1
|
Leya M, Jeong H, Yang D, Ton Nu Bao TH, Pandeya PR, Oh SI, Roh YS, Kim JW, Kim B. Hepatocyte-Specific Casein Kinase 1 Epsilon Ablation Ameliorates Metabolic Dysfunction-Associated Steatohepatitis by Up-Regulating Tumor Necrosis Factor Receptor-Associated Factor 3 in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:2106-2127. [PMID: 39179201 DOI: 10.1016/j.ajpath.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/09/2024] [Accepted: 08/02/2024] [Indexed: 08/26/2024]
Abstract
Casein kinase 1 epsilon (CK1ε), a member of the serine/threonine protein kinase family, phosphorylates a broad range of substrates. However, its role in the development of chronic liver diseases remains elusive. This study aimed to investigate the role of CK1ε in the development and progression of metabolic dysfunction-associated steatohepatitis (MASH). Hepatocyte-specific CK1ε knockout (CK1εΔHEP) mice were generated by crossbreeding mice with floxed CK1ε alleles (CK1εfl/fl) and Cre-expressing albumin mice. Mice were fed either a Western diet (WD) or a methionine- and choline-deficient diet to induce MASH. CK1εΔHEP was associated with a decreased severity of WD- or methionine- and choline-deficient diet-induced MASH, as confirmed by reduced incidence of hepatic lesions and significantly lower levels of alanine aminotransferase, aspartate aminotransferase, and proinflammatory cytokine tumor necrosis factor (TNF)-α. CK1εΔHEP WD-fed mice exhibited significant amelioration of total cholesterol, triglycerides, and de novo lipogenic genes, indicating that CK1ε could influence lipid metabolism. CK1εΔHEP WD-fed mice showed significantly down-regulated TNF receptor-associated factor (TRAF) 3, phosphorylated (p) transforming growth factor-β-activated kinase 1, p-TRAF-associated NF-κB activator (TANK)-binding kinase 1 (TBK1), and p-AKT levels, thereby affecting downstream mitogen-activated protein kinase signaling, indicating a potential mechanism for the observed rescue. Finally, pharmacologic inhibition of CK1ε with PF670462 improved palmitic acid-induced steatohepatitis in vitro and attenuated WD-induced metabolic profile in vivo. In conclusion, CK1ε up-regulates TNF receptor-associated factor 3, which, in turn, causes transforming growth factor-β-activated kinase 1-dependent signaling, amplifies downstream mitogen-activated protein kinase signaling, modifies p-c-Jun levels, and exacerbates inflammation, all of which are factors in WD-induced metabolic dysfunction-associated steatotic liver disease.
Collapse
Affiliation(s)
- Mwense Leya
- Biosafety Research Institute and College of Veterinary Medicine, Jeonbuk National University, Iksan-si, Republic of Korea; School of Veterinary Medicine, University of Namibia, Windhoek, Namibia
| | - Hyuneui Jeong
- Biosafety Research Institute and College of Veterinary Medicine, Jeonbuk National University, Iksan-si, Republic of Korea
| | - Daram Yang
- Biosafety Research Institute and College of Veterinary Medicine, Jeonbuk National University, Iksan-si, Republic of Korea
| | - Tien Huyen Ton Nu Bao
- Biosafety Research Institute and College of Veterinary Medicine, Jeonbuk National University, Iksan-si, Republic of Korea
| | - Prakash Raj Pandeya
- Department of Animal and Food Sciences, University of Kentucky, Lexington, Kentucky
| | - Sang-Ik Oh
- Biosafety Research Institute and College of Veterinary Medicine, Jeonbuk National University, Iksan-si, Republic of Korea
| | - Yoon-Seok Roh
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju-si, Republic of Korea
| | - Jong-Won Kim
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania.
| | - Bumseok Kim
- Biosafety Research Institute and College of Veterinary Medicine, Jeonbuk National University, Iksan-si, Republic of Korea.
| |
Collapse
|
2
|
Zou Y, Zhou Y, Chen Z, Zou P, Zhu Y, Zhang J, Zhang Z, Wang Y. Members of the TRAF gene family in Octopus sinensis and their response to PGN, poly I:C, and Vibrio parahaemolyticus. FISH & SHELLFISH IMMUNOLOGY 2024; 154:109905. [PMID: 39276812 DOI: 10.1016/j.fsi.2024.109905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 08/29/2024] [Accepted: 09/12/2024] [Indexed: 09/17/2024]
Abstract
Octopus sinensis, the species of Cephalopoda, is known as the highest Mollusca and is an economic and new aquaculture species in the coastal waters of southern China. The immune system has been well documented to have a function of resisting the invasion of pathogens in the external environment among mollusca species. As a kind of signaling molecule in the innate immune system, tumor necrosis factor (TNF) receptor-associated factor (TRAF) plays significant roles in TNF receptor (TNFR)/interleukin-1 receptor (IL-1R)/Toll-like receptor (TLR) signaling pathways. Until now, seven TRAF members (TRAF1-7) have been discovered, and they have been reported to participate in regulating signal pathways mediated by pattern recognition receptors and play important roles in the innate immune response of the hosts. In this study, five TRAF genes of O. sinensis (OsTRAF2, OsTRAF3, OsTRAF4, OsTRAF6, and OsTRAF7) were identified, whose full length of the open reading frame is 1473 bp, 1629 bp, 1431 bp, 1353 bp and 2121 bp respectively, encoding 490, 542, 476, 450 and 706 amino acids, respectively. Bioinformatics analysis showed that each OsTRAF has different chromosome locations. In addition to seven consecutive WD40 domains on the C-terminal of OsTRAF7 protein, the C-terminal of OsTRAF proteins all contain a conserved TRAF domain, namely the MATH domain. Phylogenetic analysis showed that OsTRAF proteins were clustered together with TRAF proteins of bivalves. Moreover, TRAF1 and TRAF2, TRAF3 and TRAF5 were clustered together in a large clade, respectively, revealing they have a close genetic relationship. The results of quantitative Real-time PCR showed that OsTRAF genes were highly expressed in the gill, hepatopancreas and white body. After stimulation with PGN, poly I:C and V. parahaemolyticus, the expression levels of OsTRAF genes were up-regulated in the gill, hepatopancreas and white body at different time points. These results indicated that OsTRAF genes play an important role in the antibacterial and antiviral immune response of O. sinensis.
Collapse
Affiliation(s)
- Yihua Zou
- State Key Laboratory of Mariculture Breeding, Fisheries College, Jimei University, Xiamen, 361021, China; Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen, 361021, China
| | - Yuquan Zhou
- State Key Laboratory of Mariculture Breeding, Fisheries College, Jimei University, Xiamen, 361021, China; Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen, 361021, China
| | - Zebin Chen
- State Key Laboratory of Mariculture Breeding, Fisheries College, Jimei University, Xiamen, 361021, China; Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen, 361021, China
| | - Pengfei Zou
- State Key Laboratory of Mariculture Breeding, Fisheries College, Jimei University, Xiamen, 361021, China; Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen, 361021, China
| | - Youfang Zhu
- Putian Municipal Institute of Fishery Science, Putian, 351100, China
| | - Jianming Zhang
- Putian Municipal Institute of Fishery Science, Putian, 351100, China
| | - Ziping Zhang
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China.
| | - Yilei Wang
- State Key Laboratory of Mariculture Breeding, Fisheries College, Jimei University, Xiamen, 361021, China; Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture and Rural Affairs, Fisheries College, Jimei University, Xiamen, 361021, China.
| |
Collapse
|
3
|
Fan H, Song C, Zhang J. Sarm1 Controls the MYD88-Mediated Inflammatory Responses in Inflammatory Bowel Disease via the Regulation of TRAF3 Recruitment. Immunol Invest 2024; 53:800-812. [PMID: 38651786 DOI: 10.1080/08820139.2024.2343889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
BACKGROUND Sterile alpha and TIR motif-containing 1 (Sarm1) is known as a negative regulator of inflammatory responses. However, its role in inflammatory bowel disease (IBD) is still unclear. OBJECTIVE This study aimed to explore the function of Sarm1 in IBD and its underlying mechanisms. Sarm1 and tumor necrosis factor (TNF) receptor associated factor 3 (TRAF3) knockout (KO) micewere established. METHODS The colitis was induced using dextran sulfate sodium (DSS). Bone marrow-derived macrophages (BMDMs) were isolated and stimulated with lipopolysaccharides (LPS) or cytidine phosphate guanosine(CpG). Inflammatory cytokines were measured viaELISA. qPCR and Western blotting were used to determine the levels of the mRNA and protein expression, respectively. RESULTS It was demonstrated that reduced expression of Sarm1 was correlated with the severity of IBD in ulcerative colitis patients, and also with the reduction of pro-inflammatory cytokines in the mouse model induced by DSS. It was further observed that Sarm1 KO enhanced the induction of pro-inflammatory cytokines in both animal and in vitro cell models. Sarm1 deficiency in macrophages increased the severity of colitis in the mouse model induced by DSS. Moreover, Sarm1 regulatedTRAF3 recruitment to myeloid differentiation primary response protein 88 (MyD88), which in turn controlled the MYD88-mediated inflammatory responses. CONCLUSIONS In summary, our data suggest that Sarm1 controls the MYD88-mediated inflammatory responses in IBD via its regulation of TRAF3 recruitment.
Collapse
Affiliation(s)
- Huijuan Fan
- Department of Gastroenterology, The Second Affiliated Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Chun Song
- Department of Gastroenterology, The Second Affiliated Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Jingyu Zhang
- Department of Gastroenterology, The Second Affiliated Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
4
|
Duan Z, Zhang F, Wang X, Li H, Zhou D, Chen Q, Tao Z, Chen Z, Yu G, Yu H. C-type lectin 12B/4E of black rockfish (Sebastes schlegelii) macrophages as pattern recognition receptors in the antibacterial mechanism of exploration. FISH & SHELLFISH IMMUNOLOGY 2024; 150:109636. [PMID: 38762095 DOI: 10.1016/j.fsi.2024.109636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 05/20/2024]
Abstract
As lower vertebrates, fish have both innate and adaptive immune systems, but the role of the adaptive immune system is limited, and the innate immune system plays an important role in the resistance to pathogen infection. C-type lectins (CLRs) are one of the major pattern recognition receptors (PRRs) of the innate immune system. CLRs can combine with pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs) to trigger NF-κB signaling pathway and exert immune efficacy. In this study, Ssclec12b and Ssclec4e of the C-type lectins, were found to be significantly up-regulated in the transcripts of Sebastes schlegelii macrophages stimulated by bacteria. The identification, expression and function of these lectins were studied. In addition, the recombinant proteins of the above two CLRs were obtained by prokaryotic expression. We found that rSsCLEC12B and rSsCLEC4E could bind to a variety of bacteria in a Ca2+-dependent manner, and promoted the agglutination of bacteria and blood cells. rSsCLEC12B and rSsCLEC4E assisted macrophages to recognize PAMPs and activate the NF-κB signaling pathway, thereby promoting the expression of inflammatory factors (TNF-α, IL-1β, IL-6, IL-8) and regulating the early immune inflammation of macrophages. These results suggested that SsCLEC12B and SsCLEC4E could serve as PRRs in S. schlegelii macrophages to recognize pathogens and participate in the host antimicrobial immune process, and provided a valuable reference for the study of CLRs involved in fish innate immunity.
Collapse
Affiliation(s)
- Zhixiang Duan
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education, Ocean University of China, 266003, Qingdao, Shandong, China
| | - Fan Zhang
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education, Ocean University of China, 266003, Qingdao, Shandong, China
| | - Xuangang Wang
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education, Ocean University of China, 266003, Qingdao, Shandong, China
| | - Hengshun Li
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education, Ocean University of China, 266003, Qingdao, Shandong, China
| | - Dianyang Zhou
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education, Ocean University of China, 266003, Qingdao, Shandong, China
| | - Qiannan Chen
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education, Ocean University of China, 266003, Qingdao, Shandong, China
| | - Ze Tao
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education, Ocean University of China, 266003, Qingdao, Shandong, China
| | - Zhentao Chen
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education, Ocean University of China, 266003, Qingdao, Shandong, China
| | - Gan Yu
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education, Ocean University of China, 266003, Qingdao, Shandong, China
| | - Haiyang Yu
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education, Ocean University of China, 266003, Qingdao, Shandong, China.
| |
Collapse
|
5
|
Gao K, Liu M, Tang H, Ma Z, Pan H, Zhang X, Inam M, Shan X, Gao Y, Wang G. Downregulation of miR-1388 Regulates the Expression of Antiviral Genes via Tumor Necrosis Factor Receptor ( TNFR)-Associated Factor 3 Targeting Following poly(I:C) Stimulation in Silver Carp ( Hypophthalmichthys molitrix). Biomolecules 2024; 14:694. [PMID: 38927097 PMCID: PMC11201635 DOI: 10.3390/biom14060694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/04/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
MicroRNAs (miRNAs) are highly conserved endogenous single-stranded non-coding RNA molecules that play a crucial role in regulating gene expression to maintain normal physiological functions in fish. Nevertheless, the specific physiological role of miRNAs in lower vertebrates, particularly in comparison to mammals, remains elusive. Additionally, the mechanisms underlying the control of antiviral responses triggered by viral stimulation in fish are still not fully understood. In this study, we investigated the regulatory impact of miR-1388 on the signaling pathway mediated by IFN regulatory factor 3 (IRF3). Our findings revealed that following stimulation with the viral analog poly(I:C), the expression of miR-1388 was significantly upregulated in primary immune tissues and macrophages. Through a dual luciferase reporter assay, we corroborated a direct targeting relationship between miR-1388 and tumor necrosis factor receptor (TNFR)-associated factor 3 (TRAF3). Furthermore, our study demonstrated a distinct negative post-transcriptional correlation between miR-1388 and TRAF3. We observed a significant negative post-transcriptional regulatory association between miR-1388 and the levels of antiviral genes following poly(I:C) stimulation. Utilizing reporter plasmids, we elucidated the role of miR-1388 in the antiviral signaling pathway activated by TRAF3. By intervening with siRNA-TRAF3, we validated that miR-1388 regulates the expression of antiviral genes and the production of type I interferons (IFN-Is) through its interaction with TRAF3. Collectively, our experiments highlight the regulatory influence of miR-1388 on the IRF3-mediated signaling pathway by targeting TRAF3 post poly(I:C) stimulation. These findings provide compelling evidence for enhancing our understanding of the mechanisms through which fish miRNAs participate in immune responses.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yunhang Gao
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China; (K.G.); (M.L.); (H.T.); (Z.M.); (H.P.); (X.Z.); (M.I.); (X.S.)
| | - Guiqin Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China; (K.G.); (M.L.); (H.T.); (Z.M.); (H.P.); (X.Z.); (M.I.); (X.S.)
| |
Collapse
|
6
|
Yang Z, Nicholas J. Biologically significant interaction of human herpesvirus 8 viral interferon regulatory factor 4 with ubiquitin-specific protease 7. J Virol 2024; 98:e0025524. [PMID: 38752725 PMCID: PMC11237418 DOI: 10.1128/jvi.00255-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/19/2024] [Indexed: 06/14/2024] Open
Abstract
Human herpesvirus 8 (HHV-8), associated with Kaposi sarcoma, primary effusion lymphoma (PEL), and multicentric Castleman disease, encodes four interferon regulatory factor homologs, vIRFs 1-4, that interact with and inhibit various mediators of host-cell defense against virus infection. A cellular protein targeted by all the vIRFs is ubiquitin-specific protease 7 (USP7); while replication-modulatory and latently infected PEL-cell pro-viability phenotypes of USP7 targeting have been identified for vIRFs 1-3, the significance of the interaction of vIRF-4 with USP7 has remained undetermined. Here we show, through genetic ablation of the vIRF-4-USP7 interaction in infected cells, that vIRF-4 association with USP7 is necessary for optimal expression of vIRF-4 and normal HHV-8 replication. Findings from experiments on transfected and infected cells identified ubiquitination of vIRF-4 via K48-linkage and USP7-binding-associated suppression of vIRF-4 ubiquitination and, in infected cells, increased vIRF-4 expression. Analysis of IFN-I induction and associated signaling as a function of vIRF-4 and its interaction with USP7 identified a role of each in innate-immune suppression. Finally, activation via K63-polyubiquitination of the innate-immune signaling mediator TRAF3 was found to be suppressed by vIRF-4 in a USP7-binding-associated manner in infected cells, but not in transfected cells, likely via binding-regulated expression of vIRF-4. Together, our data identify the first examples of vIRF ubiquitination and a vIRF substrate of USP7, enhanced expression of vIRF-4 via its interaction with USP7, and TRAF3-inhibitory activity of vIRF-4. The findings address, for the first time, the biological significance of the interaction of vIRF-4 with USP7 and reveal a mechanism of vIRF-4-mediated innate-immune evasion and pro-replication activity via TRAF3 regulation. IMPORTANCE HHV-8 homologs of cellular interferon regulatory factors (IRFs), involved in host-cell defense against virus infection, interact in an inhibitory fashion with IRFs and other mediators of antiviral innate immunity. These interactions are of demonstrated or hypothesized importance for successful primary, productive (lytic), and latent (persistent) infection by HHV-8. While HHV-8 vIRF-4 is known to interact physically with USP7 deubiquitinase, a key regulator of various cellular proteins, the functional and biological significance of the interaction has not been addressed. The present study identifies the interaction as important for HHV-8 productive replication and, indeed, for vIRF-4 expression and reveals a new function of vIRF-4 via inhibition of the activity of TRAF3, a pivotal mediator of host-cell antiviral activity through activation of cellular IRFs and induction of type-I interferons. These findings identify potential targets for the development of novel anti-HHV-8 agents, such as those able to disrupt vIRF-4-USP7 interaction or vIRF-4-stabilizing USP7 activity.
Collapse
Affiliation(s)
- Zunlin Yang
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - John Nicholas
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
7
|
Diniz CHDP, Henrique T, Stefanini ACB, De Castro TB, Tajara EH. Cetuximab chemotherapy resistance: Insight into the homeostatic evolution of head and neck cancer (Review). Oncol Rep 2024; 51:80. [PMID: 38639184 PMCID: PMC11056821 DOI: 10.3892/or.2024.8739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 04/03/2024] [Indexed: 04/20/2024] Open
Abstract
The complex evolution of genetic alterations in cancer that occurs in vivo is a selective process involving numerous factors and mechanisms. Chemotherapeutic agents that prevent the growth and spread of cancer cells induce selective pressure, leading to rapid artificial selection of resistant subclones. This rapid evolution is possible because antineoplastic drugs promote alterations in tumor‑cell metabolism, thus creating a bottleneck event. The few resistant cells that survive in this new environment obtain differential reproductive success that enables them to pass down the newly selected resistant gene pool. The present review aims to summarize key findings of tumor evolution, epithelial‑mesenchymal transition and resistance to cetuximab therapy in head and neck squamous cell carcinoma.
Collapse
Affiliation(s)
- Carlos Henrique De Paula Diniz
- Department of Molecular Biology, School of Medicine of São José do Rio Preto-FAMERP, São José do Rio Preto, São Paulo, SP 15090-000, Brazil
| | - Tiago Henrique
- Department of Molecular Biology, School of Medicine of São José do Rio Preto-FAMERP, São José do Rio Preto, São Paulo, SP 15090-000, Brazil
| | - Ana Carolina B. Stefanini
- Department of Molecular Biology, School of Medicine of São José do Rio Preto-FAMERP, São José do Rio Preto, São Paulo, SP 15090-000, Brazil
- Department of Experimental Research, Albert Einstein Education and Research Israeli Institute, IIEPAE, São Paulo, SP 05652-900, Brazil
| | - Tialfi Bergamin De Castro
- Department of Molecular Biology, School of Medicine of São José do Rio Preto-FAMERP, São José do Rio Preto, São Paulo, SP 15090-000, Brazil
- Microbial Pathogenesis Department, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA
| | - Eloiza H. Tajara
- Department of Molecular Biology, School of Medicine of São José do Rio Preto-FAMERP, São José do Rio Preto, São Paulo, SP 15090-000, Brazil
- Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, São Paulo, SP 05508-090, Brazil
| |
Collapse
|
8
|
Dupré J, Le Dimna M, Hutet E, Dujardin P, Fablet A, Leroy A, Fleurot I, Karadjian G, Roesch F, Caballero I, Bourry O, Vitour D, Le Potier MF, Caignard G. Exploring type I interferon pathway: virulent vs. attenuated strain of African swine fever virus revealing a novel function carried by MGF505-4R. Front Immunol 2024; 15:1358219. [PMID: 38529285 PMCID: PMC10961335 DOI: 10.3389/fimmu.2024.1358219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 02/15/2024] [Indexed: 03/27/2024] Open
Abstract
African swine fever virus represents a significant reemerging threat to livestock populations, as its incidence and geographic distribution have surged over the past decade in Europe, Asia, and Caribbean, resulting in substantial socio-economic burdens and adverse effects on animal health and welfare. In a previous report, we described the protective properties of our newly thermo-attenuated strain (ASFV-989) in pigs against an experimental infection of its parental Georgia 2007/1 virulent strain. In this new study, our objective was to characterize the molecular mechanisms underlying the attenuation of ASFV-989. We first compared the activation of type I interferon pathway in response to ASFV-989 and Georgia 2007/1 infections, employing both in vivo and in vitro models. Expression of IFN-α was significantly increased in porcine alveolar macrophages infected with ASFV-989 while pigs infected with Georgia 2007/1 showed higher IFN-α than those infected by ASFV-989. We also used a medium-throughput transcriptomic approach to study the expression of viral genes by both strains, and identified several patterns of gene expression. Subsequently, we investigated whether proteins encoded by the eight genes deleted in ASFV-989 contribute to the modulation of the type I interferon signaling pathway. Using different strategies, we showed that MGF505-4R interfered with the induction of IFN-α/β pathway, likely through interaction with TRAF3. Altogether, our data reveal key differences between ASFV-989 and Georgia 2007/1 in their ability to control IFN-α/β signaling and provide molecular mechanisms underlying the role of MGF505-4R as a virulence factor.
Collapse
Affiliation(s)
- Juliette Dupré
- Unité Mixte de Recherche (UMR) VIROLOGIE, Institut National Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), École Nationale Vétérinaire d’Alfort (ENVA), Agence Nationale de Sécurité Sanitaire de l'Alimentation, de l'Environnement et du Travail (ANSES) Laboratoire de Santé Animale, Université Paris-Est, Maisons-Alfort, France
- Unité Virologie Immunologie Porcines, Laboratoire de Ploufragan-Plouzané-Niort, ANSES, Ploufragan, France
| | - Mireille Le Dimna
- Unité Virologie Immunologie Porcines, Laboratoire de Ploufragan-Plouzané-Niort, ANSES, Ploufragan, France
| | - Evelyne Hutet
- Unité Virologie Immunologie Porcines, Laboratoire de Ploufragan-Plouzané-Niort, ANSES, Ploufragan, France
| | - Pascal Dujardin
- Unité Mixte de Recherche (UMR) VIROLOGIE, Institut National Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), École Nationale Vétérinaire d’Alfort (ENVA), Agence Nationale de Sécurité Sanitaire de l'Alimentation, de l'Environnement et du Travail (ANSES) Laboratoire de Santé Animale, Université Paris-Est, Maisons-Alfort, France
| | - Aurore Fablet
- Unité Mixte de Recherche (UMR) VIROLOGIE, Institut National Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), École Nationale Vétérinaire d’Alfort (ENVA), Agence Nationale de Sécurité Sanitaire de l'Alimentation, de l'Environnement et du Travail (ANSES) Laboratoire de Santé Animale, Université Paris-Est, Maisons-Alfort, France
| | - Aurélien Leroy
- UMR 1282 Infectiologie et santé publique (ISP), INRAE Centre Val de Loire, Nouzilly, France
| | - Isabelle Fleurot
- UMR 1282 Infectiologie et santé publique (ISP), INRAE Centre Val de Loire, Nouzilly, France
| | - Grégory Karadjian
- UMR Biologie moléculaire et Immunologie Parasitaires (BIPAR), ENVA-INRAE-ANSES, Laboratoire de Santé Animale, Maisons-Alfort, France
| | - Ferdinand Roesch
- UMR 1282 Infectiologie et santé publique (ISP), INRAE Centre Val de Loire, Nouzilly, France
| | - Ignacio Caballero
- UMR 1282 Infectiologie et santé publique (ISP), INRAE Centre Val de Loire, Nouzilly, France
| | - Olivier Bourry
- Unité Virologie Immunologie Porcines, Laboratoire de Ploufragan-Plouzané-Niort, ANSES, Ploufragan, France
| | - Damien Vitour
- Unité Mixte de Recherche (UMR) VIROLOGIE, Institut National Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), École Nationale Vétérinaire d’Alfort (ENVA), Agence Nationale de Sécurité Sanitaire de l'Alimentation, de l'Environnement et du Travail (ANSES) Laboratoire de Santé Animale, Université Paris-Est, Maisons-Alfort, France
| | - Marie-Frédérique Le Potier
- Unité Virologie Immunologie Porcines, Laboratoire de Ploufragan-Plouzané-Niort, ANSES, Ploufragan, France
| | - Grégory Caignard
- Unité Mixte de Recherche (UMR) VIROLOGIE, Institut National Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), École Nationale Vétérinaire d’Alfort (ENVA), Agence Nationale de Sécurité Sanitaire de l'Alimentation, de l'Environnement et du Travail (ANSES) Laboratoire de Santé Animale, Université Paris-Est, Maisons-Alfort, France
| |
Collapse
|
9
|
Duscher AA, Vroom MM, Foster JS. Impact of modeled microgravity stress on innate immunity in a beneficial animal-microbe symbiosis. Sci Rep 2024; 14:2912. [PMID: 38316910 PMCID: PMC10844198 DOI: 10.1038/s41598-024-53477-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 01/31/2024] [Indexed: 02/07/2024] Open
Abstract
The innate immune response is the first line of defense for all animals to not only detect invading microbes and toxins but also sense and interface with the environment. One such environment that can significantly affect innate immunity is spaceflight. In this study, we explored the impact of microgravity stress on key elements of the NFκB innate immune pathway. The symbiosis between the bobtail squid Euprymna scolopes and its beneficial symbiont Vibrio fischeri was used as a model system under a simulated microgravity environment. The expression of genes associated with the NFκB pathway was monitored over time as the symbiosis progressed. Results revealed that although the onset of the symbiosis was the major driver in the differential expression of NFκB signaling, the stress of simulated low-shear microgravity also caused a dysregulation of expression. Several genes were expressed at earlier time points suggesting that elements of the E. scolopes NFκB pathway are stress-inducible, whereas expression of other pathway components was delayed. The results provide new insights into the role of NFκB signaling in the squid-vibrio symbiosis, and how the stress of microgravity negatively impacts the host immune response. Together, these results provide a foundation to develop mitigation strategies to maintain host-microbe homeostasis during spaceflight.
Collapse
Affiliation(s)
- Alexandrea A Duscher
- Department of Microbiology and Cell Science, Space Life Science Lab, University of Florida, Merritt Island, FL, 32953, USA
- Chesapeake Bay Governor's School, Warsaw, VA, 22572, USA
| | - Madeline M Vroom
- Department of Microbiology and Cell Science, Space Life Science Lab, University of Florida, Merritt Island, FL, 32953, USA
- Vaxxinity, Space Life Sciences Lab, Merritt Island, FL, 32953, USA
| | - Jamie S Foster
- Department of Microbiology and Cell Science, Space Life Science Lab, University of Florida, Merritt Island, FL, 32953, USA.
| |
Collapse
|
10
|
Shang S, He D, Liu C, Bao X, Han S, Wang L. TRAF3 gene regulates macrophage migration and activation by lung epithelial cells infected with Aspergillus fumigatus. Microbiol Spectr 2024; 12:e0269923. [PMID: 38018974 PMCID: PMC10783100 DOI: 10.1128/spectrum.02699-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/21/2023] [Indexed: 11/30/2023] Open
Abstract
IMPORTANCE Aspergillus fumigatus can infect immunocompromised individuals and cause chronic and fatal invasive fungal infections. A better understanding of the molecular mechanisms of A. fumigatus-host interactions may provide new references for disease treatment. In this study, we demonstrated that the TRAF3 gene plays an important role in the early infection of A. fumigatus by regulating the resistance of lung epithelial cells to A. fumigatus. Macrophages are the most abundant innate immune cells in the alveoli; however, few studies have reported on the interactions between lung epithelial cells and macrophages in response to A. fumigatus invasion. In our study, it was demonstrated that the TRAF3 gene reduces migration to macrophages and cytokine production by negatively regulating lung epithelial cell adhesion and internalization of A. fumigatus spores. Together, our results provide new insights into lung epithelial cell-macrophage interactions during A. fumigatus infection.
Collapse
Affiliation(s)
- Shumi Shang
- Department of Pathogenobiology, Jilin University Mycology Research Center, Key Laboratory of Zoonosis Research, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Dan He
- Department of Pathogenobiology, Jilin University Mycology Research Center, Key Laboratory of Zoonosis Research, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Cong Liu
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xinyuan Bao
- Department of Pathogenobiology, Jilin University Mycology Research Center, Key Laboratory of Zoonosis Research, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Shuaishuai Han
- Beijing ZhongKai TianCheng Bio-technology Co. Ltd., Beijing, China
| | - Li Wang
- Department of Pathogenobiology, Jilin University Mycology Research Center, Key Laboratory of Zoonosis Research, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| |
Collapse
|
11
|
Hu H, Ma M, Li T, Shi L, Li P. The SMAC mimetic birinapant alleviates lipopolysaccharide-induced acute lung injury by inhibiting MAPK signaling. Immunol Lett 2023; 264:31-35. [PMID: 37913914 DOI: 10.1016/j.imlet.2023.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 10/19/2023] [Accepted: 10/28/2023] [Indexed: 11/03/2023]
Abstract
The second mitochondria-derived activator of caspases (SMAC) mimetic birinapant attenuated liver injury by inhibited the degradation of tumor necrosis factor receptor-associated factor 3 (TRAF3) and activation of mitogen-activated protein kinase (MAPK) signaling pathway in liver macrophage, but its role in LPS induced acute lung injury (ALI) is not understood. The present study was to investigate the effects of birinapant on ALI and its possible mechanism. A dose of birinapant (30 mg/kg) or a vehicle was administered intravenously 24 hours before LPS (100 μg) stimulation in mice. The levels of TNF-α, IL-6 and IL-1β in bronchoalveolar lavage fluid (BALF) were measured by ELISA. The infiltrated macrophages and expression of monocyte chemoattractant protein-1 (MCP-1) was determined by immunohistochemistry staining in the lung tissues. The JNK and p38 MAPK activation, protein expression and K48-linked polyubiquitination of TRAF3 were determined in alveolar macrophage cell line (MH-S cells) after 1μg/ml LPS stimulation. The results showed that the birinapant down-regulated the levels of TNF-α, IL-6 and IL-1β in the BALF. In addition, birinapant markedly inhibited macrophages infiltration and MCP-1 protein expression in lung tissues. At last, birinapant suppressed the MAPKsignaling pathway and K48-linked ubiquitinated degradation of TRAF3 in MH-S cells after LPS administration. In conclusion, the results proved that birinapant protected against LPS-induced ALI through inhibiting MAPK activation and K48-linked ubiquitination of TRAF3 in alveolar macrophages.
Collapse
Affiliation(s)
- Hui Hu
- Department of Traumatology, Chongqing University Central Hospital, Chongqing Emergency Medical Center, No.1 Jiankang Road, Yuzhong District, Chongqing, China
| | - Man Ma
- Department of Traumatology, Chongqing University Central Hospital, Chongqing Emergency Medical Center, No.1 Jiankang Road, Yuzhong District, Chongqing, China
| | - Tao Li
- Department of Traumatology, Chongqing University Central Hospital, Chongqing Emergency Medical Center, No.1 Jiankang Road, Yuzhong District, Chongqing, China
| | - Li Shi
- Department of Traumatology, Chongqing University Central Hospital, Chongqing Emergency Medical Center, No.1 Jiankang Road, Yuzhong District, Chongqing, China
| | - Peizhi Li
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
12
|
Tang H, Li H, Li D, Peng J, Zhang X, Yang W. The Gut Microbiota of Pregnant Rats Alleviates Fetal Growth Restriction by Inhibiting the TLR9/MyD88 Pathway. J Microbiol Biotechnol 2023; 33:1213-1227. [PMID: 37416999 PMCID: PMC10580896 DOI: 10.4014/jmb.2304.04020] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/02/2023] [Accepted: 06/14/2023] [Indexed: 07/08/2023]
Abstract
Fetal growth restriction (FGR) is a prevalent obstetric condition. This study aimed to investigate the role of Toll-like receptor 9 (TLR9) in regulating the inflammatory response and gut microbiota structure in FGR. An FGR animal model was established in rats, and ODN1668 and hydroxychloroquine (HCQ) were administered. Changes in gut microbiota structure were assessed using 16S rRNA sequencing, and fecal microbiota transplantation (FMT) was conducted. HTR-8/Svneo cells were treated with ODN1668 and HCQ to evaluate cell growth. Histopathological analysis was performed, and relative factor levels were measured. The results showed that FGR rats exhibited elevated levels of TLR9 and myeloid differentiating primary response gene 88 (MyD88). In vitro experiments demonstrated that TLR9 inhibited trophoblast cell proliferation and invasion. TLR9 upregulated lipopolysaccharide (LPS), LPS-binding protein (LBP), interleukin (IL)-1β and tumor necrosis factor (TNF)-α while downregulating IL-10. TLR9 activated the TARF3-TBK1-IRF3 signaling pathway. In vivo experiments showed HCQ reduced inflammation in FGR rats, and the relative cytokine expression followed a similar trend to that observed in vitro. TLR9 stimulated neutrophil activation. HCQ in FGR rats resulted in changes in the abundance of Eubacterium_coprostanoligenes_group at the family level and the abundance of Eubacterium_coprostanoligenes_group and Bacteroides at the genus level. TLR9 and associated inflammatory factors were correlated with Bacteroides, Prevotella, Streptococcus, and Prevotellaceae_Ga6A1_group. FMT from FGR rats interfered with the therapeutic effects of HCQ. In conclusion, our findings suggest that TLR9 regulates the inflammatory response and gut microbiota structure in FGR, providing new insights into the pathogenesis of FGR and suggesting potential therapeutic interventions.
Collapse
Affiliation(s)
- Hui Tang
- Department of Maternal and Child Health, Changsha Hospital for Maternal & Child Health Care Affiliated to Hunan Normal University, 416 Chengnan Dong Lu, Yuhua District, 410007, Changsha, Hunan, P.R. China
| | - Hanmei Li
- Department of Maternal and Child Health, Changsha Hospital for Maternal & Child Health Care Affiliated to Hunan Normal University, 416 Chengnan Dong Lu, Yuhua District, 410007, Changsha, Hunan, P.R. China
| | - Dan Li
- Department of Maternal and Child Health, Changsha Hospital for Maternal & Child Health Care Affiliated to Hunan Normal University, 416 Chengnan Dong Lu, Yuhua District, 410007, Changsha, Hunan, P.R. China
| | - Jing Peng
- Department of Maternal and Child Health, Changsha Hospital for Maternal & Child Health Care Affiliated to Hunan Normal University, 416 Chengnan Dong Lu, Yuhua District, 410007, Changsha, Hunan, P.R. China
| | - Xian Zhang
- Department of Maternal and Child Health, Changsha Hospital for Maternal & Child Health Care Affiliated to Hunan Normal University, 416 Chengnan Dong Lu, Yuhua District, 410007, Changsha, Hunan, P.R. China
| | - Weitao Yang
- Department of Maternal and Child Health, Changsha Hospital for Maternal & Child Health Care Affiliated to Hunan Normal University, 416 Chengnan Dong Lu, Yuhua District, 410007, Changsha, Hunan, P.R. China
| |
Collapse
|
13
|
Carman LE, Samulevich ML, Aneskievich BJ. Repressive Control of Keratinocyte Cytoplasmic Inflammatory Signaling. Int J Mol Sci 2023; 24:11943. [PMID: 37569318 PMCID: PMC10419196 DOI: 10.3390/ijms241511943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/17/2023] [Accepted: 07/20/2023] [Indexed: 08/13/2023] Open
Abstract
The overactivity of keratinocyte cytoplasmic signaling contributes to several cutaneous inflammatory and immune pathologies. An important emerging complement to proteins responsible for this overactivity is signal repression brought about by several proteins and protein complexes with the native role of limiting inflammation. The signaling repression by these proteins distinguishes them from transmembrane receptors, kinases, and inflammasomes, which drive inflammation. For these proteins, defects or deficiencies, whether naturally arising or in experimentally engineered skin inflammation models, have clearly linked them to maintaining keratinocytes in a non-activated state or returning cells to a post-inflamed state after a signaling event. Thus, together, these proteins help to resolve acute inflammatory responses or limit the development of chronic cutaneous inflammatory disease. We present here an integrated set of demonstrated or potentially inflammation-repressive proteins or protein complexes (linear ubiquitin chain assembly complex [LUBAC], cylindromatosis lysine 63 deubiquitinase [CYLD], tumor necrosis factor alpha-induced protein 3-interacting protein 1 [TNIP1], A20, and OTULIN) for a comprehensive view of cytoplasmic signaling highlighting protein players repressing inflammation as the needed counterpoints to signal activators and amplifiers. Ebb and flow of players on both sides of this inflammation equation would be of physiological advantage to allow acute response to damage or pathogens and yet guard against chronic inflammatory disease. Further investigation of the players responsible for repressing cytoplasmic signaling would be foundational to developing new chemical-entity pharmacologics to stabilize or enhance their function when clinical intervention is needed to restore balance.
Collapse
Affiliation(s)
- Liam E. Carman
- Graduate Program in Pharmacology & Toxicology, University of Connecticut, Storrs, CT 06269-3092, USA; (L.E.C.); (M.L.S.)
| | - Michael L. Samulevich
- Graduate Program in Pharmacology & Toxicology, University of Connecticut, Storrs, CT 06269-3092, USA; (L.E.C.); (M.L.S.)
| | - Brian J. Aneskievich
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT 06269-3092, USA
| |
Collapse
|
14
|
Zhong K, Liu X, Ding W, Peng L, Zeng X, Gu Y. TRAF inhibition drives cancer cell apoptosis and improves retinoic acid sensitivity in multiple cancers models. Discov Oncol 2023; 14:117. [PMID: 37389738 DOI: 10.1007/s12672-023-00703-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 05/26/2023] [Indexed: 07/01/2023] Open
Abstract
TNF receptor-associated factors (TRAFs) are signaling adaptor proteins that play a crucial role in regulating cellular receptors' signaling transduction to downstream pathways and exert multifaceted roles in regulating signaling pathways, cell survival, and carcinogenesis. The 13-cis-retinoic acid (RA), an active metabolite of vitamin A, exhibits anti-cancer properties, but the development of retinoic acid resistance poses a challenge in clinical application. This study aimed to investigate the relationship between TRAFs and retinoic acid sensitivity in various cancers. Here, we revealed that TRAFs' expression varied significantly across The Cancer Genome Atlas (TCGA) cancer cohorts and human cancer cell lines. Additionally, inhibiting TRAF4, TRAF5, or TRAF6 improved retinoic acid sensitivity and reduced colony formation in ovarian cancer and melanoma cells. Mechanistically, knocking down TRAF4, TRAF5, or TRAF6 in retinoic acid-treated cancer cell lines increased the levels of procaspase 9 and induced cell apoptosis. Further in vivo studies using the SK-OV-3 and MeWo xenograft models confirmed the anti-tumor effects of TRAF knockdown combined with retinoic acid treatment. These findings support that combination therapy with retinoic acid and TRAF silencing may offer significant therapeutic advantages in treating melanoma and ovarian cancers.
Collapse
Affiliation(s)
- Kun Zhong
- Medical School, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Xiaojun Liu
- Medical School, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Weihua Ding
- Medical School, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Lizhong Peng
- Medical School, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu Province, People's Republic of China
| | - Xuhui Zeng
- Medical School, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu Province, People's Republic of China.
| | - Yayun Gu
- Medical School, Nantong University, 19 Qixiu Road, Nantong, 226001, Jiangsu Province, People's Republic of China.
| |
Collapse
|
15
|
Chen HY, Tang RC, Liang JW, Zhao W, Yu SS, Yao RR, Xu R, Zhang A, Geng S, Sun XY, Ge Q, Zhang J. Deubiquitinase USP47 attenuates virus-induced type I interferon signaling. Int Immunopharmacol 2023; 118:110040. [PMID: 37001379 DOI: 10.1016/j.intimp.2023.110040] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 03/10/2023] [Accepted: 03/12/2023] [Indexed: 03/31/2023]
Abstract
The innate immune responses are tightly regulated to ensure effective clearance of invading pathogens and avoid excessive inflammation. Ubiquitination and deubiquitination are important post-translational modifications in antiviral immune responses. Here, we discovered deubiquitinase USP47 as a novel negative immune system regulator. Overexpression of USP47 repressed Sendai virus, poly(I:C) and poly(dA:dT)-induced ISRE and IFN-β activation, along with reduced IFNB1 transcription and enhanced viral replication. Knockdown of USP47 expression had the opposite effects. Dual-luciferase and phosphorylation assays showed that USP47 targeted downstream of MAVS and upstream of TBK1. Additional co-immunoprecipitation assays suggested that USP47 interacted with TRAF3 and TRAF6. Importantly, USP47 removed K63-linked polyubiquitin chains from TRAF3 and TRAF6. Hence, we describe a novel modulator of the antiviral innate immune response, USP47, which removes K63-linked polyubiquitins from TRAF3 and TRAF6, leading to reduced type I IFN signaling.
Collapse
Affiliation(s)
- Hong-Yan Chen
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology(Peking University), Peking University Health Science Center, Beijing 100191, China
| | - Rong-Chun Tang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology(Peking University), Peking University Health Science Center, Beijing 100191, China
| | - Jia-Wei Liang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology(Peking University), Peking University Health Science Center, Beijing 100191, China
| | - Weijia Zhao
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology(Peking University), Peking University Health Science Center, Beijing 100191, China
| | - Shuang-Shuang Yu
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology(Peking University), Peking University Health Science Center, Beijing 100191, China
| | - Ran-Ran Yao
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology(Peking University), Peking University Health Science Center, Beijing 100191, China
| | - Rui Xu
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology(Peking University), Peking University Health Science Center, Beijing 100191, China
| | - Ao Zhang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology(Peking University), Peking University Health Science Center, Beijing 100191, China
| | - Shijin Geng
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology(Peking University), Peking University Health Science Center, Beijing 100191, China
| | - Xiu-Yuan Sun
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology(Peking University), Peking University Health Science Center, Beijing 100191, China
| | - Qing Ge
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology(Peking University), Peking University Health Science Center, Beijing 100191, China
| | - Jun Zhang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology(Peking University), Peking University Health Science Center, Beijing 100191, China.
| |
Collapse
|
16
|
Tian WJ, Wang XJ. Broad-Spectrum Antivirals Derived from Natural Products. Viruses 2023; 15:v15051100. [PMID: 37243186 DOI: 10.3390/v15051100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
Scientific advances have led to the development and production of numerous vaccines and antiviral drugs, but viruses, including re-emerging and emerging viruses, such as SARS-CoV-2, remain a major threat to human health. Many antiviral agents are rarely used in clinical treatment, however, because of their inefficacy and resistance. The toxicity of natural products may be lower, and some natural products have multiple targets, which means less resistance. Therefore, natural products may be an effective means to solve virus infection in the future. New techniques and ideas are currently being developed for the design and screening of antiviral drugs thanks to recent revelations about virus replication mechanisms and the advancement of molecular docking technology. This review will summarize recently discovered antiviral drugs, mechanisms of action, and screening and design strategies for novel antiviral agents.
Collapse
Affiliation(s)
- Wen-Jun Tian
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, China Agricultural University, Beijing 100193, China
| | - Xiao-Jia Wang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, China Agricultural University, Beijing 100193, China
| |
Collapse
|