-DRB1 and DQB1 loci in our study. The HLA Common and Well-Documented Alleles in China (CWD2.4) (χ2=12.68, P >0.05). 94 cases of HLA-DRB1 homozygous samples detected by NGS were retested by PCR-SSOP or SBT method, and one case of allele dropout at HLA-DRB1 locus was found. SBT method confirmed that the allele of DRB1*04:03 was missed. The laboratory internal quality control system was established. Two cases of new alleles were detected and named by WHO Nomenclature Committee for Factors of the HLA System. CONCLUSION: The HLA genotyping results based on NGS showed a significantly lower ambiguity rate. The HLA class II alleles exhibit genetic polymorphism in the Han population of unrelated healthy individuals in Shenzhen. The independent method based on NGS in clinical histocompatibility testing has limitations and requires internal quality control strategies to avoid allele-dropout events.


Subject(s)
East Asian People , Genotype , High-Throughput Nucleotide Sequencing , Histocompatibility Antigens Class II , Humans , Alleles , Gene Frequency , Polymorphism, Genetic , East Asian People/genetics , Histocompatibility Antigens Class II/genetics
SELECTION OF CITATIONS
SEARCH DETAIL