Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 181
Filter
1.
Med Sci Monit ; 27: e934491, 2021 Dec 05.
Article in English | MEDLINE | ID: mdl-34864813

ABSTRACT

BACKGROUND The exact mechanisms of lung adenocarcinoma (LUAD) etiology and pathogenesis remain unclear. MATERIAL AND METHODS In this study, we evaluated alternative splicing (AS) events in LUAD. We separately analyzed LUAD data from The Cancer Genome Atlas (TCGA) database and AS-related feature lists from SpliceSeq to develop risk models for AS events and further validated risk models for AS events. The association between immune-related features and the risk model of AS events was evaluated. RESULTS We found that exon skip (ES) and mutually exclusive exons (ME) exhibited the most and least AS events, respectively. The risk score and stage of LUAD patients were strongly associated with prognosis and were independent influences on the prognosis of LUAD. The pathological stage (stage, T, N) and risk model were incorporated to construct a column line graph with better predictive ability. Risk models were associated with tumor microenvironment, and higher risk score values were associated with higher M2 macrophage content and lower M0 macrophage and helper T lymphocyte content. The correlation between core genes and immunity was further assessed by analyzing differentially-expressed genes between risk models. These results suggest an association between the level of risk for AS events and the density of immune cell infiltration. CONCLUSIONS Our findings suggest a clear association between AS risk model and patient prognosis, and was performed to confirm the biological relationship between AS and immunity.


Subject(s)
Adenocarcinoma of Lung/immunology , Alternative Splicing/immunology , Lung Neoplasms/immunology , Female , Humans , Lung/immunology , Male , Middle Aged
2.
BMC Pulm Med ; 21(1): 400, 2021 Dec 06.
Article in English | MEDLINE | ID: mdl-34872548

ABSTRACT

Lung adenocarcinoma (LUAD) is the leading cause of cancer deaths worldwide due to the lack of early diagnostic markers and specific drugs. Previous studies have shown the association of LUAD growth with aberrant alternative splicing (AS). Herein, clinical data of 535 tumor tissues and 59 normal tissues were extracted from The Cancer Genome Atlas (TCGA) database. Each sample was analyzed using the ESTIMATE algorithm; a comparison between higher and lower score groups (stromal or immune) was made to determine the overall- and progression-free survival-related differentially expressed AS (DEAS) events. We then performed unsupervised clustering of these DEASs, followed by determining their relationship with survival rate, immune cells, and the tumor microenvironment (TME). Next, two prognostic signatures were developed using bioinformatics tools to explore the prognosis of cases with LUAD. Five OS- and six PFS-associated DEAS events were implemented to establish a prognostic risk score model. When compared to the high-risk group (HRG), the PFS and OS of the low-risk group (LRG) were found to be considerable. Additionally, a better prognosis was found considerably associated with the ESTIMATE score of the patients as well as immune cells infiltration. Our analysis of AS events in LUAD not only helps to clarify the tumorigenesis mechanism of AS but also provides ideas for revealing potential prognostic biomarkers and therapeutic targets.


Subject(s)
Adenocarcinoma of Lung , Alternative Splicing , Biomarkers, Tumor , Lung Neoplasms , Risk Assessment/methods , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/immunology , Adenocarcinoma of Lung/mortality , Adenocarcinoma of Lung/pathology , Algorithms , Alternative Splicing/genetics , Alternative Splicing/immunology , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Carcinogenesis , China , Databases, Genetic , Disease-Free Survival , Humans , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Prognosis , Survival Rate , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
3.
Immunohorizons ; 5(10): 884-897, 2021 10 29.
Article in English | MEDLINE | ID: mdl-34716181

ABSTRACT

Protein arginine methyltransferase (PRMT) 5 is the type 2 methyltransferase catalyzing symmetric dimethylation of arginine. PRMT5 inhibition or deletion in CD4 Th cells reduces TCR engagement-induced IL-2 production and Th cell expansion and confers protection against experimental autoimmune encephalomyelitis, the animal model of multiple sclerosis. However, the mechanisms by which PRMT5 modulates Th cell proliferation are still not completely understood, and neither are the methylation targets in T cells. In this manuscript, we uncover the role of PRMT5 on alternative splicing in activated mouse T cells and identify several targets of PRMT5 symmetric dimethylation involved in splicing. In addition, we find a possible link between PRMT5-mediated alternative splicing of transient receptor potential cation channel subfamily M member 4 (Trpm4) and TCR/NFAT signaling/IL-2 production. This understanding may guide development of drugs targeting these processes to benefit patients with T cell-mediated diseases.


Subject(s)
Alternative Splicing/immunology , CD4-Positive T-Lymphocytes/immunology , Protein-Arginine N-Methyltransferases/metabolism , TRPM Cation Channels/genetics , Animals , CD4-Positive T-Lymphocytes/metabolism , Calcium/metabolism , Cells, Cultured , Female , Gene Knockdown Techniques , Lymphocyte Activation/genetics , Male , Methylation , Mice , Models, Animal , NFATC Transcription Factors/metabolism , Primary Cell Culture , Protein-Arginine N-Methyltransferases/genetics , RNA-Seq , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/genetics , Signal Transduction/immunology
4.
PLoS Comput Biol ; 17(9): e1009411, 2021 09.
Article in English | MEDLINE | ID: mdl-34529669

ABSTRACT

Immunotherapies provide effective treatments for previously untreatable tumors and identifying tumor-specific epitopes can help elucidate the molecular determinants of therapy response. Here, we describe a pipeline, ISOTOPE (ISOform-guided prediction of epiTOPEs In Cancer), for the comprehensive identification of tumor-specific splicing-derived epitopes. Using RNA sequencing and mass spectrometry for MHC-I associated proteins, ISOTOPE identified neoepitopes from tumor-specific splicing events that are potentially presented by MHC-I complexes. Analysis of multiple samples indicates that splicing alterations may affect the production of self-epitopes and generate more candidate neoepitopes than somatic mutations. Although there was no difference in the number of splicing-derived neoepitopes between responders and non-responders to immune therapy, higher MHC-I binding affinity was associated with a positive response. Our analyses highlight the diversity of the immunogenic impacts of tumor-specific splicing alterations and the importance of studying splicing alterations to fully characterize tumors in the context of immunotherapies. ISOTOPE is available at https://github.com/comprna/ISOTOPE.


Subject(s)
Epitopes/genetics , Epitopes/immunology , Neoplasms/genetics , Neoplasms/immunology , Alternative Splicing/genetics , Alternative Splicing/immunology , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Carcinoma, Small Cell/genetics , Carcinoma, Small Cell/immunology , Cell Line, Tumor , Computational Biology , Female , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Immunotherapy , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Male , Melanoma/genetics , Melanoma/immunology , Models, Genetic , Models, Immunological , Mutation , Neoplasms/therapy , Protein Isoforms/genetics , Protein Isoforms/immunology , RNA Splicing/genetics , RNA Splicing/immunology , RNA-Seq
5.
Immunohorizons ; 5(9): 760-771, 2021 09 28.
Article in English | MEDLINE | ID: mdl-34583937

ABSTRACT

Alternative splicing (AS) plays an important role in the development of many cell types; however, its contribution to Th subsets has been clearly defined. In this study, we compare mice naive CD4+ Th cells with Th1, Th2, Th17, and T regulatory cells and observed that the majority of AS events were retained intron, followed by skipped-exon events, with at least 1200 genes across cell types affected by AS events. A significant fraction of the AS events, especially retained intron events from the 72-h time point, were no longer observed 2 wk postdifferentiation, suggesting a role for AS in early activation and differentiation via preferential expression of specific isoforms required during T cell activation, but not for differentiation or effector function. Examining the protein consequence of the exon-skipping events revealed an abundance of structural proteins encoding for intrinsically unstructured peptide regions, followed by transmembrane helices, ß strands, and polypeptide turn motifs. Analyses of expression profiles of RNA-binding proteins (RBPs) and their cognate binding sites flanking the discovered AS events revealed an enrichment for specific RBP recognition sites in each of the Th subsets. Integration with publicly available chromatin immunoprecipitation sequencing datasets for transcription factors support a model wherein lineage-determining transcription factors impact the RBP profile within the differentiating cells, and this differential expression contributes to AS of the transcriptome via a cascade of cell type-specific posttranscriptional rewiring events.


Subject(s)
Alternative Splicing/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Binding Sites , Cells, Cultured , Datasets as Topic , Lymphocyte Activation/genetics , Mice , Models, Animal , Primary Cell Culture , RNA-Binding Proteins/metabolism , RNA-Seq , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , Transcription Factors/metabolism
6.
Front Immunol ; 12: 778204, 2021.
Article in English | MEDLINE | ID: mdl-34975865

ABSTRACT

Purpose: Interferon beta receptor 2 subunit (IFNAR2) can be produced as a transmembrane protein, but also as a soluble form (sIFNAR2) generated by alternative splicing or proteolytic cleavage, which has both agonist and antagonist activities for IFN-ß. However, its role regarding the clinical response to IFN-ß for relapsing-remitting multiple sclerosis (RRMS) is unknown. We aim to evaluate the in vitro short-term effects and after 6 and 12 months of IFN-ß therapy on sIFNAR2 production and their association with the clinical response in MS patients. Methods: Ninety-four RRMS patients were included and evaluated at baseline, 6 and 12 months from treatment onset. A subset of 41 patients were classified as responders and non-responders to IFN-ß therapy. sIFNAR2 serum levels were measured by ELISA. mRNA expression for IFNAR1, IFNAR2 splice variants, MxA and proteases were assessed by RT-PCR. The short-term effect was evaluated in PBMC from RRMS patients after IFN-ß stimulation in vitro. Results: Protein and mRNA levels of sIFNAR2 increased after IFN-ß treatment. According to the clinical response, only non-responders increased sIFNAR2 significantly at both protein and mRNA levels. sIFNAR2 gene expression correlated with the transmembrane isoform expression and was 2.3-fold higher. While MxA gene expression increased significantly after treatment, IFNAR1 and IFNAR2 only slightly increased. After short-term IFN-ß in vitro induction of PBMC, 6/7 patients increased the sIFNAR2 expression. Conclusions: IFN-ß administration induces the production of sIFNAR2 in RRMS and higher levels might be associated to the reduction of therapeutic response. Thus, levels of sIFNAR2 could be monitored to optimize an effective response to IFN-ß therapy.


Subject(s)
Drug Resistance/genetics , Interferon-beta/pharmacology , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Receptor, Interferon alpha-beta/genetics , Alternative Splicing/drug effects , Alternative Splicing/immunology , Biomarkers/blood , Biomarkers/metabolism , Drug Monitoring/methods , Female , Follow-Up Studies , Humans , Interferon-beta/therapeutic use , Male , Multiple Sclerosis, Relapsing-Remitting/blood , Multiple Sclerosis, Relapsing-Remitting/diagnosis , Multiple Sclerosis, Relapsing-Remitting/immunology , Protein Isoforms/blood , Protein Isoforms/metabolism , RNA, Messenger/blood , RNA, Messenger/metabolism , Receptor, Interferon alpha-beta/blood , Receptor, Interferon alpha-beta/metabolism , Treatment Outcome , Up-Regulation/drug effects , Up-Regulation/immunology
7.
Dev Comp Immunol ; 118: 103975, 2021 05.
Article in English | MEDLINE | ID: mdl-33383068

ABSTRACT

Alternative splicing is an essential molecular mechanism that increase the protein diversity of a species to regulate important biological processes. As a transcription factor, Interleukin-2 enhancer binding factor 2 (ILF2) regulates the functions of interleukin-2 (IL-2) at the levels of transcription, splicing and translation, and plays other critical roles in the immune system. ILF2 is well-documented in vertebrates, while little is currently known in crustacean species such as the Pacific white shrimp (Litopenaeus vannamei). In the present study, five cDNA for spliced isoforms of Lv-ILF2 were identified, in which four of them are the full-length long isoforms (Lv-ILF2-L1, Lv-ILF2-L2, Lv-ILF2-L3 and Lv-ILF2-L4) and one of them is a truncated short isoform (Lv-ILF2-S). The whole sequence of ILF2 gene from L. vannamei was obtained, which is 11,680 bp in length with 9 exons separated by 8 introns. All five isoforms contain a domain associated with zinc fingers (DZF). Two alternative splicing types (alternative 5' splice site and alternative 3' splice site) were identified in the five isoforms. The Lv-ILF2 mRNA showed a broad distribution in all detected tissues, and the Lv-ILF2-L transcript levels were higher than those of Lv-ILF2-S in corresponding tissues. The mRNA levels of Lv-ILF2-S in the hepatopancreas, heart, muscle and stomach, but not in the eyestalk, were significantly increased after challenges with Vibrio harveyi or lipopolysaccharide (LPS), while no significant changes were observed for the transcript levels of Lv-ILF2-L in these tissues under the same immune stimulants. On the contrary, the transcript levels of neither Lv-ILF2-S nor Lv-ILF2-L were affected by challenges of polyinosinic: polycytidylic acid [Poly (I:C)]. In addition, after knockdown of the Lv-ILF2 mRNA level by siRNA, the mortality of shrimp and the hepatopancreatic bacterial numbers were significantly increased under V. harveyi challenge, indicating that Lv-ILF2 might participate in the immune defenses against V. harveyi invasion. Collectively, our study here supplied the first evidence for a novel splicing mechanism of ILF2 transcripts, and provided a functional link between the Lv-ILF2 isoforms and the capacity against pathogenic Vibrio in penaeid shrimp.


Subject(s)
Arthropod Proteins/metabolism , Immunity, Innate/genetics , Nuclear Factor 45 Protein/metabolism , Penaeidae/immunology , Vibrio/immunology , Alternative Splicing/immunology , Animals , Aquaculture , Arthropod Proteins/genetics , Gene Knockdown Techniques , Nuclear Factor 45 Protein/genetics , Penaeidae/microbiology , Protein Isoforms/genetics , Protein Isoforms/metabolism
8.
Sci Rep ; 10(1): 15429, 2020 09 22.
Article in English | MEDLINE | ID: mdl-32963314

ABSTRACT

Mucosal Associated Invariant T (MAIT) cells can sense intracellular infection by a broad array of pathogens. These cells are activated upon encountering microbial antigen(s) displayed by MR1 on the surface of an infected cell. Human MR1 undergoes alternative splicing. The full-length isoform, MR1A, can activate MAIT cells, while the function of the isoforms, MR1B and MR1C, are incompletely understood. In this report, we sought to characterize the expression and function of these splice variants. Using a transcriptomic analysis in conjunction with qPCR, we find that that MR1A and MR1B transcripts are widely expressed. However only MR1A can present mycobacterial antigen to MAIT cells. Coexpression of MR1B with MR1A decreases MAIT cell activation following bacterial infection. Additionally, expression of MR1B prior to MR1A lowers total MR1A abundance, suggesting competition between MR1A and MR1B for either ligands or chaperones required for folding and/or trafficking. Finally, we evaluated CD4/CD8 double positive thymocytes expressing surface MR1. Here, we find that relative expression of MR1A/MR1B transcript is associated with the prevalence of MR1 + CD4/CD8 cells in the thymus. Our results suggest alternative splicing of MR1 represents a means of regulating MAIT activation in response to microbial ligand(s).


Subject(s)
Alternative Splicing/genetics , Alternative Splicing/immunology , Antigen Presentation/genetics , Antigen Presentation/immunology , Histocompatibility Antigens Class I/genetics , Minor Histocompatibility Antigens/genetics , Mucosal-Associated Invariant T Cells/immunology , A549 Cells , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line , Cell Line, Tumor , HEK293 Cells , Histocompatibility Antigens Class I/immunology , Humans , Ligands , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Minor Histocompatibility Antigens/immunology , Protein Isoforms/genetics , Protein Isoforms/immunology , Protein Transport/genetics , Protein Transport/immunology , Thymocytes/immunology , Transcriptome/genetics , Transcriptome/immunology
9.
Inflamm Bowel Dis ; 26(12): 1856-1868, 2020 11 19.
Article in English | MEDLINE | ID: mdl-32304568

ABSTRACT

Adherent-invasive Escherichia coli have been suggested to play a pivotal role within the pathophysiology of inflammatory bowel disease (IBD). Autoantibodies against distinct splicing variants of glycoprotein 2 (GP2), an intestinal receptor of the bacterial adhesin FimH, frequently occur in IBD patients. Hence, we aimed to functionally characterize GP2-directed autoantibodies as a putative part of IBD's pathophysiology. Ex vivo, GP2-splicing variant 4 (GP2#4) but not variant 2 was expressed on intestinal M or L cells with elevated expression patterns in IBD patients. The GP2#4 expression was induced in vitro by tumor necrosis factor (TNF)-α. The IBD-associated GP2 autoantibodies inhibited FimH binding to GP2#4 and were decreased in anti-TNFα-treated Crohn's disease patients with ileocolonic disease manifestation. In vivo, mice immunized against GP2 before infection with adherent-invasive bacteria displayed exacerbated intestinal inflammation. In summary, autoimmunity against intestinal expressed GP2#4 results in enhanced attachment of flagellated bacteria to the intestinal epithelium and thereby may drive IBD's pathophysiology.


Subject(s)
Autoantibodies/genetics , GPI-Linked Proteins/immunology , Immunity, Mucosal/genetics , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/immunology , Adhesins, Escherichia coli/genetics , Adult , Alternative Splicing/immunology , Animals , Colitis, Ulcerative/immunology , Crohn Disease/immunology , Female , Fimbriae Proteins/genetics , Humans , Male , Mice
10.
Mol Immunol ; 121: 59-71, 2020 05.
Article in English | MEDLINE | ID: mdl-32163759

ABSTRACT

Tristetraprolin (TTP) regulates inflammatory and immune responses by destabilizing target mRNAs via binding to their 3'-UTR AREs. We have recently reported that TTP preferentially up-regulates the expression level of innate immunity genes involved in the type I interferon-mediated signaling pathway and viral response in cancer cells. To elucidate the role of TTP-RNA interaction in TTP-mediated upregulation of gene expression, we performed iRIP-seq experiments to obtain the RNA interaction map consisting of direct and indirect binding sites of TTP in HeLa cells. We found substantial TTP binding signals in mRNA regions and the introns. ARE-motif AUUUA is over-represented in TTP binding peaks. Strikingly, AUUUA frequency is high both in 3'UTR and intronic regions, and the intronic peaks were more associated with TTP-regulated genes. Analysis of the over-represented motifs in TTP peaks revealed the high frequencies of UAGG and GUGUG motifs reported for hnRNPA2/B1 and CELF1 respectively in the 3'UTR and introns, and also the UGGAC motif overlapping with the m6A motif GGACU in the CDS regions. We further demonstrated that TTP binds to multiple intronic and exonic sites in the pre-mRNA/mRNA of the transcription factor RelB, correlating with the TTP-upregulated expression of RelB. TTP-up-regulated genes without a TTP binding site, but not those with, are highly enriched in innate immunity pathways and show higher tendency of harboring RelB binding sites in their promoter regions. These findings support a model in which TTP binding of RelB pre-mRNA/mRNA coordinates the RelB upregulation and activation of the innate immunity for antiviral response.


Subject(s)
Gene Regulatory Networks/immunology , Immunity, Innate/genetics , RNA, Messenger/metabolism , Transcription Factor RelB/metabolism , Tristetraprolin/metabolism , 3' Untranslated Regions/genetics , AU Rich Elements/genetics , Alternative Splicing/immunology , Binding Sites , Gene Expression Profiling , HeLa Cells , Humans , Introns/genetics , Promoter Regions, Genetic , RNA Precursors/genetics , RNA Precursors/metabolism , RNA Stability , RNA, Messenger/genetics , Up-Regulation
11.
Front Immunol ; 11: 624612, 2020.
Article in English | MEDLINE | ID: mdl-33679718

ABSTRACT

Extracellular matrix glycoprotein tenascin-C (TnC) is highly expressed in vertebrates during embryonic development and thereafter transiently in tissue niches undergoing extensive remodeling during regeneration after injury. TnC's different functions can be attributed to its multimodular structure represented by distinct domains and alternatively spliced isoforms. Upon central nervous system injury, TnC is upregulated and secreted into the extracellular matrix mainly by astrocytes. The goal of the present study was to elucidate the role of different TnC domains in events that take place after spinal cord injury (SCI). Astrocyte cultures prepared from TnC-deficient (TnC-/-) and wild-type (TnC+/+) mice were scratched and treated with different recombinantly generated TnC fragments. Gap closure, cell proliferation and expression of GFAP and cytokines were determined in these cultures. Gap closure in vitro was found to be delayed by TnC fragments, an effect mainly mediated by decreasing proliferation of astrocytes. The most potent effects were observed with fragments FnD, FnA and their combination. TnC-/- astrocyte cultures exhibited higher GFAP protein and mRNA expression levels, regardless of the type of fragment used for treatment. Application of TnC fragments induced also pro-inflammatory cytokine production by astrocytes in vitro. In vivo, however, the addition of FnD or Fn(D+A) led to a difference between the two genotypes, with higher levels of GFAP expression in TnC+/+ mice. FnD treatment of injured TnC-/- mice increased the density of activated microglia/macrophages in the injury region, while overall cell proliferation in the injury site was not affected. We suggest that altogether these results may explain how the reaction of astrocytes is delayed while their localization is restricted to the border of the injury site to allow microglia/macrophages to form a lesion core during the first stages of glial scar formation, as mediated by TnC and, in particular, the alternatively spliced FnD domain.


Subject(s)
Alternative Splicing/immunology , Astrocytes/immunology , Cicatrix/immunology , Spinal Cord Injuries/immunology , Tenascin/immunology , Animals , Astrocytes/pathology , Cicatrix/genetics , Cicatrix/pathology , Mice , Mice, Knockout , Protein Domains , Spinal Cord Injuries/genetics , Spinal Cord Injuries/pathology , Tenascin/genetics
12.
Theranostics ; 9(25): 7648-7665, 2019.
Article in English | MEDLINE | ID: mdl-31695792

ABSTRACT

Alternative splicing (AS) has emerged as a key event in tumor development and microenvironment formation. However, comprehensive analysis of AS and its clinical significance in head and neck squamous cell carcinoma (HNSC) is urgently required. Methods: Genome-wide profiling of AS events using RNA-Seq data from The Cancer Genome Atlas (TCGA) program was performed in a cohort of 464 patients with HNSC. Cancer-associated AS events (CASEs) were identified between paired HNSC and adjacent normal tissues and evaluated in functional enrichment analysis. Splicing networks and prognostic models were constructed using bioinformatics tools. Unsupervised clustering of the CASEs identified was conducted and associations with clinical, molecular and immune features were analyzed. Results: We detected a total of 32,309 AS events and identified 473 CASEs in HNSC; among these, 91 were validated in an independent cohort (n = 15). Functional protein domains were frequently altered, especially by CASEs affecting cancer drivers, such as PCSK5. CASE parent genes were significantly enriched in pathways related to HNSC and the tumor immune microenvironment, such as the viral carcinogenesis (FDR < 0.001), Human Papillomavirus infection (FDR < 0.001), chemokine (FDR < 0.001) and T cell receptor (FDR < 0.001) signaling pathways. CASEs enriched in immune-related pathways were closely associated with immune cell infiltration and cytolytic activity. AS regulatory networks suggested a significant association between splicing factor (SF) expression and CASEs and might be regulated by SF methylation. Eighteen CASEs were identified as independent prognostic factors for overall and disease-free survival. Unsupervised clustering analysis revealed distinct correlations between AS-based clusters and prognosis, molecular characteristics and immune features. Immunogenic features and immune subgroups cooperatively depict the immune features of AS-based clusters. Conclusion: This comprehensive genome-wide analysis of the AS landscape in HNSC revealed novel AS events related to carcinogenesis and immune microenvironment, with implications for prognosis and therapeutic responses.


Subject(s)
Alternative Splicing/genetics , Head and Neck Neoplasms/genetics , Squamous Cell Carcinoma of Head and Neck/genetics , Tumor Microenvironment/genetics , Alternative Splicing/immunology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Carcinogenesis/immunology , Carcinogenesis/pathology , Cohort Studies , Disease-Free Survival , Female , Gene Expression Profiling/methods , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/pathology , Humans , Male , Papillomavirus Infections/genetics , Papillomavirus Infections/immunology , Papillomavirus Infections/pathology , Prognosis , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/pathology , Tumor Microenvironment/immunology
13.
Proc Natl Acad Sci U S A ; 116(31): 15625-15634, 2019 07 30.
Article in English | MEDLINE | ID: mdl-31308239

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is an inflammatory condition associated with abnormal immune responses, leading to airflow obstruction. Lungs of COPD subjects show accumulation of proinflammatory T helper (Th) 1 and Th17 cells resembling that of autoreactive immune responses. As regulatory T (Treg) cells play a central role in the control of autoimmune responses and their generation and function are controlled by the adipocytokine leptin, we herein investigated the association among systemic leptin overproduction, reduced engagement of glycolysis in T cells, and reduced peripheral frequency of Treg cells in different COPD stages. These phenomena were also associated with an impaired capacity to generate inducible Treg (iTreg) cells from conventional T (Tconv) cells. At the molecular level, we found that leptin inhibited the expression of forkhead-boxP3 (FoxP3) and its splicing variants containing the exon 2 (FoxP3-E2) that correlated inversely with inflammation and weakened lung function during COPD progression. Our data reveal that the immunometabolic pathomechanism leading to COPD progression is characterized by leptin overproduction, a decline in the expression of FoxP3 splicing forms, and an impairment in Treg cell generation and function. These results have potential implications for better understanding the autoimmune-like nature of COPD and the pathogenic events leading to lung damage.


Subject(s)
Alternative Splicing/immunology , Forkhead Transcription Factors , Leptin , Pulmonary Disease, Chronic Obstructive , T-Lymphocytes, Regulatory , Female , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/immunology , Humans , Leptin/biosynthesis , Leptin/immunology , Male , Middle Aged , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/pathology , Th17 Cells/immunology , Th17 Cells/metabolism , Th17 Cells/pathology
14.
Immunology ; 157(4): 296-303, 2019 08.
Article in English | MEDLINE | ID: mdl-31162836

ABSTRACT

The characterization of the architecture, structure and extracellular interactions of the CD6 glycoprotein, a transmembrane receptor expressed in medullary thymocytes and all mature T-cell populations, has been enhanced by the existence of monoclonal antibodies (mAbs) that specifically recognize the various scavenger receptor cysteine-rich (SRCR) domains of the ectodomain. Using engineered isoforms of CD6 including or excluding each of the three SRCR domains, either expressed at the membranes of cells or in soluble forms, we provide conclusive and definitive evidence that domain 2 of CD6, previously not identifiable, can be recognized by the CD6 mAbs OX125 and OX126, and that OX124 targets domain 3 and can block the interaction at the cell surface of CD6 with its major ligand CD166. Alternative splicing-dependent CD6 isoforms can now be confidently identified. We confirm that following T-cell activation there is a partial replacement of full-length CD6 by the CD6Δd3 isoform, which lacks the CD166-binding domain, and we find no evidence for the expression of other CD6 isoforms at the mRNA or protein levels.


Subject(s)
Alternative Splicing/immunology , Antibodies, Monoclonal, Murine-Derived/chemistry , Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , Lymphocyte Activation , T-Lymphocytes/immunology , Antibodies, Monoclonal, Murine-Derived/immunology , Humans , Jurkat Cells , Protein Domains , Protein Isoforms/immunology , T-Lymphocytes/cytology
15.
Sci Rep ; 9(1): 7306, 2019 05 13.
Article in English | MEDLINE | ID: mdl-31086276

ABSTRACT

Signaling of the pleiotropic cytokine Interleukin-6 (IL-6) via its soluble IL-6R (sIL-6R) has been termed trans-signaling and is thought to be responsible for the pro-inflammatory properties of IL-6. The sIL-6R can be generated by alternative mRNA splicing or proteolytic cleavage of the membrane-bound IL-6R. However, which stimuli induce sIL-6R release and which endogenous signaling pathways are required for this process is poorly understood. Here, we show that activation of Toll-like receptor 2 (TLR2) on primary human peripheral blood mononuclear cells (PBMCs) and on the monocytic cell line THP-1 induces expression and secretion of IL-6 and the generation of sIL-6R. We show by flow cytometry that monocytes are a PBMC subset that expresses TLR2 in conjunction with the IL-6R and are the major cellular source for both IL-6 and sIL-6R. Mechanistically, we find that the metalloproteases ADAM10 and ADAM17 are responsible for cleavage of the IL-6R and therefore sIL-6R generation. Finally, we identify the Extracellular-signal Regulated Kinase (ERK) cascade as a critical pathway that differentially regulates both IL-6 and sIL-6R generation in monocytes.


Subject(s)
Interleukin-6/metabolism , MAP Kinase Signaling System/immunology , Receptors, Interleukin-6/metabolism , Sepsis/immunology , Toll-Like Receptor 2/metabolism , ADAM10 Protein/metabolism , ADAM17 Protein/metabolism , Alternative Splicing/immunology , Amyloid Precursor Protein Secretases/metabolism , Case-Control Studies , Cohort Studies , Healthy Volunteers , Humans , Interleukin-6/blood , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Membrane Proteins/metabolism , Monocytes/immunology , Monocytes/metabolism , Primary Cell Culture , RNA, Messenger/metabolism , Receptors, Interleukin-6/blood , Receptors, Interleukin-6/genetics , Sepsis/blood , THP-1 Cells
16.
Fish Shellfish Immunol ; 91: 188-193, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31077849

ABSTRACT

Columnaris disease has long been recognized as a serious problem worldwide which affects both wild and cultured freshwater fish including the commercially important channel catfish (Ictalurus punctatus). The fundamental molecular mechanisms of the host immune response to the causative agent Flavobacterium columnare remain unclear, though gene expression analysis after the bacterial infection has been conducted. Alternative splicing, a post-transcriptional regulation process to modulate gene expression and increase the proteomic diversity, has not yet been studied in channel catfish following infection with F. columnare. In this study, genomic information and RNA-Seq datasets of channel catfish were used to characterize the changes of alternative splicing after the infection. Alternative splicing was shown to be induced by F. columnare infection, with 8.0% increase in alternative splicing event at early infection stage. Intriguingly, genes involved in RNA binding and RNA splicing themselves were significantly enriched in differentially alternatively spliced (DAS) gene sets after infection. This finding was consistent with our previous study in channel catfish following infection with Edwardsiella ictaluri. It was suggested to be a universal mechanism that genes involved in RNA binding and splicing were regulated to undergo differential alternative splicing after stresses in channel catfish. Moreover, many immune genes were observed to be differentially alternatively spliced after infection. Further studies need to be performed to get a deeper view of molecular regulation on alternative splicing after stresses, setting a foundation for developing catfish broodstocks with enhanced disease resistance.


Subject(s)
Alternative Splicing/immunology , Fish Diseases/immunology , Flavobacteriaceae Infections/veterinary , Ictaluridae , Transcription, Genetic/immunology , Animals , Fish Diseases/microbiology , Flavobacteriaceae Infections/immunology , Flavobacteriaceae Infections/microbiology , Flavobacterium/physiology , Random Allocation
17.
Nat Commun ; 10(1): 1671, 2019 04 11.
Article in English | MEDLINE | ID: mdl-30975994

ABSTRACT

Host and environmental factors contribute to variation in human immune responses, yet the genetic and evolutionary drivers of alternative splicing in response to infection remain largely uncharacterised. Leveraging 970 RNA-sequencing profiles of resting and stimulated monocytes from 200 individuals of African- and European-descent, we show that immune activation elicits a marked remodelling of the isoform repertoire, while increasing the levels of erroneous splicing. We identify 1,464 loci associated with variation in isoform usage (sQTLs), 9% of them being stimulation-specific, which are enriched in disease-related loci. Furthermore, we detect a longstanding increased plasticity of immune gene splicing, and show that positive selection and Neanderthal introgression have both contributed to diversify the splicing landscape of human populations. Together, these findings suggest that differential isoform usage has been an important substrate of innovation in the long-term evolution of immune responses and a more recent vehicle of population local adaptation.


Subject(s)
Alternative Splicing/immunology , Immunity/genetics , Infections/immunology , Selection, Genetic/immunology , Transcriptome/immunology , Animals , Biological Evolution , Black People/genetics , Genetic Variation/immunology , Healthy Volunteers , Humans , Male , Neanderthals/genetics , Polymorphism, Single Nucleotide , Protein Isoforms/genetics , Protein Isoforms/immunology , Quantitative Trait Loci/immunology , Sequence Analysis, RNA , White People/genetics , Exome Sequencing
18.
Dev Comp Immunol ; 96: 135-143, 2019 07.
Article in English | MEDLINE | ID: mdl-30885554

ABSTRACT

CD44 gene is a cell surface receptor which undergoes complex alternative splicing and extensive post-translational modifications. Although many studies have showed that CD44 is involved in the process of host defense, the function of piscine CD44 in antibacterial or antiviral defense response remains unclear. In the present study, we report the functional characterization of zebrafish CD44c, which is more similar to CD44b antigen isoforms rather than CD44a based on amino acid composition and phylogenetic analysis. The expression of zebrafish CD44c was inducible in response to bacterial and viral infections. During SVCV infection, the in vivo studies revealed that CD44c overexpression led to the increased virus loads and decreased survival rate. The attenuated response by zebrafish CD44c in response to SVCV infection were characterized by the impaired production of inflammatory cytokines and the impaired expressions of IFNs, IFN-stimulated genes, MHC class I and II genes. During Edwardsiella piscicida infection, the overexpression of zebrafish CD44c facilitated bacterial growth and dissemination, but did not impact on larvae survival. The detrimental role of CD44c in host defense against E. piscicida infection was supported by a decreased production of several antibacterial molecules including defbl2, defbl3, NK-lysin and RNase3. All together, these results firstly demonstrate the negative regulation of piscine CD44c in viral and bacterial infection.


Subject(s)
Enterobacteriaceae Infections/immunology , Fish Diseases/immunology , Hyaluronan Receptors/immunology , Rhabdoviridae Infections/immunology , Zebrafish/immunology , Alternative Splicing/immunology , Amino Acid Sequence , Animals , Cell Line , Cytokines/immunology , Cytokines/metabolism , Edwardsiella/immunology , Edwardsiella/pathogenicity , Enterobacteriaceae Infections/microbiology , Fish Diseases/microbiology , Hyaluronan Receptors/chemistry , Hyaluronan Receptors/genetics , Immunity, Innate , Larva/immunology , Larva/metabolism , Phylogeny , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/immunology , Rhabdoviridae/immunology , Rhabdoviridae/pathogenicity , Rhabdoviridae Infections/virology , Structure-Activity Relationship , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish/virology
19.
PLoS One ; 14(1): e0210193, 2019.
Article in English | MEDLINE | ID: mdl-30629639

ABSTRACT

The human natural killer-1 (HNK-1) carbohydrate epitope, composed of a unique sulfated trisaccharide (HSO3-3GlcAß1-3Galß1-4GlcNAc-R), is highly expressed during brain development and regulates higher brain function. However, it remains unclear which glycoprotein carries the HNK-1 epitope in the embryonic brain and the functional role it plays. Here, we showed that one of the major HNK-1 carrier proteins in the embryonic brain is tenascin-C (TNC), an extracellular matrix protein that regulates neurite outgrowth by interacting with the GPI-anchored protein contactin-1 (CNTN). Because the alternatively spliced fibronectin type-III (FNIII) repeats in TNC give rise to many isoforms and affect neuronal function, we evaluated neurite outgrowth of primary hippocampal neurons on purified recombinant FNIII repeats with or without the HNK-1 epitope as a substrate. We found that the presence of the HNK-1 epitope on the C domain of TNC promoted neurite outgrowth, and that this signal was mediated by CNTN, which is an HNK-1-expressing neuronal receptor. The neurite-promoting activity of the HNK-1 epitope on TNC required neuronal HNK-1 expression, which was defective in neurons lacking the glucuronyltransferases GlcAT-P and GlcAT-S. These results suggest that the HNK-1 epitope is a key modifier of TNC and CNTN in the regulation of embryonic brain development.


Subject(s)
CD57 Antigens/immunology , Contactin 1/physiology , Hippocampus/growth & development , Neuronal Outgrowth/immunology , Tenascin/immunology , Alternative Splicing/immunology , Animals , Embryo, Mammalian , Epitopes/immunology , Fibronectin Type III Domain/genetics , Fibronectin Type III Domain/immunology , Glucuronosyltransferase/genetics , HEK293 Cells , Hippocampus/cytology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurites/physiology , Neuronal Outgrowth/genetics , Primary Cell Culture , Tenascin/genetics
20.
J Virol ; 93(7)2019 04 01.
Article in English | MEDLINE | ID: mdl-30651364

ABSTRACT

Influenza A viruses (IAV) are known to modulate and "hijack" several cellular host mechanisms, including gene splicing and RNA maturation machineries. These modulations alter host cellular responses and enable an optimal expression of viral products throughout infection. The interplay between the host protein p53 and IAV, in particular through the viral nonstructural protein NS1, has been shown to be supportive for IAV replication. However, it remains unknown whether alternatively spliced isoforms of p53, known to modulate p53 transcriptional activity, are affected by IAV infection and contribute to IAV replication. Using a TP53 minigene, which mimics intron 9 alternative splicing, we have shown here that the NS1 protein of IAV changes the expression pattern of p53 isoforms. Our results demonstrate that CPSF4 (cellular protein cleavage and polyadenylation specificity factor 4) independently and the interaction between NS1 and CPSF4 modulate the alternative splicing of TP53 transcripts, which may result in the differential activation of p53-responsive genes. Finally, we report that CPSF4 and most likely beta and gamma spliced p53 isoforms affect both viral replication and IAV-associated type I interferon secretion. All together, our data show that cellular p53 and CPSF4 factors, both interacting with viral NS1, have a crucial role during IAV replication that allows IAV to interact with and alter the expression of alternatively spliced p53 isoforms in order to regulate the cellular innate response, especially via type I interferon secretion, and perform efficient viral replication.IMPORTANCE Influenza A viruses (IAV) constitute a major public health issue, causing illness and death in high-risk populations during seasonal epidemics or pandemics. IAV are known to modulate cellular pathways to promote their replication and avoid immune restriction via the targeting of several cellular proteins. One of these proteins, p53, is a master regulator involved in a large panel of biological processes, including cell cycle arrest, apoptosis, or senescence. This "cellular gatekeeper" is also involved in the control of viral infections, and viruses have developed a wide diversity of mechanisms to modulate/hijack p53 functions to achieve an optimal replication in their hosts. Our group and others have previously shown that p53 activity is finely modulated by different multilevel mechanisms during IAV infection. Here, we characterized IAV nonstructural protein NS1 and the cellular factor CPSF4 as major partners involved in the IAV-induced modulation of the TP53 alternative splicing that was associated with a strong modulation of p53 activity and notably the p53-mediated antiviral response.


Subject(s)
Influenza A virus/immunology , Influenza, Human/immunology , Tumor Suppressor Protein p53/immunology , Viral Nonstructural Proteins/immunology , mRNA Cleavage and Polyadenylation Factors/immunology , A549 Cells , Alternative Splicing/immunology , Cell Line, Tumor , Humans , Immunity, Innate/immunology , Influenza, Human/virology , Interferons/immunology , Virus Replication/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...