1,953 results on '"Interferons genetics"'
Search Results
2. SARS-CoV-2 variants induce increased inflammatory gene expression but reduced interferon responses and heme synthesis as compared with wild type strains.
- Author
-
Merchant M, Ashraf J, Masood KI, Yameen M, Hussain R, Nasir A, and Hasan Z
- Subjects
- Humans, Interferons metabolism, Interferons genetics, Inflammation genetics, Inflammation virology, Gene Expression Profiling, Transcriptome, Male, Female, COVID-19 genetics, COVID-19 virology, COVID-19 immunology, SARS-CoV-2 genetics, Heme biosynthesis, Heme metabolism
- Abstract
SARS-CoV-2 variants of concern (VOC) have been associated with increased viral transmission and disease severity. We investigated the mechanisms of pathogenesis caused by variants using a host blood transcriptome profiling approach. We analysed transcriptional signatures of COVID-19 patients comparing those infected with wildtype (wt), alpha, delta or omicron strains seeking insights into infection in Asymptomatic cases.Comparison of transcriptional profiles of Symptomatic and Asymptomatic COVID-19 cases showed increased differentially regulated gene (DEGs) of inflammatory, apoptosis and blood coagulation pathways, with decreased T cell and Interferon stimulated genes (ISG) activation. Between SARS-CoV-2 strains, an increasing number of DEGs occurred in comparisons between wt and alpha (196), delta (1425) or, omicron (2313) infections. COVID-19 cases with alpha or, delta variants demonstrated suppression transcripts of innate immune pathways. EGR1 and CXCL8 were highly upregulated in those infected with VOC; heme biosynthetic pathway genes (ALAS2, HBB, HBG1, HBD9) and ISGs were downregulated. Delta and omicron infections upregulated ribosomal pathways, reflecting increased viral RNA translation. Asymptomatic COVID-19 cases infected with delta infections showed increased cytokines and ISGs expression. Overall, increased inflammation, with reduced host heme synthesis was associated with infections caused by VOC infections, with raised type I interferon in cases with less severe disease., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
3. Engineered chimeric receptors for dissecting interferon signaling.
- Author
-
Lin AE, Mesev EV, Toettcher JE, and Ploss A
- Subjects
- Humans, Animals, Receptors, Erythropoietin metabolism, Receptors, Erythropoietin genetics, Receptors, Interferon metabolism, Receptors, Interferon genetics, Protein Engineering, Signal Transduction, Interferons metabolism, Interferons genetics
- Abstract
Though interferons (IFNs) were once heralded as panaceas to numerous diseases, how cells decode varying IFN stimuli and subsequently produce (in)appropriate signaling remain unclear. Our labs recently engineered novel erythropoietin receptor-IFN chimeric receptors, and we highlight their utility in two cases uncovering differential genetic determinants of type I (IFN-α/β) and type III (IFN-λ) IFN signaling. These and other types of synthetic (cytokine) receptors could be expanded to real-time signaling dynamics and in vivo studies., Competing Interests: The authors declare no conflict of interest.
- Published
- 2024
- Full Text
- View/download PDF
4. Interference without interferon: interferon-independent induction of interferon-stimulated genes and its role in cellular innate immunity.
- Author
-
Swaraj S and Tripathi S
- Subjects
- Humans, Animals, Virus Diseases immunology, Virus Diseases genetics, Janus Kinases genetics, Janus Kinases metabolism, Janus Kinases immunology, STAT Transcription Factors genetics, STAT Transcription Factors metabolism, STAT Transcription Factors immunology, Immunity, Innate, Interferons immunology, Interferons genetics, Interferons metabolism, Signal Transduction
- Abstract
Interferons (IFNs) are multifaceted proteins that play pivotal roles in orchestrating robust antiviral immune responses and modulating the intricate landscape of host immunity. The major signaling pathway activated by IFNs is the JAK/STAT (Janus kinase/signal transducer and activator of transcription) pathway, which leads to the transcription of a battery of genes, collectively known as IFN-stimulated genes (ISGs). While the well-established role of IFNs in coordinating the innate immune response against viral infections is widely acknowledged, recent years have provided a more distinct comprehension of the functional significance attributed to non-canonical, IFN-independent induction of ISGs. In this review, we summarize the non-conventional signaling pathways of ISG induction. These alternative pathways offer new avenues for developing antiviral strategies or immunomodulation in various diseases., Competing Interests: The authors declare no conflict of interest.
- Published
- 2024
- Full Text
- View/download PDF
5. FBXL19 in endothelial cells protects the heart from influenza A infection by enhancing antiviral immunity and reducing cellular senescence programs.
- Author
-
Xia B, Chen H, Taleb SJ, Xi X, Shaheen N, Baoyinna B, Soni S, Mebratu YA, Yount JS, Zhao J, and Zhao Y
- Subjects
- Animals, Mice, Inbred C57BL, Mice, F-Box Proteins metabolism, F-Box Proteins genetics, Humans, Influenza A virus pathogenicity, Myocardium metabolism, Myocardium immunology, Myocardium pathology, Disease Models, Animal, Signal Transduction, Interferons metabolism, Interferons genetics, Male, Chemokine CCL5, Cellular Senescence, Orthomyxoviridae Infections immunology, Orthomyxoviridae Infections metabolism, Endothelial Cells metabolism, Endothelial Cells immunology, Endothelial Cells virology, Interferon Regulatory Factor-3 metabolism, Interferon Regulatory Factor-3 genetics
- Abstract
Influenza A virus (IAV) infection while primarily affecting the lungs, is often associated with cardiovascular complications. However, the mechanisms underlying this association are not fully understood. Here, we investigated the potential role of FBXL19, a member of the Skp1-Cullin-1-F-box family of E3 ubiquitin ligase, in IAV-induced cardiac inflammation. We demonstrated that FBXL19 overexpression in endothelial cells (ECs) reduced viral titers and IAV matrix protein 1 (M1) levels while increasing antiviral gene expression, including interferon (IFN)-α, -β, and -γ and RANTES (regulated on activation normal T cell expressed and secreted) in the cardiac tissue of IAV-infected mice. Moreover, EC-specific overexpression of FBXL19 attenuated the IAV infection-reduced interferon regulatory factor 3 (IRF3) level without altering its mRNA level and suppressed cardiac inflammation. Furthermore, IAV infection triggered cellular senescence programs in the heart as indicated by the upregulation of p16 and p21 mRNA levels and the downregulation of lamin-B1 levels, which were partially reversed by FBXL19 overexpression in ECs. Our findings indicate that EC-specific overexpression of FBXL19 protects against IAV-induced cardiac damage by enhancing interferon-mediated antiviral signaling, reducing cardiac inflammation, and suppressing cellular senescence programs. NEW & NOTEWORTHY Our study reveals a novel facet of IAV infection, demonstrating that it can trigger cellular senescence within the heart. Intriguingly, upregulation of endothelial FBXL19 promotes host innate immunity, reduces cardiac senescence, and diminishes inflammation. These findings highlight the therapeutic potential of targeting FBXL19 to mitigate IAV-induced cardiovascular complications.
- Published
- 2024
- Full Text
- View/download PDF
6. CgIκB2 negatively regulates the expression of interferon-like protein by Rel/NF-κB signal in Crassostrea gigas.
- Author
-
Niu J, Wang S, Qiao X, Yu S, Yu Z, Jin Y, Huang M, Wang L, and Song L
- Subjects
- Animals, Immunity, Innate genetics, I-kappa B Proteins genetics, I-kappa B Proteins metabolism, Hemocytes immunology, Hemocytes metabolism, Crassostrea genetics, Crassostrea immunology, Signal Transduction, Poly I-C pharmacology, NF-kappa B genetics, NF-kappa B metabolism, Gene Expression Regulation immunology, Interferons genetics, Interferons immunology, Interferons metabolism
- Abstract
Inhibitors of NF-κB (IκBs) have been implicated as major components of the Rel/NF-κB signaling pathway, playing an important negative regulatory role in host antiviral immunity such as in the activation of interferon (IFN) in vertebrates. In the present study, the immunomodulatory effect of IκB (CgIκB2) on the expression of interferon-like protein (CgIFNLP) was evaluated in Pacific oyster (Crassostrea gigas). After poly (I:C) stimulation, the mRNA expression level of CgIκB2 in haemocytes was significantly down-regulated at 3-12 h while up-regulated at 48-72 h. The mRNA expression of CgIκB2 in haemocytes was significantly up-regulated at 3 h after rCgIFNLP stimulation. In the CgIκB2-RNAi oysters, the mRNA expression of CgIFNLP, interferon regulatory factor-8 (CgIRF8) and NF-κB subunit (CgRel), the abundance of CgIFNLP and CgIRF8 protein in haemocytes, as well as the abundance of CgRel protein in nucleus were significantly increased after poly (I:C) stimulation. Immunofluorescence assay showed that nuclear translocation of CgIRF8 and CgRel protein was promoted in CgIκB2-RNAi oysters compared with that in EGFP-RNAi group. In the CgRel-RNAi oysters, the mRNA and protein expression level of CgIFNLP significantly down-regulated after poly (I:C) stimulation. The collective results indicated that CgIκB2 plays an important role in regulating CgIFNLP expression through its effects on Rel/NF-κB and IRF signaling pathways., Competing Interests: Declaration of competing interest The authors declare that they have no conflict of interests., (Copyright © 2024 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
7. Bioinformatic-Experimental Screening Uncovers Multiple Targets for Increase of MHC-I Expression through Activating the Interferon Response in Breast Cancer.
- Author
-
Li X, Ruan Z, Yang S, Yang Q, Li J, and Hu M
- Subjects
- Humans, Female, Interferons metabolism, Interferons genetics, Histocompatibility Antigens Class I genetics, Histocompatibility Antigens Class I metabolism, Cell Line, Tumor, Prognosis, Gene Expression Profiling, Breast Neoplasms genetics, Breast Neoplasms immunology, Breast Neoplasms metabolism, Breast Neoplasms pathology, Computational Biology methods, Gene Expression Regulation, Neoplastic, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating metabolism
- Abstract
Expression of major histocompatibility complex I (MHC-I) on tumor cells is extremely important for the antitumor immune response for its essential role in activating various immune cells, including tumor-specific CD8+ T cells. Cancers of lower MHC-I expression commonly exhibit less immune cell infiltration and worse prognosis in clinic. In this study, we conducted bioinformatic-experimental screening to identify potential gene targets to enhance MHC-I expression in breast cancer (BRCA). Through a combination of MHC-I scoring, gene expression correlation analysis, survival prognostication, and Cibersort tumor-infiltrated lymphocytes (TILs) scoring, we identify 144 genes negatively correlated with both MHC-I expression and TILs in breast cancer. Furthermore, we verified partially according to KEGG functional enrichment or gene-dependency analysis and figured out multiple genes, including PIP5K1A , NCKAP1 , CYFIP1 , DIS3 , TBP , and EXOC1 , as effective gene targets for increasing MHC-I expression in breast cancer. Mechanistically, knockout of each of these genes activated the intrinsic interferon response in breast cancer cells, which not only promoted MHC-I expression but also caused immunogenic cell death of breast cancer. Finally, the scRNA-seq confirmed the negative correlation of PIP5K1A et al. with TILs in breast cancer patients. Collectively, we identified multiple gene targets for an increase in MHC-I expression in breast cancer in this study.
- Published
- 2024
- Full Text
- View/download PDF
8. Research Progress into the Biological Functions of IFITM3.
- Author
-
Xie Q, Wang L, Liao X, Huang B, Luo C, Liao G, Yuan L, Liu X, Luo H, and Shu Y
- Subjects
- Humans, Animals, Signal Transduction, Interferons metabolism, Interferons immunology, Interferons genetics, Membrane Proteins metabolism, Membrane Proteins genetics, RNA-Binding Proteins metabolism, RNA-Binding Proteins genetics
- Abstract
Interferon-induced transmembrane proteins (IFITMs) are upregulated by interferons. They are not only highly conserved in evolution but also structurally consistent and have almost identical structural domains and functional domains. They are all transmembrane proteins and have multiple heritable variations in genes. The IFITM protein family is closely related to a variety of biological functions, including antiviral immunity, tumor formation, bone metabolism, cell adhesion, differentiation, and intracellular signal transduction. The progress of the research on its structure and related functions, as represented by IFITM3, is reviewed.
- Published
- 2024
- Full Text
- View/download PDF
9. A genome-wide arrayed CRISPR screen identifies PLSCR1 as an intrinsic barrier to SARS-CoV-2 entry that recent virus variants have evolved to resist.
- Author
-
Le Pen J, Paniccia G, Kinast V, Moncada-Velez M, Ashbrook AW, Bauer M, Hoffmann HH, Pinharanda A, Ricardo-Lax I, Stenzel AF, Rosado-Olivieri EA, Dinnon KH 3rd, Doyle WC, Freije CA, Hong SH, Lee D, Lewy T, Luna JM, Peace A, Schmidt C, Schneider WM, Winkler R, Yip EZ, Larson C, McGinn T, Menezes MR, Ramos-Espiritu L, Banerjee P, Poirier JT, Sànchez-Rivera FJ, Cobat A, Zhang Q, Casanova JL, Carroll TS, Glickman JF, Michailidis E, Razooky B, MacDonald MR, and Rice CM
- Subjects
- Humans, HEK293 Cells, CRISPR-Cas Systems genetics, Spike Glycoprotein, Coronavirus genetics, Spike Glycoprotein, Coronavirus metabolism, Serine Endopeptidases genetics, Serine Endopeptidases metabolism, Interferons metabolism, Interferons genetics, Clustered Regularly Interspaced Short Palindromic Repeats genetics, Antigens, Differentiation, SARS-CoV-2 genetics, Virus Internalization, COVID-19 virology, COVID-19 genetics
- Abstract
Interferons (IFNs) play a crucial role in the regulation and evolution of host-virus interactions. Here, we conducted a genome-wide arrayed CRISPR knockout screen in the presence and absence of IFN to identify human genes that influence Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection. We then performed an integrated analysis of genes interacting with SARS-CoV-2, drawing from a selection of 67 large-scale studies, including our own. We identified 28 genes of high relevance in both human genetic studies of Coronavirus Disease 2019 (COVID-19) patients and functional genetic screens in cell culture, with many related to the IFN pathway. Among these was the IFN-stimulated gene PLSCR1. PLSCR1 did not require IFN induction to restrict SARS-CoV-2 and did not contribute to IFN signaling. Instead, PLSCR1 specifically restricted spike-mediated SARS-CoV-2 entry. The PLSCR1-mediated restriction was alleviated by TMPRSS2 overexpression, suggesting that PLSCR1 primarily restricts the endocytic entry route. In addition, recent SARS-CoV-2 variants have adapted to circumvent the PLSCR1 barrier via currently undetermined mechanisms. Finally, we investigate the functional effects of PLSCR1 variants present in humans and discuss an association between PLSCR1 and severe COVID-19 reported recently., Competing Interests: The authors have declared that no competing interests exist., (Copyright: © 2024 Le Pen et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2024
- Full Text
- View/download PDF
10. Protocol for linking enhanced interferon immunity to virus resistance in gene-knockout zebrafish.
- Author
-
Qu ZL and Zhang YB
- Subjects
- Animals, Microinjections methods, Disease Resistance genetics, Disease Resistance immunology, Larva virology, Larva immunology, Larva genetics, Plasmids genetics, Zebrafish genetics, Gene Knockout Techniques methods, Interferons genetics, Interferons metabolism, Interferons immunology
- Abstract
A gene-rescue experiment under a mutant background is essential to clarify gene function and the resulting biological potential in vivo. Here, we present a protocol for determining the change in interferon response by microinjecting plasmids into one-cell-stage zebrafish embryos. We describe steps for comparing the resistance potential to virus infection in wild-type and knockout zebrafish larvae following plasmid microinjection. We then detail how to link the enhanced interferon immunity to the improved resistance in knockout zebrafish larvae by gene-rescue experiments. For complete details on the use and execution of this protocol, please refer to Qu et al.
1 ., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
11. Anti-infective immune functions of type IV interferon in grass carp ( Ctenopharyngodon idella ): A novel antibacterial and antiviral interferon in lower vertebrates.
- Author
-
Liu Y, Zhu W, Zhang Y, Zhang J, Lv M, and Su J
- Subjects
- Animals, Antiviral Agents pharmacology, Gene Expression Regulation drug effects, Interferons metabolism, Interferons genetics, Anti-Bacterial Agents pharmacology, Bacterial Infections veterinary, Bacterial Infections immunology, Fish Proteins genetics, Fish Proteins metabolism, Amino Acid Sequence, Phylogeny, Carps immunology, Fish Diseases immunology, Fish Diseases virology
- Abstract
Type IV interferon (IFN-υ) is a recently discovered cytokine crucial for host defense against viral infections. However, the role and mechanisms of IFN-υ in bacterial infections remain unexplored. This study investigated the antibacterial and antiviral functions and mechanisms of grass carp ( Ctenopharyngodon idella ) IFN-υ (CiIFN-υ) both in vivo and in vitro . The CiIFN-υ gene was first identified and characterized in grass carp. Subsequently, the immune expression of CiIFN-υ significantly increased following bacterial challenge, indicating its response to bacterial infections. The eukaryotic recombinant expression plasmid of CiIFN-υ was then constructed and transfected into fathead minnow (FHM) cells. Supernatants were collected and incubated with four bacterial strains, followed by plate spreading and colony counting. Results indicated that CiIFN-υ exhibited more potent antibacterial activity against gram-negative bacteria compared to gram-positive bacteria and aggregated gram-negative bacteria but not gram-positive bacteria. In vivo experiments further confirmed the antibacterial function, showing high survival rates, low tissue edema and damage, reduced tissue bacterial load, and elevated proinflammatory response at the early stages of bacterial infection. In addition, the antiviral function of CiIFN-υ was confirmed through in vitro and in vivo experiments, including crystal violet staining, survival rates, tissue viral burden, and RT-qPCR. This study highlights the antibacterial function and preliminary mechanism of IFN-υ, demonstrating that IFN-υ possesses dual functions against bacterial and viral infections.
- Published
- 2024
- Full Text
- View/download PDF
12. Molecular similarities between the genes for Trypanosoma cruzi microtubule-associated proteins, mammalian interferons, and TRIMs.
- Author
-
Winkler MA and Pan AA
- Subjects
- Humans, Animals, Protozoan Proteins genetics, Interferons genetics, Computational Biology, Molecular Sequence Data, Sequence Homology, Amino Acid, Tripartite Motif Proteins genetics, Trypanosoma cruzi genetics, Trypanosoma cruzi immunology
- Abstract
Initial studies using bioinformatics analysis revealed DNA sequence similarities between Trypanosoma cruzi GenBank® M21331, coding for Antigen 36 (Ag 36), and tripartite motif (TRIM) genes. TRIM40 showed 9.7% identity to GenBank M21331, and four additional TRIM genes had identities greater than 5.0%. TRIM37 showed a continuous stretch of identity of 12 nucleotides, that is, at least 25% longer than any of the other TRIMs. When we extended our analysis on the relationships of GenBank M21331 to further innate immune genes, using the Needleman-Wunsch (NW) algorithm for alignment, identities to human IFN-α, IFN-β, and IFN-γ genes of 13.6%, 12.6%, and 17.9%, respectively, were found. To determine the minimum number of genes coding for proteins closely related to Ag 36, a BLAST-p search was conducted with it versus the T. cruzi genome. The BLAST-p search revealed that T. cruzi GenBank M21331 had 14 gene sequences homologous to microtubule-associated protein (MAP) genes with 100% amino acid sequence identity. To verify the similarities in non-human genes, a study comparing TRIM21 region sequences among mammalian species to the comparable human TRIM21 region showed that related sequences were also present in 11 mammalian species. The MAP genes homologous to Ag 36 form a family of at least 14 genes which mimic human immune genes in the IFN and TRIM families. This mimicry is of gene sequences and not their protein products or epitopes. These results appear to be the first description of molecular mimicry of immune genes in humans by a protozoan parasite., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
13. The history of interferon-stimulated genes in pregnant cattle, sheep, and pigs.
- Author
-
Johnson GA, Bazer FW, Burghardt RC, Seo H, Wu G, Cain JW, and Pohler KG
- Subjects
- Animals, Female, Pregnancy, Cattle, Sheep, Swine, Endometrium metabolism, Interferons metabolism, Interferons genetics
- Abstract
In Brief: The trophectoderm of the elongating conceptuses of cattle, sheep, and pigs secrete high amounts of interferons that increase or induce the expression of interferon-stimulated genes (ISGs) in the endometrium. Research concerning ISGs, performed from 1995 through 2023, is reviewed in this manuscript., Abstract: Expression of the classical interferon (IFN) stimulated genes (ISGs) increases in the endometrial stroma and glandular epithelium (GE) through activation of signal transducer and activator of transcription (STAT) signaling in response to the secretion of IFN tau (IFNT) and IFN gamma (IFNG) by the conceptuses of ruminants, including cattle and sheep, and pigs, respectively. The first of the classical ISGs to be characterized was ISG15 in cattle. Classical ISGs are not expressed by the endometrial luminal epithelium (LE) due to the expression of interferon regulatory factor 2 (IRF2) in the LE that prevents the expression of ISGs in the LE. Classical ISG expression in the endometrium serves as a reliable indicator of conceptus health and elongation in cattle. There are also nonclassical ISGs that are upregulated in endometrial LE in response to progesterone (P4) that are further stimulated by IFNT in sheep, the intracellular signaling pathway responsible for IFN effects on expression is unknown. ISGs are also upregulated in extrauterine tissues including CL and peripheral blood mononuclear cells (PBMCs). The expression of ISGs by the PBMCs of cattle serves as an early prognosticator of pregnancy. The physiological roles of ISGs remain obscure, but evidence suggests that they are at least in part involved in modifying the immune system to support endometrial remodeling necessary for the successful implantation of the conceptus. Our understanding of these ISGs is primarily the result of work from the laboratories of Drs Fuller Bazer, Thomas (Tod) Hansen, Gregory Johnson, Hakhyun Ka, Patrick Lonergan, Troy Ott, and Thomas Spencer.
- Published
- 2024
- Full Text
- View/download PDF
14. Multi-omics landscape of Interferon-stimulated gene OASL reveals a potential biomarker in pan-cancer: from prognosis to tumor microenvironment.
- Author
-
Liu Y, Yang R, Zhang M, Yang B, Du Y, Feng H, Wang W, Xue B, Niu F, and He P
- Subjects
- Humans, Prognosis, Cell Line, Tumor, Gene Expression Regulation, Neoplastic, Neoplasms immunology, Neoplasms genetics, 2',5'-Oligoadenylate Synthetase genetics, 2',5'-Oligoadenylate Synthetase metabolism, Interferons metabolism, Interferons genetics, Gene Expression Profiling, B7-H1 Antigen genetics, B7-H1 Antigen metabolism, Pancreatic Neoplasms immunology, Pancreatic Neoplasms genetics, Pancreatic Neoplasms pathology, Multiomics, Tumor Microenvironment immunology, Biomarkers, Tumor genetics
- Abstract
Background: OASL (Oligoadenylate Synthetase-Like), an interferon-induced protein in the OAS family, plays a significant role in anti-viral response. Studies have demonstrated its association with prognosis of certain tumors. However, the mechanism through which OASL affects tumors is unclear. A systemic pan-cancer study of OASL needs to be illustrated., Methods: Analysis of OASL expression across 33 tumors was conducted utilizing TCGA, GTEx and CPTAC databases. COX and Log-Rank regressions were employed to calculate the prognosis. We validated the impact of OASL on apoptosis, migration, and invasion in pancreatic cancer cell lines. Moreover, we employed seven algorithms in bulk data to investigate the association of OASL expression and immune cell infiltration within tumor immune microenvironment (TIME) and ultimately validated at single-cell transcriptome level., Results: We discovered elevated expression of OASL and its genetic heterogeneity in certain tumors, which link closely to prognosis. Validation experiments were conducted in PAAD and confirmed these findings. Additionally, OASL regulates immune checkpoint ligand such as programmed death ligand 1 (PD-L1), through IFN-γ/STAT1 and IL-6/JAK/STAT3 pathways in tumor cells. Meanwhile, OASL affects macrophages infiltration in TIME. By these mechanisms OASL could cause dysfunction of cytotoxic T lymphocytes (CTLs) in tumors., Discussion: Multi-omics analysis reveals OASL as a prognostic and immunological biomarker in pan-cancer., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2024 Liu, Yang, Zhang, Yang, Du, Feng, Wang, Xue, Niu and He.)
- Published
- 2024
- Full Text
- View/download PDF
15. A novel STING variant triggers endothelial toxicity and SAVI disease.
- Author
-
Valeri E, Breggion S, Barzaghi F, Abou Alezz M, Crivicich G, Pagani I, Forneris F, Sartirana C, Costantini M, Costi S, Marino A, Chiarotto E, Colavito D, Cimaz R, Merelli I, Vicenzi E, Aiuti A, and Kajaste-Rudnitski A
- Subjects
- Humans, Infant, Gain of Function Mutation, Golgi Apparatus metabolism, Interferons metabolism, Interferons genetics, Lung Diseases, Interstitial genetics, Lung Diseases, Interstitial pathology, Lung Diseases, Interstitial immunology, Mutation, Signal Transduction, Vascular Diseases genetics, Vascular Diseases pathology, Infant, Newborn, Child, Preschool, Female, Endothelial Cells metabolism, Endothelial Cells pathology, Membrane Proteins genetics, Membrane Proteins metabolism
- Abstract
Gain-of-function mutations in STING cause STING-associated vasculopathy with onset in infancy (SAVI) characterized by early-onset systemic inflammation, skin vasculopathy, and interstitial lung disease. Here, we report and characterize a novel STING variant (F269S) identified in a SAVI patient. Single-cell transcriptomics of patient bone marrow revealed spontaneous activation of interferon (IFN) and inflammatory pathways across cell types and a striking prevalence of circulating naïve T cells was observed. Inducible STING F269S expression conferred enhanced signaling through ligand-independent translocation of the protein to the Golgi, protecting cells from viral infections but preventing their efficient immune priming. Additionally, endothelial cell activation was promoted and further exacerbated by cytokine secretion by SAVI immune cells, resulting in inflammation and endothelial damage. Our findings identify STING F269S mutation as a novel pathogenic variant causing SAVI, highlight the importance of the crosstalk between endothelial and immune cells in the context of lung disease, and contribute to a better understanding of how aberrant STING activation can cause pathology., (© 2024 Valeri et al.)
- Published
- 2024
- Full Text
- View/download PDF
16. Evaluation of the effect of sustained-release progesterone injection on the expression of interferon-related genes in repeat-breeder dairy cows.
- Author
-
Modaresi J, Kadivar A, Esfandabadi NS, Khosravian P, and Mohebbi A
- Subjects
- Animals, Cattle physiology, Female, Interferons genetics, Interferons metabolism, Gene Expression drug effects, Insemination, Artificial veterinary, Pregnancy, Injections, Intramuscular veterinary, Progesterone administration & dosage, Progesterone blood, Delayed-Action Preparations
- Abstract
Background: Repeat-breeder cows repeatedly fail to conceive after at least three attempts and return to oestrus at apparently normal intervals. Repeat-breeder cows cause economic losses in dairy farms in different ways., Objective: In the present study, we investigated the effect of sustained-release progesterone injection in two different doses on the expression of interferon-related genes in repeat-breeder dairy cows., Methods: A total of 96 repeat-breeder primiparous and multiparous cows were assigned among three groups: control group, inseminated and do not receive progesterone treatment; P
400 and P600 groups, inseminated and received a single-intramuscular injection of 400 and 600 mg slow-release progesterone 5 days after insemination, respectively. Blood sampling was carried out on Day 20 after AI for progesterone measurement and evaluation of gene expression for ISG15, MX1 and MX2 genes., Results: One injection of sustained-release progesterone increased the expression of ISG15, MX1 and MX2 genes with differences between two different progesterone concentrations. For all three genes, the level of gene expression was higher in progesterone-supplemented group than in control group, when P400 and P600 groups considered together. The level of MX2 gene expression was significantly higher in pregnant cows than non-pregnant cows. There was a significant positive correlation between expression level of all three genes and blood progesterone concentration. The expression level of ISG15 gene showed a significant positive correlation with MX1 and MX2 gene expression., Conclusion: The use of this sustained-release progesterone is simple and can be used in repeat-breeder cows to improve fertility., (© 2024 The Author(s). Veterinary Medicine and Science published by John Wiley & Sons Ltd.)- Published
- 2024
- Full Text
- View/download PDF
17. Black carp A20 inhibits interferon signaling through de-ubiquitinating IKKβ.
- Author
-
Yang X, Xie L, Yin Y, Yang C, Xiao J, Wu H, Wang C, Tian Y, and Feng H
- Subjects
- Animals, Interferons genetics, Interferons immunology, Interferons metabolism, Fish Diseases immunology, Tumor Necrosis Factor alpha-Induced Protein 3 genetics, Tumor Necrosis Factor alpha-Induced Protein 3 immunology, Immunity, Innate genetics, Gene Expression Regulation immunology, Sequence Alignment veterinary, Phylogeny, Gene Expression Profiling veterinary, Amino Acid Sequence, I-kappa B Kinase genetics, I-kappa B Kinase immunology, I-kappa B Kinase metabolism, Fish Proteins genetics, Fish Proteins immunology, Fish Proteins chemistry, Carps immunology, Carps genetics, Signal Transduction immunology, Ubiquitination
- Abstract
IkappaB kinase beta (IKKβ) is a key member of IκB kinases and functions importantly in interferon (IFN) signaling. Phosphorylation and ubiquitination are involved in the activation of IKKβ. A20 is a de-ubiquitin enzyme and functions as a suppressor in inflammation signaling, which has been reported to be phosphorylated and activated by IKKβ. However, the role and relationship of IKKβ and A20 in teleost remains unclear. In this study, IKKβ (bcIKKβ) and A20 (bcA20) of black carp (Mylopharyngodon piceus) have been cloned and characterized. Overexpressed bcIKKβ in EPC cells showed strong anti-viral ability by activating both NF-κB and IFN signaling. EPC cells stable expressing bcIKKβ presented improved anti-viral activity as well. The interaction between bcA20 and bcIKKβ was identified, and overexpression of bcA20 was able to suppress bcIKKβ-mediated activation of NF-κB and IFN signaling. Meanwhile, knock-down of A20 increased host the antiviral ability of host cells. Importantly, it has been identified that bcA20 was able to remove K27-linked ubiquitination and decrease the phosphorylation of bcIKKβ. Thus, our data conclude that bcA20 suppresses the anti-viral activity of bcIKKβ and removes its K27-linked ubiquitination, which presents a new mechanism of IKKβ regulation., Competing Interests: Declaration of competing interest The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2024 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
18. Dissecting transposable elements and endogenous retroviruses upregulation by HDAC inhibitors in leiomyosarcoma cells: Implications for the interferon response.
- Author
-
Gualandi N, Minisini M, Bertozzo A, and Brancolini C
- Subjects
- Humans, Cell Line, Tumor, Histones metabolism, Endogenous Retroviruses genetics, Histone Deacetylase Inhibitors pharmacology, Leiomyosarcoma genetics, Leiomyosarcoma virology, Leiomyosarcoma drug therapy, Leiomyosarcoma metabolism, DNA Transposable Elements, Up-Regulation, Interferons genetics, Interferons metabolism
- Abstract
Transposable elements (TEs) are of interest as immunomodulators for cancer therapies. TEs can fold into dsRNAs that trigger the interferon response. Here, we investigated the effect of different HDAC inhibitors (HDACIs) on the expression of TEs in leiomyosarcoma cells. Our data show that endogenous retroviruses (ERVs), especially ERV1 elements, are upregulated after treatment with HDAC1/2/3-specific inhibitors. Surprisingly, the interferon response was not activated. We observed an increase in A-to-I editing of upregulated ERV1. This could have an impact on the stability of dsRNAs and the activation of the interferon response. We also found that H3K27ac levels are increased in the LTR12 subfamilies, which could be regulatory elements controlling the expression of proapoptotic genes such as TNFRSF10B. In summary, we provide a detailed characterization of TEs modulation in response to HDACIs and suggest the use of HDACIs in combination with ADAR inhibitors to induce cell death and support immunotherapy in cancer., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
19. IFITM3 reduces infectious bursal disease virus proliferation by regulating interferon expression.
- Author
-
Liu Y, Ma J, Gao P, Li C, Wang Q, Wang L, Xu Z, and Yu Y
- Subjects
- Animals, Cell Line, Birnaviridae Infections veterinary, Birnaviridae Infections virology, Birnaviridae Infections immunology, Poultry Diseases virology, RNA-Binding Proteins genetics, RNA-Binding Proteins metabolism, RNA, Small Interfering genetics, Gene Expression Regulation, Bursa of Fabricius virology, Bursa of Fabricius metabolism, Interferon-beta metabolism, Interferon-beta genetics, Infectious bursal disease virus physiology, Chickens virology, Membrane Proteins genetics, Membrane Proteins metabolism, Interferons metabolism, Interferons genetics, Virus Replication
- Abstract
Interferon-inducible transmembrane protein 3 (IFITM3), a member of the interferon-stimulating factor (ISG) family, has various antiviral functions. Infectious bursal disease virus (IBDV) mainly invades the bursa of Fabricius in chickens, causing a reduction in their immunity and resulting in death from secondary infections. Our previous study found that IBDV infection promotes the expression of chicken IFITM3. However, the role of chicken IFITM3 in IBDV infection remains unknown. To explore this role, the overexpression vector for IFITM3 was constructed and transfected into HD-11 and DF-1 cells. The results showed that the overexpression of IFITM3 significantly reduced IBDV proliferation. While the IBDV proliferation increased when IFITM3 was inhibited by using siRNA. To further explore the mechanism by which IFITM3 reduces IBDV proliferation, the effects of IFITM3 on interferon (IFN) were investigated. Transfecting the constructed IFITM3 vectors into HD-11 and DF-1 cells demonstrated that IFITM3 promoted the expression of IFN-α, IFN-β, and IFN-γ. To investigate the mechanism by which IFITM3 regulates IFN expression, the effects of IFITM3 on IFN production were explored. The results showed that the IKB gene mainly affected the regulatory effects of IFITM3 on IFN. Taken together, IFITM3 may reduce viral proliferation by regulating changes in IFNs, and this process may involve a positive feedback effect of IFITM3 on IFN. IKB plays an important role in the regulation of IFN effects by IFITM3., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
20. Luciferase Reporter Assay for Determining the Signaling Activity of Interferons.
- Author
-
Zhang R and Zheng H
- Subjects
- Humans, Transfection, Animals, Genes, Reporter, Interferons metabolism, Interferons genetics, Signal Transduction, Luciferases metabolism, Luciferases genetics, Promoter Regions, Genetic, Response Elements
- Abstract
The classic dual luciferase reporter assay has been widely used to rapidly and accurately determine the transcriptional activity of a given promoter induced by certain signal pathways in the cells. In particular, the sensitive characteristics of luciferase highlight its significance in many experiments, such as weak promoter analysis, transfection studies using small amounts of DNA, and detection in cell lines with low transfection efficiency. This chapter presents detailed information and experimental procedures for measuring interferon (IFN)-induced Interferon-Stimulated Response Element (ISRE) promoter activity using the dual luciferase reporter assay., (© 2025. The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2025
- Full Text
- View/download PDF
21. SRP54 of black carp negatively regulates MDA5-mediated antiviral innate immunity.
- Author
-
Chu J, Chen Y, Wu Y, Qin W, Yan J, Xiao J, and Feng H
- Subjects
- Animals, Interferons metabolism, Interferons immunology, Interferons genetics, Signal Recognition Particle metabolism, Signal Recognition Particle immunology, Signal Recognition Particle genetics, Humans, Fish Diseases immunology, Fish Diseases virology, Ubiquitination, Rhabdoviridae Infections immunology, Rhabdoviridae Infections veterinary, Rhabdoviridae, Immunity, Innate, Carps immunology, Carps genetics, Fish Proteins genetics, Fish Proteins metabolism, Fish Proteins immunology, Signal Transduction immunology, Interferon-Induced Helicase, IFIH1 metabolism, Interferon-Induced Helicase, IFIH1 genetics
- Abstract
Signal Recognition Particle 54 kDa (SRP54) is a subunit of the signal recognition particle (SRP), a cytoplasmic ribonucleoprotein complex guiding the transportation of newly synthesized proteins from polyribosomes to endoplasmic reticulum. In mammals, it has been reported to regulate the RLR signaling pathway negatively by impairing the association between MAVS and MDA5/RIG-I. However, the role of SRP54 in teleost antiviral innate immune response remains obscure. In this study, the SRP54 homolog of black carp (bcSRP54) has been cloned, and its function in antiviral innate immunity has been elucidated. The CDS of bcSRP54 gene consists of 1515 nucleotides and encodes 504 amino acids. Immunofluorescence (IF) showed that bcSRP54 was mainly distributed in the cytoplasm. Overexpressed bcSRP54 significantly reduced bcMDA5-mediated transcription of interferon (IFN) promoter in reporter assay. Co-expression of bcSRP54 and bcMDA5 significantly suppressed bcMDA5-mediated IFN signaling and antiviral activity, while bcSRP54 knockdown increased the antiviral ability of host cells. In addition, the results of the immunofluorescence staining demonstrated the subcellular overlapping between bcSRP54 and bcMDA5, and the co-immunoprecipitation (co-IP) experiment identified their association. Furthermore, the over-expression of bcSRP54 did not influence the protein expression and ubiquitination modification level of bcMDA5, however, hindered the binding of bcMDA5 to bcMAVS. In summary, our results conclude that bcSRP54 targets bcMDA5 and inhibits the interaction between bcMDA5 and bcMAVS, thereby negatively regulating antiviral innate immunity, which provides insight into how teleost SRP54 regulates IFN signaling., Competing Interests: Declaration of competing interest All authors declare that they have no conflict of interest., (Copyright © 2024 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
22. The nuclear localization signal of monkeypox virus protein P2 orthologue is critical for inhibition of IRF3-mediated innate immunity.
- Author
-
Jiao P, Ma J, Zhao Y, Jia X, Zhang H, Fan W, Jia X, Bai X, Zhao Y, Lu Y, Zhang H, Guo J, Pang G, Zhang K, Fang M, Li M, Liu W, Smith GL, and Sun L
- Subjects
- Animals, Mice, Humans, HEK293 Cells, alpha Karyopherins genetics, alpha Karyopherins metabolism, Immune Evasion, Cell Nucleus metabolism, Interferons genetics, Interferons immunology, Interferons metabolism, Poxviridae Infections immunology, Poxviridae Infections virology, Poxviridae Infections veterinary, Mice, Inbred C57BL, Immunity, Innate, Interferon Regulatory Factor-3 metabolism, Interferon Regulatory Factor-3 genetics, Viral Proteins genetics, Viral Proteins metabolism, Viral Proteins immunology, Nuclear Localization Signals genetics, Monkeypox virus genetics, Monkeypox virus immunology
- Abstract
The Orthopoxvirus (OPXV) genus of the Poxviridae includes human pathogens variola virus (VARV), monkeypox virus (MPXV), vaccinia virus (VACV), and a number of zoonotic viruses. A number of Bcl-2-like proteins of VACV are involved in escaping the host innate immunity. However, little work has been devoted to the evolution and function of their orthologues in other OPXVs. Here, we found that MPXV protein P2, encoded by the P2L gene, and P2 orthologues from other OPXVs, such as VACV protein N2, localize to the nucleus and antagonize interferon (IFN) production. Exceptions to this were the truncated P2 orthologues in camelpox virus (CMLV) and taterapox virus (TATV) that lacked the nuclear localization signal (NLS). Mechanistically, the NLS of MPXV P2 interacted with karyopherin α-2 (KPNA2) to facilitate P2 nuclear translocation, and competitively inhibited KPNA2-mediated IRF3 nuclear translocation and downstream IFN production. Deletion of the NLS in P2 or orthologues significantly enhanced IRF3 nuclear translocation and innate immune responses, thereby reducing viral replication. Moreover, deletion of NLS from N2 in VACV attenuated viral replication and virulence in mice. These data demonstrate that the NLS-mediated translocation of P2 is critical for P2-induced inhibition of innate immunity. Our findings contribute to an in-depth understanding of the mechanisms of OPXV P2 orthologue in innate immune evasion.
- Published
- 2024
- Full Text
- View/download PDF
23. Research progress on the nonstructural protein 1 (NS1) of influenza a virus.
- Author
-
Zhang X, Zhang Y, and Wei F
- Subjects
- Humans, Animals, Autophagy, Virulence, Host-Pathogen Interactions, Apoptosis, Interferons metabolism, Interferons immunology, Interferons genetics, Viral Nonstructural Proteins genetics, Viral Nonstructural Proteins metabolism, Influenza A virus genetics, Influenza A virus pathogenicity, Influenza A virus physiology, Influenza, Human virology
- Abstract
Influenza A virus (IAV) is the leading cause of highly contagious respiratory infections, which poses a serious threat to public health. The non-structural protein 1 (NS1) is encoded by segment 8 of IAV genome and is expressed in high levels in host cells upon IAV infection. It is the determinant of virulence and has multiple functions by targeting type Ι interferon (IFN-I) and type III interferon (IFN-III) production, disrupting cell apoptosis and autophagy in IAV-infected cells, and regulating the host fitness of influenza viruses. This review will summarize the current research on the NS1 including the structure and related biological functions of the NS1 as well as the interaction between the NS1 and host cells. It is hoped that this will provide some scientific basis for the prevention and control of the influenza virus.
- Published
- 2024
- Full Text
- View/download PDF
24. Evolutionary insights of interferon lambda genes in tetrapods.
- Author
-
Gautam D, Sindhu A, Vats A, Rajput S, Rana C, and De S
- Subjects
- Animals, Interleukins genetics, DNA Copy Number Variations, Cattle genetics, Mammals genetics, Buffaloes genetics, Interferon Lambda, Evolution, Molecular, Interferons genetics, Interferons metabolism, Phylogeny
- Abstract
Type III interferon (IFN), also known as IFN-λ, is an innate antiviral protein. We retrieved the sequences of IFN-λ and their receptors from 42 tetrapod species and conducted a computational evolutionary analysis to understand the diversity of these genes. The copy number variation (CNV) of IFN-λ was determined through qPCR in Indian cattle and buffalo. The tetrapod species feature intron-containing type III IFN genes. Some reptiles and placental mammals have 2 IFN-λ loci, while marsupials, monotremes, and birds have a single IFN-λ locus. Some placental mammals and amphibians exhibit multiple IFN-λ genes, including both intron-less and intron-containing forms. Placental mammals typically possess 3-4 functional IFN-λ genes, some of them lack signal peptides. IFN-λ of these tetrapod species formed 3 major clades. Mammalian IFN-λ4 appears as an ancestral form, with syntenic conservation in most mammalian species. The intron-less IFN-λ1 and both type III IFN receptors have conserved synteny in tetrapod. Purifying selection was noted in their evolutionary analysis that plays a crucial role in minimizing genetic diversity and maintaining the integrity of biological function. This indicates that these proteins have successfully retained their biological function and indispensability, even in the presence of the type I IFNs. The expansion of IFN-λ genes in amphibians and camels have led to the evolution of multiple IFN-λ. The CNV can arise from gene duplication and conversion events. The qPCR-based absolute quantification revealed that IFN-λ3 and IFN-λ4 have more than 1 copy in buffalo (Murrah) and 6 cattle breeds (Sahiwal, Tharparkar, Kankrej, Red Sindhi, Jersey, and Holstein Friesian). Overall, these findings highlight the evolutionary diversity and functional significance of IFN-λ in tetrapod species., (© The Author(s) 2024. Published by Oxford University Press on behalf of the European Society of Evolutionary Biology. All rights reserved. For commercial re-use, please contact reprints@oup.com for reprints and translation rights for reprints. All other permissions can be obtained through our RightsLink service via the Permissions link on the article page on our site—for further information please contact journals.permissions@oup.com.)
- Published
- 2024
- Full Text
- View/download PDF
25. Innate immune signatures in the nasopharynx after SARS-CoV-2 infection and links with the clinical outcome of COVID-19 in Omicron-dominant period.
- Author
-
Cha H, Lee CM, Kim S, Kang CK, Choe PG, Jeon YK, Jo HJ, Kim NJ, Park WB, and Kim HJ
- Subjects
- Humans, Male, Female, Middle Aged, Adult, Aged, COVID-19 immunology, COVID-19 virology, Nasopharynx virology, Nasopharynx immunology, Immunity, Innate, SARS-CoV-2 immunology, SARS-CoV-2 physiology, Interferons metabolism, Interferons genetics, Interferons immunology
- Abstract
While severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is characterized by impaired induction of interferons (IFNs) and IFN-stimulated genes (ISGs), the IFNs and ISGs in upper airway is essential to restrict the spread of respiratory virus. Here, we identified the prominent IFN and ISG upregulation in the nasopharynx (NP) of mild and even severe coronavirus disease 2019 (COVID-19) patients (CoV2+) in Omicron era and to compare their clinical outcome depending on the level of IFNs and ISGs. Whereas the induction of IFNB was minimal, transcription of IFNA, IFNG, and IFNLs was significantly increased in the NP of CoV2 + patients. IFNs and ISGs may be more upregulated in the NP of CoV2 + patients at early phases of infection according to viral RNA levels and this is observed even in severe cases. IFN-related innate immune response might be characteristic in macrophages and monocytes at the NP and the CoV2 + patients with higher transcription of IFNs and ISGs in the NP showed a correlation with good prognosis of COVID-19. This study presents that IFNs and ISGs may be upregulated in the NP, even in severe CoV2 + patients depending on viral replication during Omicron-dominant period and the unique IFN-responsiveness in the NP links with COVID-19 clinical outcomes., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
26. A Potential Role for the Amyloid Precursor Protein in the Regulation of Interferon Signaling, Cholesterol Homeostasis, and Tau Phosphorylation in Niemann-Pick Disease Type C.
- Author
-
Sanchez KL, Shin SD, Rajagopal NP, White JB, Currais A, Soriano-Castell D, Maher P, and Soriano S
- Subjects
- Animals, Mice, Cerebral Cortex metabolism, Cerebral Cortex pathology, Disease Models, Animal, Mice, Knockout, Phosphorylation, Amyloid beta-Protein Precursor genetics, Amyloid beta-Protein Precursor metabolism, Cerebellum metabolism, Cerebellum pathology, Cholesterol metabolism, Homeostasis, Interferons metabolism, Interferons genetics, Niemann-Pick C1 Protein, Niemann-Pick Disease, Type C metabolism, Niemann-Pick Disease, Type C genetics, Niemann-Pick Disease, Type C pathology, Signal Transduction, tau Proteins metabolism, tau Proteins genetics
- Abstract
Niemann-Pick disease type C (NPC) is a rare and fatal neurological disorder caused by mutations in Npc1 or Npc2 , with Npc1 accounting for 95% of cases. These mutations result in the functional loss of their respective proteins, causing cellular abnormalities characterized by disrupted lipid dysregulation, calcium dysfunction, elevated damage associated molecular patterns (DAMPs), and a pro-inflammatory environment. This cellular pathology ultimately triggers neurodegeneration, with the cerebellum being the earliest and most affected region. We have recently shown atypical activation of interferon signaling in the presymptomatic Npc1
-/- mouse cerebellum and, to a lesser extent, in the cerebral cortex. In addition, we reported that the Amyloid Precursor Protein (APP) is an NPC disease modifier. Loss of APP function leads to widespread neurodegeneration in the NPC brain, including exacerbated interferon signaling in the cerebellum. To better understand the role of APP as a disease modifier throughout the NPC brain, here we carried out a transcriptomic analysis of the cerebral cortex and cerebellum from 3-week-old Npc1-/- mice as well as age-matched controls in the presence and absence of APP. We report differential effects of APP loss of function in the cerebral cortex and cerebellum, including cholesterol and tau dysregulation, in both brain regions. Our findings demonstrate a novel link between APP loss and early pathogenic mechanisms in NPC.- Published
- 2024
- Full Text
- View/download PDF
27. Competition between two HUSH complexes orchestrates the immune response to retroelement invasion.
- Author
-
Danac JMC, Matthews RE, Gungi A, Qin C, Parsons H, Antrobus R, Timms RT, and Tchasovnikarova IA
- Subjects
- Humans, HEK293 Cells, Histones metabolism, Histones genetics, Retroelements genetics, Epigenesis, Genetic, Long Interspersed Nucleotide Elements genetics, Signal Transduction, Interferons metabolism, Interferons immunology, Interferons genetics, HeLa Cells, Gene Silencing
- Abstract
The human silencing hub (HUSH) preserves genome integrity through the epigenetic repression of invasive genetic elements. However, despite our understanding of HUSH as an obligate complex of three subunits, only loss of MPP8 or Periphilin, but not TASOR, triggers interferon signaling following derepression of endogenous retroelements. Here, we resolve this paradox by characterizing a second HUSH complex that shares MPP8 and Periphilin but assembles around TASOR2, an uncharacterized paralog of TASOR. Whereas HUSH represses LINE-1 retroelements marked by the repressive histone modification H3K9me3, HUSH2 is recruited by the transcription factor IRF2 to repress interferon-stimulated genes. Mechanistically, HUSH-mediated retroelement silencing sequesters the limited pool of the shared subunits MPP8 and Periphilin, preventing TASOR2 from forming HUSH2 complexes and hence relieving the HUSH2-mediated repression of interferon-stimulated genes. Thus, competition between two HUSH complexes intertwines retroelement silencing with the induction of an immune response, coupling epigenetic and immune aspects of genome defense., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
28. Differential Contributions of Interferon Classes to Host Inflammatory Responses and Restricting Virus Progeny Production.
- Author
-
Lum KK, Reed TJ, Yang J, and Cristea IM
- Subjects
- Humans, Inflammation virology, Inflammation immunology, Interferon-gamma immunology, Interferon-beta metabolism, Interferon-beta immunology, Interferon-beta genetics, Immunity, Innate, Keratinocytes virology, Keratinocytes immunology, Keratinocytes drug effects, Keratinocytes metabolism, Virus Replication drug effects, Macrophages immunology, Macrophages virology, Macrophages metabolism, Macrophages drug effects, Cytomegalovirus immunology, Herpesvirus 1, Human physiology, Herpesvirus 1, Human immunology, Interferons immunology, Interferons metabolism, Interferons genetics, Lipopolysaccharides pharmacology, Monocytes immunology, Monocytes virology, Monocytes metabolism, Monocytes drug effects, Host-Pathogen Interactions immunology, Proteome, Proteomics methods
- Abstract
Fundamental to mammalian intrinsic and innate immune defenses against pathogens is the production of Type I and Type II interferons, such as IFN-β and IFN-γ, respectively. The comparative effects of IFN classes on the cellular proteome, protein interactions, and virus restriction within cell types that differentially contribute to immune defenses are needed for understanding immune signaling. Here, a multilayered proteomic analysis, paired with biochemical and molecular virology assays, allows distinguishing host responses to IFN-β and IFN-γ and associated antiviral impacts during infection with several ubiquitous human viruses. In differentiated macrophage-like monocytic cells, we classified proteins upregulated by IFN-β, IFN-γ, or pro-inflammatory LPS. Using parallel reaction monitoring, we developed a proteotypic peptide library for shared and unique ISG signatures of each IFN class, enabling orthogonal confirmation of protein alterations. Thermal proximity coaggregation analysis identified the assembly and maintenance of IFN-induced protein interactions. Comparative proteomics and cytokine responses in macrophage-like monocytic cells and primary keratinocytes provided contextualization of their relative capacities to restrict virus production during infection with herpes simplex virus type-1, adenovirus, and human cytomegalovirus. Our findings demonstrate how IFN classes induce distinct ISG abundance and interaction profiles that drive antiviral defenses within cell types that differentially coordinate mammalian immune responses.
- Published
- 2024
- Full Text
- View/download PDF
29. [Myxovirus resistance protein A (MxA) induces the expression of interferon-stimulated genes in HepG2 cells by enhancing the activity of the interferon-stimulated response element (ISRE)].
- Author
-
Yang K, Pan Y, Liu P, Yu F, Wei X, Zhang F, and Wang Q
- Subjects
- Humans, Hep G2 Cells, Interferon-alpha pharmacology, Interferon-alpha genetics, Interferon-alpha metabolism, Response Elements genetics, Signal Transduction, STAT2 Transcription Factor genetics, STAT2 Transcription Factor metabolism, Interferons genetics, Interferons metabolism, Interferon-Stimulated Gene Factor 3, gamma Subunit genetics, Interferon-Stimulated Gene Factor 3, gamma Subunit metabolism, Gene Expression Regulation, Myxovirus Resistance Proteins genetics, Myxovirus Resistance Proteins metabolism, 2',5'-Oligoadenylate Synthetase genetics, 2',5'-Oligoadenylate Synthetase metabolism, eIF-2 Kinase genetics, eIF-2 Kinase metabolism, STAT1 Transcription Factor genetics, STAT1 Transcription Factor metabolism
- Abstract
Objective To explore the effects of Myxovirus resistance protein A (MxA) on the Janus kinase/Signal transducer and activator of transcription (JAK/STAT) pathway in HepG2 cells. Methods HepG2 cells were transfected with the pcDNA3.1-Flag-MxA construct, and subsequent localization and expression of the MxA protein were detected through immunofluorescence cytochemistry. The presence of MxA protein was further confirmed by using Western blot analysis. Following transfection with MxA small interfering RNA (si-MxA) and subsequent treatment with alpha interferon (IFN-α), real-time fluorescent quantitative PCR was employed to measure the mRNA levels of myxovirus resistance protein A (MxA), protein kinase R (PKR), and oligoadenylate synthase (OAS). Western blot analysis was used to detect the protein expression of MxA, PKR, OAS, signal transducer and activator of transcription 1 (STAT1), phosphorylated STAT1 (pSTAT1), STAT2, phosphorylated STAT2 (p-STAT2) and interferon regulatory factor 9 (IRF9). Additionally, pcDNA3.1-Flag-MxA and pISRE-TA-luc were co-transfected into HepG2 and HepG2.2.15 cells, respectively, to assess the activity of the interferon-stimulated response element (ISRE) by using a luciferase activity assay. Results MxA protein was expressed in both the cytoplasm and nucleus of HepG2 cells, with higher expression levels in the cytoplasm than in the nucleus. Knocking down MxA expression in HepG2 cells did not affect the expression of STAT1, p-STAT1, STAT2, p-STAT2, and IRF9 proteins induced by IFN-α, but significantly reduced the expression of antiviral proteins PKR and OAS. Overexpression of MxA in HepG2 cells enhanced ISRE activity and increased the expression of PKR and OAS proteins, but this effect was inhibited in HepG2.2.15 cells. Conclusion MxA induces the expression of antiviral proteins by enhancing the activity of the JAK/STAT signaling pathway ISRE.
- Published
- 2024
30. Pseudorabies virus VHS protein abrogates interferon responses by blocking NF-κB and IRF3 nuclear translocation.
- Author
-
Yan Z, Yue J, Zhang Y, Hou Z, Li D, Yang Y, Li X, Idris A, Li H, Li S, Xie J, and Feng R
- Subjects
- Animals, Swine, Humans, Interferons immunology, Interferons metabolism, Interferons genetics, Pseudorabies virology, Pseudorabies immunology, Cell Line, Host-Pathogen Interactions immunology, Viral Proteins genetics, Viral Proteins metabolism, Viral Proteins immunology, HEK293 Cells, Phosphorylation, Protein Transport, Herpesvirus 1, Suid immunology, Herpesvirus 1, Suid genetics, Interferon Regulatory Factor-3 metabolism, Interferon Regulatory Factor-3 genetics, Immune Evasion, NF-kappa B metabolism, NF-kappa B genetics, NF-kappa B immunology
- Abstract
Herpesviruses antagonize host antiviral responses through a myriad of molecular strategies culminating in the death of the host cells. Pseudorabies virus (PRV) is a significant veterinary pathogen in pigs, causing neurological sequalae that ultimately lead to the animal's demise. PRV is known to trigger apoptotic cell death during the late stages of infection. The virion host shutdown protein (VHS) encoded by UL41 plays a crucial role in the PRV infection process. In this study, we demonstrate that UL41 inhibits PRV-induced activation of inflammatory cytokine and negatively regulates the cGAS-STING-mediated antiviral activity by targeting IRF3, thereby inhibiting the translocation and phosphorylation of IRF3. Notably, mutating the conserved amino acid sites (E192, D194, and D195) in the RNase domain of UL41 or knocking down UL41 inhibits the immune evasion of PRV, suggesting that UL41 may play a crucial role in PRV's evasion of the host immune response during infection. These results enhance our understanding of how PRV structural proteins assist the virus in evading the host immune response., Competing Interests: Conflict of interest All authors declare that there are no competing interests., (Copyright © 2024 The Authors. Publishing services by Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
31. Singapore grouper iridovirus VP146 modulates the cGAS-STING signaling pathway to escape the interferon immune response.
- Author
-
Xu L, Xu Q, Mo W, Chen H, Wu S, Qin Q, and Wei J
- Subjects
- Cell Line, Spleen cytology, Gene Expression Regulation immunology, Virus Replication immunology, Interferons genetics, Interferons immunology, Fish Proteins immunology, Animals, Iridovirus immunology, Nucleotidyltransferases genetics, Nucleotidyltransferases immunology, Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing immunology, Signal Transduction immunology, Bass genetics, Bass immunology, Bass virology
- Abstract
Singapore grouper iridovirus (SGIV) is a large double-stranded DNA virus that has caused significant economic losses to the grouper aquaculture industry. So far, the structure and function of SGIV proteins have been successively reported. In the present paper, the protein of SGIV VP146 was cloned and identified. VP146 was whole-cell distributed in GS cells. VP146 promoted SGIV replication and inhibited the transcription of interferon-related genes as well as pro-inflammatory cytokines in GS cells. In addition, VP146 was involved in the regulation of the cGAS-STING signaling pathway, and decreased cGAS-STING induced the promoter of ISRE and NF-κB. VP146 interacted with the proteins of cGAS, STING, TBK1, and IRF3 from grouper, but did not affect the binding of grouper STING to grouper TBK1 and grouper IRF3. Interestingly, grouper STING was able to affect the intracellular localization of VP146. Four segment structural domains of grouper STING were constructed, and grouper STING-CTT could affect the intracellular localization of VP146. VP146 had no effect on the self-binding of EcSITNG, nor on the binding of EcSTING to EcTBK1 and EcIRF3. Together, the results demonstrated that SGIV VP146 modulated the cGAS-STING signaling pathway to escape the interferon immune response., Competing Interests: Declaration of competing interest The authors declare no conflict of interest., (Copyright © 2024 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
32. Two cytokine receptor family B (CRFB) members in orange-spotted grouper Epinephelus coioides, EcCRFB3 and EcCRFB4, negatively regulate interferon immune responses to assist nervous necrosis virus replication.
- Author
-
Huang S, Kang Y, Zheng R, Yang L, Gao J, Tang W, Jiang J, He J, and Xie J
- Subjects
- Animals, Phylogeny, Sequence Alignment veterinary, Receptors, Cytokine genetics, Receptors, Cytokine immunology, Gene Expression Profiling veterinary, Interferons immunology, Interferons genetics, Nodaviridae physiology, Fish Diseases immunology, Fish Diseases virology, RNA Virus Infections immunology, RNA Virus Infections veterinary, Fish Proteins genetics, Fish Proteins immunology, Virus Replication, Bass immunology, Bass genetics, Immunity, Innate genetics, Amino Acid Sequence, Gene Expression Regulation immunology
- Abstract
Receptors of type I interferon (IFNR) play a vital role in the antiviral immune response. However, little is known about the negative regulatory role of the IFNR. Nervous necrosis virus (NNV) is one of the most significant viruses in cultured fish, resulting in great economic losses for the aquaculture industry. In this study, two orange-spotted grouper (Epinephelus coioides) cytokine receptor family B (CRFB) members, EcCRFB3 and EcCRFB4 were cloned and characterized from NNV infected grouper brain (GB) cells. The open reading frame (ORF) of EcCRFB3 consists of 852 bp encoding 283 amino acids, while EcCRFB4 has an ORF of 990 bp encoding 329 amino acids. The mRNA levels of EcCRFB3 or EcCRFB4 were significantly upregulated after NNV infection and the stimulation of poly (I:C) or NNV-encoded Protein A. In addition, EcCRFB3 or EcCRFB4 overexpression facilitated NNV replication, whereas EcCRFB3 or EcCRFB4 silencing resisted NNV replication. Overexpressed EcCRFB3 or EcCRFB4 inhibited the expression of IFN-I-induced ISGs. Taken together, our research provides the first evidence in fish demonstrating the role of IFNRs to regulate the IFN signaling pathway negatively. Our findings enrich the understanding of the functions of IFNRs and reveal a novel escape mechanism of NNV., Competing Interests: Declaration of competing interest The authors declare that they have no competing interests., (Copyright © 2024 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
33. Type III interferon drives pathogenicity to Staphylococcus aureus via the airway epithelium.
- Author
-
Pires S, Kaiser K, and Parker D
- Subjects
- Animals, Mice, Mice, Inbred C57BL, Host-Pathogen Interactions, Signal Transduction, Respiratory Mucosa microbiology, Interferon Lambda, Receptors, Interferon genetics, Receptors, Interferon metabolism, Virulence, Staphylococcus aureus pathogenicity, Staphylococcus aureus genetics, Macrophages, Alveolar microbiology, Macrophages, Alveolar immunology, Staphylococcal Infections microbiology, Mice, Knockout, Interferons metabolism, Interferons genetics, Interferons immunology
- Abstract
Type III interferon signaling contributes to the pathogenesis of the important human pathogen Staphylococcus aureus in the airway. Little is known of the cellular factors important in this response. Using Ifnl2 -green fluorescent protein reporter mice combined with flow cytometry and cellular depletion strategies, we demonstrate that the alveolar macrophage is the primary producer of interferon lambda (IFN-λ) in response to S. aureus in the airway. Bone marrow chimeras showed reduced bacterial burden in IFN-λ receptor (IFNLR1)-deficient recipient mice, indicative that non-hematopoietic cells were important for pathogenesis, in addition to significant reductions in pulmonary inflammation. These observations were confirmed through the use of an airway epithelial-specific IFNLR knockout mouse. Our data suggest that upon entry to the airway, S. aureus activates alveolar macrophages to produce type III IFN that is subsequently sensed by the airway epithelium. Future steps will determine how signaling from the epithelium then exerts its influence on bacterial clearance. These results highlight the important, yet sometimes detrimental, role of type III IFN signaling during infection and the impact the airway epithelium plays during host-pathogen interactions.IMPORTANCEThe contribution of type III interferon signaling to the control of bacterial infections is largely unknown. We have previously demonstrated that it contributes to the pathogenesis of acute Staphylococcus aureus respiratory infection. In this report, we document the importance of two cell types that underpin this pathogenesis. We demonstrate that the alveolar macrophage is the cell that is responsible for the production of type III interferon and that this molecule is sensed by airway epithelial cells, which impacts both bacterial clearance and induction of inflammation. This work sheds light on the first two aspects of this important pathogenic cascade., Competing Interests: The authors declare no conflict of interest.
- Published
- 2024
- Full Text
- View/download PDF
34. Genomic profiling of a multi-lineage and multi-passage patient-derived xenograft biobank reflects heterogeneity of ovarian cancer.
- Author
-
Qin T, Hu Z, Zhang L, Lu F, Xiao R, Liu Y, Fan J, Guo E, Yang B, Fu Y, Zhuang X, Kang X, Wu Z, Fang Z, Cui Y, Hu X, Yin J, Yan M, Li F, Song K, Chen G, and Sun C
- Subjects
- Humans, Female, Animals, Mice, Xenograft Model Antitumor Assays, Wnt Signaling Pathway genetics, Drug Resistance, Neoplasm genetics, Genomics methods, Biological Specimen Banks, Genetic Heterogeneity, DNA Damage genetics, Interferons metabolism, Interferons genetics, Cell Lineage genetics, Ovarian Neoplasms genetics, Ovarian Neoplasms pathology, Ovarian Neoplasms metabolism
- Abstract
Ovarian cancer (OC) manifests as a complex disease characterized by inter- and intra-patient heterogeneity. Despite enhanced biological and genetic insights, OC remains a recalcitrant malignancy with minimal survival improvement. Based on multi-site sampling and a multi-lineage patient-derived xenograft (PDX) establishment strategy, we present herein the establishment of a comprehensive PDX biobank from histologically and molecularly heterogeneous OC patients. Comprehensive profiling of matched PDX and patient samples demonstrates that PDXs closely recapitulate parental tumors. By leveraging multi-lineage models, we reveal that the previously reported genomic disparities of PDX could be mainly attributed to intra-patient spatial heterogeneity instead of substantial model-independent genomic evolution. Moreover, DNA damage response pathway inhibitor (DDRi) screening uncovers heterogeneous responses across models. Prolonged iterative drug exposure recapitulates acquired drug resistance in initially sensitive models. Meanwhile, interrogation of induced drug-resistant (IDR) models reveals that suppressed interferon (IFN) response and activated Wnt/β-catenin signaling contribute to acquired DDRi drug resistance., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
35. PELI2 is a negative regulator of STING signaling that is dynamically repressed during viral infection.
- Author
-
Ritchie C and Li L
- Subjects
- Humans, Phosphorylation, Animals, HEK293 Cells, Lupus Erythematosus, Systemic genetics, Lupus Erythematosus, Systemic immunology, Lupus Erythematosus, Systemic metabolism, Lupus Erythematosus, Systemic virology, Immunity, Innate, Host-Pathogen Interactions, Ubiquitin-Protein Ligases metabolism, Ubiquitin-Protein Ligases genetics, Mice, Interferons metabolism, Interferons immunology, Interferons genetics, Feedback, Physiological, Mice, Inbred C57BL, Exodeoxyribonucleases, Phosphoproteins, Signal Transduction, Protein Serine-Threonine Kinases metabolism, Protein Serine-Threonine Kinases genetics, Membrane Proteins metabolism, Membrane Proteins genetics, Interferon Regulatory Factor-3 metabolism, Interferon Regulatory Factor-3 genetics, Ubiquitination
- Abstract
The innate immune cGAS-STING pathway is activated by cytosolic double-stranded DNA (dsDNA), a ubiquitous danger signal, to produce interferon, a potent anti-viral and anti-cancer cytokine. However, STING activation must be tightly controlled because aberrant interferon production leads to debilitating interferonopathies. Here, we discover PELI2 as a crucial negative regulator of STING. Mechanistically, PELI2 inhibits the transcription factor IRF3 by binding to phosphorylated Thr354 and Thr356 on the C-terminal tail of STING, leading to ubiquitination and inhibition of the kinase TBK1. PELI2 sets a threshold for STING activation that tolerates low levels of cytosolic dsDNA, such as that caused by silenced TREX1, RNASEH2B, BRCA1, or SETX. When this threshold is reached, such as during viral infection, STING-induced interferon production temporarily downregulates PELI2, creating a positive feedback loop allowing a robust immune response. Lupus patients have insufficient PELI2 levels and high basal interferon production, suggesting that PELI2 dysregulation may drive the onset of lupus and other interferonopathies., Competing Interests: Declaration of interests C.R. and L.L. have filed a patent on using PELI2 as a biomarker (application no. PCT/US24/24438)., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
36. PSMD11 loss-of-function variants correlate with a neurobehavioral phenotype, obesity, and increased interferon response.
- Author
-
Deb W, Rosenfelt C, Vignard V, Papendorf JJ, Möller S, Wendlandt M, Studencka-Turski M, Cogné B, Besnard T, Ruffier L, Toutain B, Poirier L, Cuinat S, Kritzer A, Crunk A, diMonda J, Vengoechea J, Mercier S, Kleinendorst L, van Haelst MM, Zuurbier L, Sulem T, Katrínardóttir H, Friðriksdóttir R, Sulem P, Stefansson K, Jonsdottir B, Zeidler S, Sinnema M, Stegmann APA, Naveh N, Skraban CM, Gray C, Murrell JR, Isikay S, Pehlivan D, Calame DG, Posey JE, Nizon M, McWalter K, Lupski JR, Isidor B, Bolduc FV, Bézieau S, Krüger E, Küry S, and Ebstein F
- Subjects
- Adolescent, Animals, Child, Child, Preschool, Female, Humans, Male, Interferons metabolism, Interferons genetics, Loss of Function Mutation, Phenotype, Drosophila melanogaster genetics, Intellectual Disability genetics, Neurodevelopmental Disorders genetics, Obesity genetics, Proteasome Endopeptidase Complex genetics, Proteasome Endopeptidase Complex metabolism
- Abstract
Primary proteasomopathies have recently emerged as a new class of rare early-onset neurodevelopmental disorders (NDDs) caused by pathogenic variants in the PSMB1, PSMC1, PSMC3, or PSMD12 proteasome genes. Proteasomes are large multi-subunit protein complexes that maintain cellular protein homeostasis by clearing ubiquitin-tagged damaged, misfolded, or unnecessary proteins. In this study, we have identified PSMD11 as an additional proteasome gene in which pathogenic variation is associated with an NDD-causing proteasomopathy. PSMD11 loss-of-function variants caused early-onset syndromic intellectual disability and neurodevelopmental delay with recurrent obesity in 10 unrelated children. Our findings demonstrate that the cognitive impairment observed in these individuals could be recapitulated in Drosophila melanogaster with depletion of the PMSD11 ortholog Rpn6, which compromised reversal learning. Our investigations in subject samples further revealed that PSMD11 loss of function resulted in impaired 26S proteasome assembly and the acquisition of a persistent type I interferon (IFN) gene signature, mediated by the integrated stress response (ISR) protein kinase R (PKR). In summary, these data identify PSMD11 as an additional member of the growing family of genes associated with neurodevelopmental proteasomopathies and provide insights into proteasomal biology in human health., Competing Interests: Declaration of interests A.C. and K.M. are employees of GeneDx, LLC. J.R.L. has stock in 23andMe and is a paid consultant for Genome International., (Copyright © 2024 American Society of Human Genetics. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
37. Translation Inhibition Mediated by Interferon-Stimulated Genes during Viral Infections.
- Author
-
Smart A, Gilmer O, and Caliskan N
- Subjects
- Animals, Humans, Host-Pathogen Interactions, Viral Proteins genetics, Viral Proteins metabolism, Virus Replication, Viruses immunology, Viruses genetics, Viruses drug effects, Interferons immunology, Interferons metabolism, Interferons genetics, Protein Biosynthesis, RNA, Viral genetics, Virus Diseases immunology, Virus Diseases virology, Virus Diseases genetics
- Abstract
Viruses often pose a significant threat to the host through the exploitation of cellular machineries for their own benefit. In the context of immune responses, myriad host factors are deployed to target viral RNAs and inhibit viral protein translation, ultimately hampering viral replication. Understanding how "non-self" RNAs interact with the host translation machinery and trigger immune responses would help in the development of treatment strategies for viral infections. In this review, we explore how interferon-stimulated gene products interact with viral RNA and the translation machinery in order to induce either global or targeted translation inhibition., Competing Interests: The authors declare no conflicts of interest.
- Published
- 2024
- Full Text
- View/download PDF
38. CgADAR1 involved in regulating the synthesis of interferon-like protein in Crassostrea gigas.
- Author
-
He Q, Liu C, Liu Q, Wang L, and Song L
- Subjects
- Animals, Amino Acid Sequence, Adenosine Deaminase genetics, Adenosine Deaminase metabolism, Phylogeny, Gene Expression Profiling, Sequence Alignment, Base Sequence, Crassostrea immunology, Crassostrea genetics, Immunity, Innate genetics, Gene Expression Regulation immunology, Interferons genetics, Interferons immunology
- Abstract
Adenosine deaminases acting on RNA 1 (ADAR1) is a dsRNA adenosine (A)-to-inosine (I) editing enzyme that regulates the innate immune response against virus invasion. In the present study, a novel CgADAR1 was identified from the oyster Crassostrea gigas. The open reading frame (ORF) of CgADAR1 was of 3444 bp encoding a peptide of 1147 amino acid residues with two Zα domains, one dsRNA binding motif (DSRM) and one RNA adenosine deaminase domain (ADEAMc). The mRNA transcripts of CgADAR1 were detected in all the examined tissues, with higher expression levels in mantle and gill, which were 7.11-fold and 4.90-fold (p < 0.05) of that in labial palp, respectively. The mRNA transcripts of CgADAR1 in haemocytes were significantly induced at 24 h and 36 h after Poly (A: U) stimulation, which were 6.03-fold (p < 0.01) and 1.37-fold (p < 0.001) of that in control group, respectively. At 48 h after Poly (A:U) stimulation, the mRNA expression of CgRIG-Ⅰ, CgIRF8 and CgIFNLP significantly increased, which were 4.36-fold (p < 0.001), 1.82-fold (p < 0.05) and 1.92-fold (p < 0.05) of that in control group. After CgADAR1 expression was inhibited by RNA interference (RNAi), the mRNA expression levels of CgMDA5, CgRIG-Ⅰ, CgTBK1, CgIRF8 and CgIFNLP were significantly increased, which were 11.88-fold, 11.51-fold, 2.22-fold, 2.85-fold and 2.52-fold of that in control group (p < 0.001), and the phosphorylation level of CgTBK1 was also significantly increased. These results suggested that CgADAR1 played a regulation role in the early stages of viral infection by inhibiting the synthesis of interferon-like protein., (Copyright © 2024 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
39. Variations in the interferon and TLR3 genes may be associated with susceptibility to systemic lupus erythematosus and its clinical presentation.
- Author
-
Modzelewska E, Wajda A, Lutkowska A, Felis-Giemza A, Stypińska B, Matusiewicz A, Puszczewicz M, Majewski D, Jagodziński PP, Haładyj E, and Paradowska-Gorycka A
- Subjects
- Humans, Female, Male, Adult, Middle Aged, Gene Frequency, Alleles, Case-Control Studies, Interferons genetics, Genetic Association Studies, Lupus Erythematosus, Systemic genetics, Toll-Like Receptor 3 genetics, Genetic Predisposition to Disease, Polymorphism, Single Nucleotide, Genotype
- Abstract
The study aimed to explore the pontential impact of 10 polymorphisms within IFN-α, IFN-β1, IFN-γ and TLR3 genes on SLE phenotype and susceptibility and to study the relationship between specific genotypes and clinics. Whole blood samples from SLE patients and healthy controls was obtained. DNA was extracted from the peripheral blood by the QIAamp DNA Blood Mini Kit (Qiagen). The quality and quantity of isolated DNA was estimated by the Quawell Q5000 spectrophotometer. We genotyped SLE patients and healthy subjects using real-time PCR (QuantStudio 5 thermocycler). The study suggests that IFN-γ rs2069705, IFN-γ rs2069718 and IFN-α rs3758236 polymorphisms have a protective role in SLE. We observed relations between TLR3 rs3775292, IFN-β1 rs7873167, IFN-γ rs2069705, TLR3 rs3775291 and TLR3 rs5743305 polymorphisms and clinical picture of SLE patients. We found associations between the IFN-α rs3758236, IFN-γ rs2069705, IFN-γ rs2069718, IFN-γ rs1861493 and IFN-β1 rs10964831 polymorphisms and the clinical manifestation of the SLE and/or its comorbidities. We perceived links between IFN-γ rs2069705, IFN-γ rs2069718, IFN-γ rs1861493, TLR3 rs3775291, TLR3 rs3775292 and TLR3 rs5743305 polymorphisms and the occurrence of autoantibodies. Our study presented the relationship between IFN and TLR gene polymorphisms with SLE susceptibility, phenotype and autoantibodies profile. This study propose that polymorphisms within interferons and TLR3 genes can be engaged in the SLE pathogenesis and course., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Authors. Published by Elsevier GmbH.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
40. USP46 promotes the interferon antiviral signaling in black carp by deubiquitinating TBK1.
- Author
-
Shu J, Yang C, Miao Y, Li J, Zheng T, Xiao J, Kong W, Xu Z, and Feng H
- Subjects
- Animals, Humans, Ubiquitin-Specific Proteases metabolism, Ubiquitin-Specific Proteases genetics, Carps immunology, Immunity, Innate, Fish Proteins metabolism, Fish Proteins genetics, Signal Transduction, Protein Serine-Threonine Kinases metabolism, Protein Serine-Threonine Kinases genetics, Ubiquitination, Interferons metabolism, Interferons genetics
- Abstract
Ubiquitin-specific peptidase 46 (USP46) functions as a deubiquitinating enzyme, facilitating the removal of ubiquitin molecules attached to substrate proteins and playing a critical role in cancer and neurodegenerative diseases. However, its function in innate antiviral immunity is unknown. In this study we cloned and identified bcUSP46, a homolog of USP46 from black carp. We discovered that overexpression of bcUSP46 enhanced the transcription of interferon (IFN) promoters and increased the expression of IFN, PKR, and Mx1. In addition, bcUSP46 knockdown significantly inhibited the expression of ISG genes, as well as the antiviral activity of the host cells. Interestingly, when bcUSP46 was co-expressed with the RLR factors, it significantly enhanced the activity of the IFN promoter mediated by these factors, especially TANK-binding kinase 1 (TBK1). The subsequent co-immunoprecipitation (co-IP) and immunofluorescence (IF) assay confirmed the association between bcUSP46 and bcTBK1. Noteworthily, co-expression of bcUSP46 with bcTBK1 led to an elevation of bcTBK1 protein level. Further analysis revealed that bcUSP46 stabilized bcTBK1 by eliminating the K48-linked ubiquitination of bcTBK1. Overall, our findings highlight the unique role of USP46 in modulating TBK1/IFN signaling and enrich our knowledge of the function of deubiquitination in regulating innate immunity in vertebrates., Competing Interests: Declaration of competing interest The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2024 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
41. SGIV VP82 inhibits the interferon response by degradation of IRF3 and IRF7.
- Author
-
Wang Y, Liu S, Wang W, Liu L, Zhao Y, Qin Q, Huang X, and Huang Y
- Subjects
- Animals, Immunity, Innate genetics, Interferons genetics, Interferons immunology, Interferons metabolism, Immune Evasion, Bass immunology, Bass genetics, Virus Replication, Zebrafish Proteins, Interferon Regulatory Factors, Interferon Regulatory Factor-3 genetics, Interferon Regulatory Factor-3 immunology, Interferon Regulatory Factor-3 metabolism, Interferon Regulatory Factor-7 genetics, Interferon Regulatory Factor-7 metabolism, Interferon Regulatory Factor-7 immunology, Fish Proteins genetics, Fish Proteins immunology, Fish Proteins metabolism, Fish Diseases immunology, Fish Diseases virology, DNA Virus Infections immunology, DNA Virus Infections veterinary, Ranavirus physiology, Viral Proteins genetics, Viral Proteins metabolism
- Abstract
During virus-host co-evolution, viruses have developed multiple strategies to dampen IFN response and prevent its antiviral activity in host cells. To date, the interactions between host IFN response and the immune evasion strategies exploited by fish iridoviruses still remain largely uncertain. Here, a potential immune evasion protein candidate of Singapore grouper iridovirus (SGIV), VP82 (encoded by SGIV ORF82) was screened and its roles during viral replication were investigated in detail. Firstly, VP82 overexpression dramatically decreased IFN or ISRE promoter activity and the transcription levels of IFN stimulated genes (ISGs) stimulated by grouper cyclic GMP-AMP synthase (EccGAS)/stimulator of interferon genes (EcSTING), TANK-binding kinase 1 (EcTBK1), IFN regulatory factor 3 (EcIRF3)and EcIRF7. Secondly, Co-IP assays indicated that VP82 interacted with EcIRF3 and EcIRF7, but not EcSTING and EcTBK1, which was consistent with the co-localization between VP82 and EcIRF3 or EcIRF7. Furthermore, VP82 promoted the degradation of EcIRF3 and EcIRF7 in a dose-dependent manner via the autophagy pathway. Finally, VP82 overexpression accelerated SGIV replication, evidenced by the increased transcriptions of viral core genes and viral production. Moreover, the antiviral action of EcIRF3 or EcIRF7 was significantly depressed in VP82 overexpressed cells. Together, VP82 was speculated to exert crucial roles for SGIV replication by inhibiting the IFN response via the degradation of IRF3 and IRF7. Our findings provided new insights into understanding the immune evasion strategies utilized by fish iridovirus through IFN regulation., (Copyright © 2024 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
42. Assessing the Potential of an Enzymatically Liberated Salmon Oil to Support Immune Health Recovery from Acute SARS-CoV-2 Infection via Change in the Expression of Cytokine, Chemokine and Interferon-Related Genes.
- Author
-
Currie C, Myklebust TÅ, Bjerknes C, and Framroze B
- Subjects
- Humans, Male, Female, Middle Aged, Adult, COVID-19 Drug Treatment, Fatty Acids, Omega-3 pharmacology, Fish Oils pharmacology, Fish Oils therapeutic use, COVID-19 immunology, COVID-19 virology, Interferons metabolism, Interferons genetics, SARS-CoV-2 immunology, Cytokines metabolism, Chemokines metabolism, Chemokines genetics
- Abstract
Cytokines, chemokines, and interferons are released in response to viral infection with the ultimate aim of viral clearance. However, in SARS-CoV-2 infection, there is an imbalanced immune response, with raised cytokine levels but only a limited interferon response with inefficient viral clearance. Furthermore, the inflammatory response can be exaggerated, which risks both acute and chronic sequelae. Several observational studies have suggested a reduced risk of progression to severe COVID-19 in subjects with a higher omega-3 index. However, randomized studies of omega-3 supplementation have failed to replicate this benefit. Omega-3 fats provide important anti-inflammatory effects; however, fatty fish contains many other fatty acids that provide health benefits distinct from omega-3. Therefore, the immune health benefit of whole salmon oil (SO) was assessed in adults with mild to moderate COVID-19. Eleven subjects were randomized to best supportive care (BSC) with or without a full spectrum, enzymatically liberated SO, dosed at 4g daily, for twenty-eight days. Nasal swabs were taken to measure the change in gene expression of markers of immune response and showed that the SO provided both broad inflammation-resolving effects and improved interferon response. The results also suggest improved lung barrier function and enhanced immune memory, although the clinical relevance needs to be assessed in longer-duration studies. In conclusion, the salmon oil was well tolerated and provided broad inflammation-resolving effects, indicating a potential to enhance immune health.
- Published
- 2024
- Full Text
- View/download PDF
43. Senescence of human pancreatic beta cells enhances functional maturation through chromatin reorganization and promotes interferon responsiveness.
- Author
-
Patra M, Klochendler A, Condiotti R, Kaffe B, Elgavish S, Drawshy Z, Avrahami D, Narita M, Hofree M, Drier Y, Meshorer E, Dor Y, and Ben-Porath I
- Subjects
- Humans, Insulin Secretion, Insulin metabolism, Chromatin metabolism, Cyclin-Dependent Kinase Inhibitor p16 metabolism, Cyclin-Dependent Kinase Inhibitor p16 genetics, Cells, Cultured, Senescence-Associated Secretory Phenotype genetics, Transcriptome, Single-Cell Analysis, Insulin-Secreting Cells metabolism, Cellular Senescence genetics, Chromatin Assembly and Disassembly, Interferons metabolism, Interferons genetics
- Abstract
Senescent cells can influence the function of tissues in which they reside, and their propensity for disease. A portion of adult human pancreatic beta cells express the senescence marker p16, yet it is unclear whether they are in a senescent state, and how this affects insulin secretion. We analyzed single-cell transcriptome datasets of adult human beta cells, and found that p16-positive cells express senescence gene signatures, as well as elevated levels of beta-cell maturation genes, consistent with enhanced functionality. Senescent human beta-like cells in culture undergo chromatin reorganization that leads to activation of enhancers regulating functional maturation genes and acquisition of glucose-stimulated insulin secretion capacity. Strikingly, Interferon-stimulated genes are elevated in senescent human beta cells, but genes encoding senescence-associated secretory phenotype (SASP) cytokines are not. Senescent beta cells in culture and in human tissue show elevated levels of cytoplasmic DNA, contributing to their increased interferon responsiveness. Human beta-cell senescence thus involves chromatin-driven upregulation of a functional-maturation program, and increased responsiveness of interferon-stimulated genes, changes that could increase both insulin secretion and immune reactivity., (© The Author(s) 2024. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2024
- Full Text
- View/download PDF
44. IFN-signaling gene expression as a diagnostic biomarker for monogenic interferonopathies.
- Author
-
Adang LA, D'Aiello R, Takanohashi A, Woidill S, Gavazzi F, Behrens EM, Sullivan KE, Goldbach-Mansky R, de Jesus AA, Vanderver A, and Shults J
- Subjects
- Humans, Male, Female, Child, Interferons genetics, Interferons metabolism, Ubiquitin Thiolesterase genetics, Child, Preschool, Interferon Type I genetics, Interferon Type I metabolism, Membrane Proteins genetics, Adult, Adolescent, RNA, Messenger metabolism, RNA, Messenger genetics, Tumor Suppressor Proteins, Biomarkers metabolism, Nervous System Malformations genetics, Nervous System Malformations diagnosis, Signal Transduction genetics, Autoimmune Diseases of the Nervous System genetics, Autoimmune Diseases of the Nervous System diagnosis
- Abstract
IFN-signaling gene (ISG) expression scores are potential markers of inflammation with significance from cancer to genetic syndromes. In Aicardi Goutières Syndrome (AGS), a disorder of abnormal DNA and RNA metabolism, this score has potential as a diagnostic biomarker, although the approach to ISG calculation has not been standardized or validated. To optimize ISG calculation and validate ISG as a diagnostic biomarker, mRNA levels of 36 type I IFN response genes were quantified from 997 samples (including 334 AGS), and samples were randomized into training and test data sets. An independent validation cohort (n = 122) was also collected. ISGs were calculated using all potential combinations up to 6 genes. A 4-gene approach (IFI44L, IFI27, USP18, IFI6) was the best-performing model (AUC of 0.8872 [training data set], 0.9245 [test data set]). The majority of top-performing gene combinations included IFI44L. Performance of IFI44L alone was 0.8762 (training data set) and 0.9580 (test data set) by AUC. The top approaches were able to discriminate individuals with genetic interferonopathy from control samples. This study validates the context of use for the ISG score as a diagnostic biomarker and underscores the importance of IFI44L in diagnosis of genetic interferonopathies.
- Published
- 2024
- Full Text
- View/download PDF
45. Transcriptome Analysis in Air-Liquid Interface Porcine Respiratory Epithelial Cell Cultures Reveals That the Betacoronavirus Porcine Encephalomyelitis Hemagglutinating Virus Induces a Robust Interferon Response to Infection.
- Author
-
Davila KMS, Nelli RK, Mora-Díaz JC, Sang Y, Miller LC, and Giménez-Lirola LG
- Subjects
- Animals, Swine, Immunity, Innate, Virus Replication, Coronavirus Infections immunology, Coronavirus Infections virology, Coronavirus Infections veterinary, Cytokines metabolism, Cytokines genetics, Cytokines immunology, Transcriptome, Respiratory Mucosa virology, Respiratory Mucosa immunology, Swine Diseases virology, Swine Diseases immunology, Swine Diseases genetics, Cells, Cultured, Deltacoronavirus, Gene Expression Profiling, Epithelial Cells virology, Epithelial Cells immunology, Interferons genetics, Interferons metabolism, Interferons immunology, Betacoronavirus 1 immunology, Betacoronavirus 1 genetics
- Abstract
Porcine hemagglutinating encephalomyelitis virus (PHEV) replicates in the upper respiratory tract and tonsils of pigs. Using an air-liquid interface porcine respiratory epithelial cells (ALI-PRECs) culture system, we demonstrated that PHEV disrupts respiratory epithelia homeostasis by impairing ciliary function and inducing antiviral, pro-inflammatory cytokine, and chemokine responses. This study explores the mechanisms driving early innate immune responses during PHEV infection through host transcriptome analysis. Total RNA was collected from ALI-PRECs at 24, 36, and 48 h post inoculation (hpi). RNA-seq analysis was performed using an Illumina Hiseq 600 to generate 100 bp paired-end reads. Differential gene expression was analyzed using DeSeq2. PHEV replicated actively in ALI-PRECs, causing cytopathic changes and progressive mucociliary disruption. Transcriptome analysis revealed downregulation of cilia-associated genes such as CILK1 , DNAH11 , LRRC-23 , -49 , and -51 , and acidic sialomucin CD164L2 . PHEV also activated antiviral signaling pathways, significantly increasing the expression of interferon-stimulated genes ( RSAD2 , MX1 , IFIT , and ISG15 ) and chemokine genes ( CCL5 and CXCL10 ), highlighting inflammatory regulation. This study contributes to elucidating the molecular mechanisms of the innate immune response to PHEV infection of the airway epithelium, emphasizing the critical roles of the mucociliary, interferon, and chemokine responses.
- Published
- 2024
- Full Text
- View/download PDF
46. Interferon-Regulated Expression of Cellular Splicing Factors Modulates Multiple Levels of HIV-1 Gene Expression and Replication.
- Author
-
Roesmann F, Müller L, Klaassen K, Heß S, and Widera M
- Subjects
- Humans, Gene Expression Regulation, Viral, RNA Splicing Factors metabolism, RNA Splicing Factors genetics, Interferon Type I metabolism, Interferon Type I genetics, Host-Pathogen Interactions immunology, Host-Pathogen Interactions genetics, Interferons metabolism, Interferons genetics, Interferons immunology, RNA-Binding Proteins genetics, RNA-Binding Proteins metabolism, HIV-1 genetics, HIV-1 physiology, Virus Replication, HIV Infections virology, HIV Infections genetics, HIV Infections immunology, Immunity, Innate
- Abstract
Type I interferons (IFN-Is) are pivotal in innate immunity against human immunodeficiency virus I (HIV-1) by eliciting the expression of IFN-stimulated genes (ISGs), which encompass potent host restriction factors. While ISGs restrict the viral replication within the host cell by targeting various stages of the viral life cycle, the lesser-known IFN-repressed genes (IRepGs), including RNA-binding proteins (RBPs), affect the viral replication by altering the expression of the host dependency factors that are essential for efficient HIV-1 gene expression. Both the host restriction and dependency factors determine the viral replication efficiency; however, the understanding of the IRepGs implicated in HIV-1 infection remains greatly limited at present. This review provides a comprehensive overview of the current understanding regarding the impact of the RNA-binding protein families, specifically the two families of splicing-associated proteins SRSF and hnRNP, on HIV-1 gene expression and viral replication. Since the recent findings show specifically that SRSF1 and hnRNP A0 are regulated by IFN-I in various cell lines and primary cells, including intestinal lamina propria mononuclear cells (LPMCs) and peripheral blood mononuclear cells (PBMCs), we particularly discuss their role in the context of the innate immunity affecting HIV-1 replication.
- Published
- 2024
- Full Text
- View/download PDF
47. PR-SET7 epigenetically restrains uterine interferon response and cell death governing proper postnatal stromal development.
- Author
-
Bao H, Sun Y, Deng N, Zhang L, Jia Y, Li G, Gao Y, Li X, Tang Y, Cai H, Lu J, Wang H, Deng W, and Kong S
- Subjects
- Female, Animals, Mice, Interferons metabolism, Interferons genetics, Endogenous Retroviruses genetics, Apoptosis genetics, Mice, Inbred C57BL, Cell Death genetics, Necroptosis genetics, RNA-Binding Proteins metabolism, RNA-Binding Proteins genetics, Histones metabolism, Single-Cell Analysis, Mice, Knockout, Cell Differentiation genetics, Uterus metabolism, Stromal Cells metabolism, Epigenesis, Genetic, Histone-Lysine N-Methyltransferase metabolism, Histone-Lysine N-Methyltransferase genetics
- Abstract
The differentiation of the stroma is a hallmark event during postnatal uterine development. However, the spatiotemporal changes that occur during this process and the underlying regulatory mechanisms remain elusive. Here, we comprehensively delineated the dynamic development of the neonatal uterus at single-cell resolution and characterized two distinct stromal subpopulations, inner and outer stroma. Furthermore, single-cell RNA sequencing revealed that uterine ablation of Pr-set7, the sole methyltransferase catalyzing H4K20me1, led to a reduced proportion of the inner stroma due to massive cell death, thus impeding uterine development. By combining RNA sequencing and epigenetic profiling of H4K20me1, we demonstrated that PR-SET7-H4K20me1 either directly repressed the transcription of interferon stimulated genes or indirectly restricted the interferon response via silencing endogenous retroviruses. Declined H4K20me1 level caused viral mimicry responses and ZBP1-mediated apoptosis and necroptosis in stromal cells. Collectively, our study provides insight into the epigenetic machinery governing postnatal uterine stromal development mediated by PR-SET7., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
48. Interferon-Stimulated Genes that Target Retrovirus Translation.
- Author
-
Jäger N, Pöhlmann S, Rodnina MV, and Ayyub SA
- Subjects
- Humans, Immunity, Innate, Animals, Signal Transduction, Retroviridae Infections virology, Retroviridae Infections immunology, Retroviridae Infections genetics, Interferons immunology, Interferons metabolism, Interferons genetics, Retroviridae genetics, Retroviridae physiology, Protein Biosynthesis
- Abstract
The innate immune system, particularly the interferon (IFN) system, constitutes the initial line of defense against viral infections. IFN signaling induces the expression of interferon-stimulated genes (ISGs), and their products frequently restrict viral infection. Retroviruses like the human immunodeficiency viruses and the human T-lymphotropic viruses cause severe human diseases and are targeted by ISG-encoded proteins. Here, we discuss ISGs that inhibit the translation of retroviral mRNAs and thereby retrovirus propagation. The Schlafen proteins degrade cellular tRNAs and rRNAs needed for translation. Zinc Finger Antiviral Protein and RNA-activated protein kinase inhibit translation initiation factors, and Shiftless suppresses translation recoding essential for the expression of retroviral enzymes. We outline common mechanisms that underlie the antiviral activity of multifunctional ISGs and discuss potential antiretroviral therapeutic approaches based on the mode of action of these ISGs.
- Published
- 2024
- Full Text
- View/download PDF
49. hnRNPM protects against the dsRNA-mediated interferon response by repressing LINE-associated cryptic splicing.
- Author
-
Zheng R, Dunlap M, Bobkov GOM, Gonzalez-Figueroa C, Patel KJ, Lyu J, Harvey SE, Chan TW, Quinones-Valdez G, Choudhury M, Le Roux CA, Bartels MD, Vuong A, Flynn RA, Chang HY, Van Nostrand EL, Xiao X, and Cheng C
- Subjects
- Humans, Interferons metabolism, Interferons genetics, Animals, HEK293 Cells, Mice, Transcriptome, Exons, RNA Splice Sites, Alu Elements genetics, Heterogeneous-Nuclear Ribonucleoprotein Group M genetics, Heterogeneous-Nuclear Ribonucleoprotein Group M metabolism, RNA, Double-Stranded genetics, RNA, Double-Stranded metabolism, Introns, RNA Splicing, Long Interspersed Nucleotide Elements genetics
- Abstract
RNA splicing is pivotal in post-transcriptional gene regulation, yet the exponential expansion of intron length in humans poses a challenge for accurate splicing. Here, we identify hnRNPM as an essential RNA-binding protein that suppresses cryptic splicing through binding to deep introns, maintaining human transcriptome integrity. Long interspersed nuclear elements (LINEs) in introns harbor numerous pseudo splice sites. hnRNPM preferentially binds at intronic LINEs to repress pseudo splice site usage for cryptic splicing. Remarkably, cryptic exons can generate long dsRNAs through base-pairing of inverted ALU transposable elements interspersed among LINEs and consequently trigger an interferon response, a well-known antiviral defense mechanism. Significantly, hnRNPM-deficient tumors show upregulated interferon-associated pathways and elevated immune cell infiltration. These findings unveil hnRNPM as a guardian of transcriptome integrity by repressing cryptic splicing and suggest that targeting hnRNPM in tumors may be used to trigger an inflammatory immune response, thereby boosting cancer surveillance., Competing Interests: Declaration of interests R.A.F. is a co-founder, board of directors member, and stockholder of GanNA Bio and a board of directors member and stockholder of Chronus Health. H.Y.C. is a co-founder of Accent Therapeutics, Boundless Bio, Cartography Biosciences, and Orbital Therapeutics, an advisor of 10× Genomics, Arsenal Biosciences, Chroma Medicine, and Spring Discovery, and a member of the Molecular Cell advisory board. E.L.V.N. is a co-founder, member of the board of directors, on the SAB, an equity holder, and a paid consultant for Eclipse BioInnovations, on the SAB of RNAConnect, and an inventor of intellectual property owned by University of California, San Diego. E.L.V.N.’s interests have been reviewed and approved by the Baylor College of Medicine in accordance with its conflict-of-interest policies., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
50. SFTSV nucleoprotein mediates DNA sensor cGAS degradation to suppress cGAS-dependent antiviral responses.
- Author
-
Jiang Z-z, Chu M, Yan L-n, Zhang W-k, Li B, Xu J, Zhao Z-x, Han H-J, Zhou C-m, and Yu X-j
- Subjects
- Humans, HEK293 Cells, Severe Fever with Thrombocytopenia Syndrome virology, Severe Fever with Thrombocytopenia Syndrome immunology, Severe Fever with Thrombocytopenia Syndrome metabolism, Autophagy, Animals, DNA, Mitochondrial genetics, DNA, Mitochondrial metabolism, Interferons metabolism, Interferons immunology, Interferons genetics, Viral Proteins metabolism, Viral Proteins genetics, Phlebovirus genetics, Phlebovirus immunology, Nucleotidyltransferases metabolism, Nucleotidyltransferases genetics, Immunity, Innate, Nucleoproteins metabolism, Nucleoproteins genetics, Nucleoproteins immunology
- Abstract
Cyclic GMP-AMP synthase (cGAS) is an important DNA pattern recognition receptor that senses double-stranded DNA derived from invading pathogens or self DNA in cytoplasm, leading to an antiviral interferon response. A tick-borne Bunyavirus, severe fever with thrombocytopenia syndrome virus (SFTSV), is an RNA virus that causes a severe emerging viral hemorrhagic fever in Asia with a high case fatality rate of up to 30%. However, it is unclear whether cGAS interacts with SFTSV infection. In this study, we found that SFTSV infection upregulated cGAS RNA transcription and protein expression, indicating that cGAS is an important innate immune response against SFTSV infection. The mechanism of cGAS recognizing SFTSV is by cGAS interacting with misplaced mitochondrial DNA in the cytoplasm. Depletion of mitochondrial DNA significantly inhibited cGAS activation under SFTSV infection. Strikingly, we found that SFTSV nucleoprotein (N) induced cGAS degradation in a dose-dependent manner. Mechanically, N interacted with the 161-382 domain of cGAS and linked the cGAS to LC3. The cGAS-N-LC3 trimer was targeted to N-induced autophagy, and the cGAS was degraded in autolysosome. Taken together, our study discovered a novel antagonistic mechanism of RNA viruses, SFTSV is able to suppress the cGAS-dependent antiviral innate immune responses through N-hijacking cGAS into N-induced autophagy. Our results indicated that SFTSV N is an important virulence factor of SFTSV in mediating host antiviral immune responses., Importance: Severe fever with thrombocytopenia syndrome virus (SFTSV) is a tick-borne RNA virus that is widespread in East and Southeast Asian countries with a high fatality rate of up to 30%. Up to now, many cytoplasmic pattern recognition receptors, such as RIG-I, MDA5, and SAFA, have been reported to recognize SFTSV genomic RNA and trigger interferon-dependent antiviral responses. However, current knowledge is not clear whether SFTSV can be recognized by DNA sensor cyclic GMP-AMP synthase (cGAS). Our study demonstrated that cGAS could recognize SFTSV infection via ectopic mitochondrial DNA, and the activated cGAS-stimulator of interferon genes signaling pathway could significantly inhibit SFTSV replication. Importantly, we further uncovered a novel mechanism of SFTSV to inhibit innate immune responses by the degradation of cGAS. cGAS was degraded in N-induced autophagy. Collectively, this study illustrated a novel virulence factor of SFTSV to suppress innate immune responses through autophagy-dependent cGAS degradation., Competing Interests: The authors declare no conflict of interest.
- Published
- 2024
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.