1
|
Su WC, Xia Y. Virus targeting as a dominant driver of interfacial evolution in the structurally resolved human-virus protein-protein interaction network. Cell Syst 2025; 16:101202. [PMID: 40023148 DOI: 10.1016/j.cels.2025.101202] [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: 08/05/2024] [Revised: 11/28/2024] [Accepted: 02/04/2025] [Indexed: 03/04/2025]
Abstract
Regions on a host protein that interact with virus proteins (exogenous interfaces) frequently overlap with those that interact with other host proteins (endogenous interfaces), resulting in competition between hosts and viruses for these shared interfaces (mimic-targeted interfaces). Yet, the evolutionary consequences of this competitive relationship on the host are not well understood. Here, we integrate experimentally determined structures and homology-based templates of protein complexes with protein-protein interaction networks to construct a high-resolution human-virus structural interaction network. We perform site-specific evolutionary rate analyses on this structural interaction network and find that exogenous-specific interfaces evolve faster than endogenous-specific interfaces. Mimic-targeted interfaces evolve as fast as exogenous-specific interfaces, despite being targeted by both human and virus proteins. Our findings suggest that virus targeting plays a dominant role in host interfacial evolution within the context of domain-domain interactions and that mimic-targeted interfaces on human proteins are the key battleground for a mammalian-specific host-virus evolutionary arms race.
Collapse
Affiliation(s)
- Wan-Chun Su
- Graduate Program in Quantitative Life Sciences, McGill University, Montréal, QC, Canada
| | - Yu Xia
- Graduate Program in Quantitative Life Sciences, McGill University, Montréal, QC, Canada; Department of Bioengineering, McGill University, Montréal, QC, Canada.
| |
Collapse
|
2
|
Nussinov R. Pioneer in Molecular Biology: Conformational Ensembles in Molecular Recognition, Allostery, and Cell Function. J Mol Biol 2025:169044. [PMID: 40015370 DOI: 10.1016/j.jmb.2025.169044] [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: 11/25/2024] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/01/2025]
Abstract
In 1978, for my PhD, I developed the efficient O(n3) dynamic programming algorithm for the-then open problem of RNA secondary structure prediction. This algorithm, now dubbed the "Nussinov algorithm", "Nussinov plots", and "Nussinov diagrams", is still taught across Europe and the U.S. As sequences started coming out in the 1980s, I started seeking genome-encoded functional signals, later becoming a bioinformatics trend. In the early 1990s I transited to proteins, co-developing a powerful computer vision-based docking algorithm. In the late 1990s, I proposed the foundational role of conformational ensembles in molecular recognition and allostery. At the time, conformational ensembles and free energy landscapes were viewed as physical properties of proteins but were not associated with function. The classical view of molecular recognition and binding was based on only two conformations captured by crystallography: open and closed. I proposed that all conformational states preexist. Proteins always have not one folded form-nor two-but many folded forms. Thus, rather than inducing fit, binding can work by shifting the ensembles between states, and this shifting, or redistributing the ensembles to maintain equilibrium, is the origin of the allosteric effect and protein, thus cell, function. This transformative paradigm impacted community views in allosteric drug design, catalysis, and regulation. Dynamic conformational ensemble shifts are now acknowledged as the origin of recognition, allostery, and signaling, underscoring that conformational ensembles-not proteins-are the workhorses of the cell, pioneering the fundamental idea that dynamic ensembles are the driving force behind cellular processes. Nussinov was recognized as pioneer in molecular biology by JMB.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD 21702, USA; Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
3
|
Wu S, Yao L, Zhang W, Chen P, Jiang J, Ma Y. Bioinformatics analysis and validation of novel biomarkers and competitive endogenous RNA networks involved in pyroptosis in diabetic nephropathy. Sci Rep 2025; 15:5530. [PMID: 39953123 PMCID: PMC11829041 DOI: 10.1038/s41598-025-87854-3] [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: 07/06/2024] [Accepted: 01/22/2025] [Indexed: 02/17/2025] Open
Abstract
Diabetic nephropathy (DN) is one of the major complications of diabetes mellitus. Pyroptosis is a type of programmed cell death that is closely related to the development of DN, however the molecular mechanism of pyroptosis in the development of DN is still unclear. The aim of this study is to identify pyroptosis-related potential biomarkers and competing endogenous RNA (ceRNA) networks in DN. The differentially expressed pyroptosis-related genes (DEPRGs) were identified using R software from Gene Expression Omnibus (GEO) database. In total, 4 significantly upregulated hub DEPRGs (CASP1, TXNIP, IRF9, and TRAF3) were selected and verified by machine learning techniques. Receiver Operating Curve (ROC) to assess the diagnostic value of pivotal DEPRGs. Immune infiltration was analysed using the CIBERSORT algorithm in R software. Then, differentially expressed miRNAs (DEmiRNAs) and lncRNAs (DElncRNAs) were obtained from the GEO database, respectively. The hub DEPRGs-associated ceRNA network was constructed. Finally, DN rats were induced by high-sugar and high-fat diet combined with an intraperitoneal injection of STZ. The expression of pyroptosis-related proteins and 4 hub DEPRGs were detected in rats' kidney tissues using Western blotting. The DN pyroptosis-related ceRNA networks constructed by hub genes were validated both in clinical samples and DN rat model using real-time PCR (qRT-PCR). Our results indicated that the ceRNA network consisting of key genes might be a potential regulatory axis for pyroptosis in DN.
Collapse
Affiliation(s)
- Siyu Wu
- College of Traditional Chinese Medicine, Xinjiang Medical University, Urumuqi, 830017, China
- Department of Pharmacy, General Hospital of Xin-Jiang Military Region, Urumqi, 830099, China
| | - Lan Yao
- College of Traditional Chinese Medicine, Xinjiang Medical University, Urumuqi, 830017, China.
| | - Wenxiang Zhang
- College of Traditional Chinese Medicine, Xinjiang Medical University, Urumuqi, 830017, China
| | - Pengde Chen
- College of Traditional Chinese Medicine, Xinjiang Medical University, Urumuqi, 830017, China
| | - Jie Jiang
- College of Traditional Chinese Medicine, Xinjiang Medical University, Urumuqi, 830017, China
| | - Yao Ma
- Department of Endocrinology, The Second Mercy Hospital of Xinjiang Uygur Autonomous Region, Urumuqi, 830013, China.
| |
Collapse
|
4
|
Woodfin S, Hall S, Ramerth A, Chapple B, Fausnacht D, Moore W, Alkhalidy H, Liu D. Potential Application of Plant-Derived Compounds in Multiple Sclerosis Management. Nutrients 2024; 16:2996. [PMID: 39275311 PMCID: PMC11397714 DOI: 10.3390/nu16172996] [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: 07/30/2024] [Revised: 08/23/2024] [Accepted: 08/29/2024] [Indexed: 09/16/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disorder characterized by inflammation, demyelination, and neurodegeneration, resulting in significant disability and reduced quality of life. Current therapeutic strategies primarily target immune dysregulation, but limitations in efficacy and tolerability highlight the need for alternative treatments. Plant-derived compounds, including alkaloids, phenylpropanoids, and terpenoids, have demonstrated anti-inflammatory effects in both preclinical and clinical studies. By modulating immune responses and promoting neuroregeneration, these compounds offer potential as novel adjunctive therapies for MS. This review provides insights into the molecular and cellular basis of MS pathogenesis, emphasizing the role of inflammation in disease progression. It critically evaluates emerging evidence supporting the use of plant-derived compounds to attenuate inflammation and MS symptomology. In addition, we provide a comprehensive source of information detailing the known mechanisms of action and assessing the clinical potential of plant-derived compounds in the context of MS pathogenesis, with a focus on their anti-inflammatory and neuroprotective properties.
Collapse
Affiliation(s)
- Seth Woodfin
- Department of Biology and Chemistry, School of Health Sciences, Liberty University, Lynchburg, VA 24515, USA
| | - Sierra Hall
- Department of Biology and Chemistry, School of Health Sciences, Liberty University, Lynchburg, VA 24515, USA
| | - Alexis Ramerth
- Department of Biology and Chemistry, School of Health Sciences, Liberty University, Lynchburg, VA 24515, USA
| | - Brooke Chapple
- Department of Biology and Chemistry, School of Health Sciences, Liberty University, Lynchburg, VA 24515, USA
| | - Dane Fausnacht
- Department of Biology, School of Sciences and Agriculture, Ferrum College, Ferrum, VA 24088, USA
| | - William Moore
- Department of Biology and Chemistry, School of Health Sciences, Liberty University, Lynchburg, VA 24515, USA
| | - Hana Alkhalidy
- Department of Human Nutrition, Foods and Exercise, College of Agriculture and Life Sciences, Virginia Tech, Blacksburg, VA 24061, USA
- Department of Nutrition and Food Technology, Faculty of Agriculture, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan
| | - Dongmin Liu
- Department of Human Nutrition, Foods and Exercise, College of Agriculture and Life Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| |
Collapse
|
5
|
Kalita E, Panda M, Rao A, Pandey RK, Prajapati VK. Viral mimicry and endocrine system: Divulging the importance in host-microbial crosstalk. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 142:421-436. [PMID: 39059993 DOI: 10.1016/bs.apcsb.2024.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
Host-pathogen interactions are complex associations which evolve over long co-evolutionary histories. Pathogens exhibit different mechanisms to gain advantage over their host. Mimicry of host factors is an influential tool in subverting host mechanisms to ensure pathogenesis. This chapter discusses such molecular mimicry exhibited during viral infections. Understanding the evolutionary relationships, shared identity and functional impact of the virus encoded mimics is critical. With a particular emphasis on viral mimics and their association with cancer and autoimmune diseases, this chapter highlights the importance of molecular mimicry in virus biology.
Collapse
Affiliation(s)
- Elora Kalita
- Life Science Research Centre, Faculty of Science, University of Ostrava, Ostrava, Czechia
| | - Mamta Panda
- Department of Neurology, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf Martinistraße, Hamburg, Germany
| | - Abhishek Rao
- Department of Biochemistry, Central University of Rajasthan, Rajasthan, India
| | - Rajan Kumar Pandey
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Vijay Kumar Prajapati
- Department of Biochemistry, University of Delhi South Campus, Dhaula Kuan, New Delhi, India.
| |
Collapse
|
6
|
Shang L, Du Y, Zhao Y, Zhang Y, Liu C. The Interaction of OTUB1 and TRAF3 Mediates NLRP3 Inflammasome Activity to Regulate TGF-β1-induced BEAS-2B Cell Injury. Appl Biochem Biotechnol 2023; 195:7060-7074. [PMID: 36976509 DOI: 10.1007/s12010-023-04434-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2023] [Indexed: 03/29/2023]
Abstract
Asthma is a frequently chronic respiratory disease with inflammation and remodeling in the airway. OTUB1 has been reported to be associated with pulmonary diseases. However, the role and potential mechanism of OTUB1 in asthma remain unclear. The expressions of OTUB1 in the bronchial mucosal tissues of asthmatic children and TGF-β1-induced BEAS-2B cells were determined. The biological behaviors were assessed in an asthma in vitro model using a loss-function approach. The contents of inflammatory cytokines were detected by ELISA kits. The related protein expressions were performed using western blot assay. Besides, the interaction between OTUB1 and TRAF3 was detected by Co-IP and ubiquitination assays. Our results showed that OTUB1 level was increased in asthmatic bronchial mucosal tissues and TGF-β1-induced BEAS-2B cells. OTUB1 knockdown promoted proliferation, inhibited apoptosis and EMT of TGF-β1-treated cells. The inhibition of OTUB1 attenuated the TGF-β1-induced inflammation and remodeling. Furthermore, OTUB1 knockdown inhibited the deubiquitination of TRAF3 and further suppressed the activation of NLRP3 inflammasome. The overexpression of TRAF3 or NLRP3 reversed the positive role of OTUB1 knockdown in TGF-β1-induced cells injury. Collectively, OTUB1 deubiquitinates TRAF3 to activate NLRP3 inflammasome, thereby leading to inflammation and remodeling of TGF-β1-induced cells, and further promoting the pathogenesis of asthma.
Collapse
Affiliation(s)
- Liqun Shang
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Yujie Du
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Yali Zhao
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Yongqing Zhang
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
| | - Cuicui Liu
- Department of Respiratory and Asthma, Xi'an Children's Hospital, Xi'an, 710003, Shaanxi, China.
| |
Collapse
|
7
|
Le Y, Zhang J, Gong Z, Zhang Z, Nian X, Li X, Yu D, Ma N, Zhou R, Zhang G, Liu B, Yang L, Fu B, Xu X, Yang X. TRAF3 deficiency in MDCK cells improved sensitivity to the influenza A virus. Heliyon 2023; 9:e19246. [PMID: 37681145 PMCID: PMC10481187 DOI: 10.1016/j.heliyon.2023.e19246] [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: 01/23/2023] [Revised: 06/29/2023] [Accepted: 08/16/2023] [Indexed: 09/09/2023] Open
Abstract
Tumor necrosis factor receptor-associated factor 3 (TRAF3), an adaptor protein, has significant and varying effects on immunity depending on cell types. The role of TRAF3 in Madin-Darby Canine Kidney Epithelial (MDCK) cell resistance to influenza A virus (IVA) remains elusive. In the present study, CRISPR-Cas9 gene editing technology was used to construct the TRAF3 knockout MDCK cells (MDCK-TRAF3-/-). Hemagglutination assay, plaque assay, transcriptome, and quantitative real-time PCR were performed after IVA infection. The results showed that after IVA infection, HA titers and virus titers were promoted, interferon I-related pathways were significantly blocked, and transcription of several antiviral-related genes was significantly decreased in MDCK-TRAF3-/- cells. Thus, our study suggests that TRAF3 gene knockout reduced MDCK cell's resistance to IVA, thereby resulting in a promising way for IVA isolation and vaccine manufacturing.
Collapse
Affiliation(s)
- Yang Le
- National Engineering Technology Research Center for Combined Vaccines, 430207, Wuhan, China
- Wuhan Institute of Biological Products Co.Ltd., 430207, Wuhan, China
| | - Jiayou Zhang
- National Engineering Technology Research Center for Combined Vaccines, 430207, Wuhan, China
- Wuhan Institute of Biological Products Co.Ltd., 430207, Wuhan, China
| | - Zheng Gong
- National Engineering Technology Research Center for Combined Vaccines, 430207, Wuhan, China
- Wuhan Institute of Biological Products Co.Ltd., 430207, Wuhan, China
| | - Zhegang Zhang
- National Engineering Technology Research Center for Combined Vaccines, 430207, Wuhan, China
- Wuhan Institute of Biological Products Co.Ltd., 430207, Wuhan, China
| | - Xuanxuan Nian
- National Engineering Technology Research Center for Combined Vaccines, 430207, Wuhan, China
- Wuhan Institute of Biological Products Co.Ltd., 430207, Wuhan, China
| | - Xuedan Li
- National Engineering Technology Research Center for Combined Vaccines, 430207, Wuhan, China
- Wuhan Institute of Biological Products Co.Ltd., 430207, Wuhan, China
| | - Daiguan Yu
- National Engineering Technology Research Center for Combined Vaccines, 430207, Wuhan, China
- Wuhan Institute of Biological Products Co.Ltd., 430207, Wuhan, China
| | - Ning Ma
- National Engineering Technology Research Center for Combined Vaccines, 430207, Wuhan, China
- Wuhan Institute of Biological Products Co.Ltd., 430207, Wuhan, China
| | - Rong Zhou
- National Engineering Technology Research Center for Combined Vaccines, 430207, Wuhan, China
- Wuhan Institute of Biological Products Co.Ltd., 430207, Wuhan, China
| | - Guomei Zhang
- National Engineering Technology Research Center for Combined Vaccines, 430207, Wuhan, China
- Wuhan Institute of Biological Products Co.Ltd., 430207, Wuhan, China
| | - Bo Liu
- National Engineering Technology Research Center for Combined Vaccines, 430207, Wuhan, China
- Wuhan Institute of Biological Products Co.Ltd., 430207, Wuhan, China
| | - Lu Yang
- National Engineering Technology Research Center for Combined Vaccines, 430207, Wuhan, China
- Wuhan Institute of Biological Products Co.Ltd., 430207, Wuhan, China
| | - Baiqi Fu
- Wuhan Institute of Biotechnology, Wuhan, 430075, China
| | - Xiuqin Xu
- Wuhan Institute of Biotechnology, Wuhan, 430075, China
| | - Xiaoming Yang
- National Engineering Technology Research Center for Combined Vaccines, 430207, Wuhan, China
- Wuhan Institute of Biological Products Co.Ltd., 430207, Wuhan, China
- China National Biotech Group Company Limited, 100029, Bejing, China
| |
Collapse
|
8
|
Li J, Ding J, Chen M, Chen K, Zou Y, Xu X, Zhang D, Yu X, Ding Z. Transcriptome-wide N6-methyladenosine modification profiling of mRNAs during infection of Newcastle disease virus in chicken macrophages. Virus Res 2023; 323:198993. [PMID: 36326508 PMCID: PMC10194374 DOI: 10.1016/j.virusres.2022.198993] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/15/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022]
Abstract
N6-methyladenosine (m6A) modification, the most prevalent post-transcriptional modification of eukaryotic mRNAs, is reported to play a crucial role in viral infection. However, the role of m6A modification during Newcastle disease virus (NDV) infection has remained unclear. In this study, we performed MeRIP-seq to investigate the transcriptome-wide m6A methylome and m6A-modified genes in NDV-infected chicken macrophages. A total of 9496 altered peaks were identified, of which 7015 peaks were significantly upregulated across 3320 genes, and 2481 peaks were significantly down-regulated across 1264 genes. Combined analysis of m6A peaks and mRNA expression showed that 1234 mRNAs had significantly altered levels of methylation and expression after NDV infection, and m6A modification tended to have a negative relationship with mRNA expression, suggesting that m6A modification may regulate the process of NDV infection by regulating gene expression, particularly of the genes important in the innate immune response. To the best of our knowledge, this is the first comprehensive characterization of m6A patterns in chicken macrophage mRNA after NDV infection, providing a valuable basis for further exploring the role of m6A modification mechanisms during the course of NDV infection.
Collapse
Affiliation(s)
- Jindou Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Jiaxin Ding
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Minghua Chen
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Kainan Chen
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yinxue Zou
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xiaohong Xu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Di Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xibing Yu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Zhuang Ding
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| |
Collapse
|
9
|
Guven-Maiorov E, Sakakibara N, Ponnamperuma RM, Dong K, Matar H, King KE, Weinberg WC. Delineating functional mechanisms of the p53/p63/p73 family of transcription factors through identification of protein-protein interactions using interface mimicry. Mol Carcinog 2022; 61:629-642. [PMID: 35560453 PMCID: PMC9949960 DOI: 10.1002/mc.23405] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 11/08/2022]
Abstract
Members of the p53 family of transcription factors-p53, p63, and p73-share a high degree of homology; however, members can be activated in response to different stimuli, perform distinct (sometimes opposing) roles and are expressed in different tissues. The level of complexity is increased further by the transcription of multiple isoforms of each homolog, which may interact or interfere with each other and can impact cellular outcome. Proteins perform their functions through interacting with other proteins (and/or with nucleic acids). Therefore, identification of the interactors of a protein and how they interact in 3D is essential to fully comprehend their roles. By utilizing an in silico protein-protein interaction prediction method-HMI-PRED-we predicted interaction partners of p53 family members and modeled 3D structures of these protein interaction complexes. This method recovered experimentally known interactions while identifying many novel candidate partners. We analyzed the similarities and differences observed among the interaction partners to elucidate distinct functions of p53 family members and provide examples of how this information may yield mechanistic insight to explain their overlapping versus distinct/opposing outcomes in certain contexts. While some interaction partners are common to p53, p63, and p73, the majority are unique to each member. Nevertheless, most of the enriched pathways associated with these partners are common to all members, indicating that the members target the same biological pathways but through unique mediators. p63 and p73 have more common enriched pathways compared to p53, supporting their similar developmental roles in different tissues.
Collapse
Affiliation(s)
- Emine Guven-Maiorov
- Laboratory of Molecular Oncology, Office of Biotechnology Products, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States.,National Cancer Institute, Bethesda, MD, United States.,Postal and email addresses of corresponding authors FDA/CDER/OPQ/OBP, Building 52-72/2306, 10903 New Hampshire Avenue, Silver Spring, MD 20993, United States, ,
| | - Nozomi Sakakibara
- Laboratory of Molecular Oncology, Office of Biotechnology Products, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Roshini M. Ponnamperuma
- Laboratory of Molecular Oncology, Office of Biotechnology Products, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Kun Dong
- Laboratory of Molecular Oncology, Office of Biotechnology Products, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States.,National Cancer Institute, Bethesda, MD, United States
| | - Hector Matar
- Laboratory of Molecular Oncology, Office of Biotechnology Products, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Kathryn E. King
- Laboratory of Molecular Oncology, Office of Biotechnology Products, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States
| | - Wendy C. Weinberg
- Laboratory of Molecular Oncology, Office of Biotechnology Products, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, United States.,Postal and email addresses of corresponding authors FDA/CDER/OPQ/OBP, Building 52-72/2306, 10903 New Hampshire Avenue, Silver Spring, MD 20993, United States, ,
| |
Collapse
|
10
|
Schrank TP, Prince AC, Sathe T, Wang X, Liu X, Alzhanov DT, Burtness B, Baldwin AS, Yarbrough WG, Issaeva N. NF-κB over-activation portends improved outcomes in HPV-associated head and neck cancer. Oncotarget 2022; 13:707-722. [PMID: 35634245 PMCID: PMC9131933 DOI: 10.18632/oncotarget.28232] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/03/2022] [Indexed: 12/12/2022] Open
Abstract
Evolving understanding of head and neck squamous cell carcinoma (HNSCC) is leading to more specific diagnostic disease classifications. Among HNSCC caused by the human papilloma virus (HPV), tumors harboring defects in TRAF3 or CYLD are associated with improved clinical outcomes and maintenance of episomal HPV. TRAF3 and CYLD are negative regulators of NF-κB and inactivating mutations of either leads to NF-κB overactivity. Here, we developed and validated a gene expression classifier separating HPV+ HNSCCs based on NF-κB activity. As expected, the novel classifier is strongly enriched in NF-κB targets leading us to name it the NF-κB Activity Classifier (NAC). High NF-κB activity correlated with improved survival in two independent cohorts. Using NAC, tumors with high NF-κB activity but lacking defects in TRAF3 or CYLD were identified; thus, while TRAF3 or CYLD gene defects identify the majority of tumors with NF-κB activation, unknown mechanisms leading to NF-kB activity also exist. The NAC correctly classified the functional consequences of two novel CYLD missense mutations. Using a reporter assay, we tested these CYLD mutations revealing that their activity to inhibit NF-kB was equivalent to the wild-type protein. Future applications of the NF-κB Activity Classifier may be to identify HPV+ HNSCC patients with better or worse survival with implications for treatment strategies.
Collapse
Affiliation(s)
- Travis P. Schrank
- Department of Otolaryngology/Head and Neck Surgery, UNC, Chapel Hill, NC 27599, USA
- These authors contributed equally to this work
| | - Andrew C. Prince
- Department of Otolaryngology/Head and Neck Surgery, UNC, Chapel Hill, NC 27599, USA
- These authors contributed equally to this work
| | - Tejas Sathe
- Department of Surgery, Otolaryngology, Yale, New Haven, CT 06519, USA
- Current address: Department of Surgery, Columbia University, New York, NY 10032, USA
| | - Xiaowei Wang
- Department of Pharmacology and Bioengineering, University of Illinois at Chicago, Chicago, IL 60612, USA
- Bioinformatics Core, University of Illinois Cancer Center, Chicago, IL 60612, USA
| | - Xinyi Liu
- Department of Pharmacology and Bioengineering, University of Illinois at Chicago, Chicago, IL 60612, USA
- Bioinformatics Core, University of Illinois Cancer Center, Chicago, IL 60612, USA
| | - Damir T. Alzhanov
- Department of Otolaryngology/Head and Neck Surgery, UNC, Chapel Hill, NC 27599, USA
| | - Barbara Burtness
- Department of Medicine, Yale School of Medicine, New Haven, CT 06510, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT 06510, USA
| | - Albert S. Baldwin
- Department of Medicine, Yale School of Medicine, New Haven, CT 06510, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT 06510, USA
| | - Wendell G. Yarbrough
- Department of Otolaryngology/Head and Neck Surgery, UNC, Chapel Hill, NC 27599, USA
- Lineberger Cancer Center, UNC, Chapel Hill, NC 27599, USA
- Department of Pathology and Laboratory Medicine, UNC, Chapel Hill, NC 27599, USA
- Senior authors
| | - Natalia Issaeva
- Department of Otolaryngology/Head and Neck Surgery, UNC, Chapel Hill, NC 27599, USA
- Lineberger Cancer Center, UNC, Chapel Hill, NC 27599, USA
- Department of Pathology and Laboratory Medicine, UNC, Chapel Hill, NC 27599, USA
- Senior authors
| |
Collapse
|
11
|
Abstract
Immunity could be viewed as the common factor in neurodevelopmental disorders and cancer. The immune and nervous systems coevolve as the embryo develops. Immunity can release cytokines that activate MAPK signaling in neural cells. In specific embryonic brain cell types, dysregulated signaling that results from germline or embryonic mutations can promote changes in chromatin organization and gene accessibility, and thus expression levels of essential genes in neurodevelopment. In cancer, dysregulated signaling can emerge from sporadic somatic mutations during human life. Neurodevelopmental disorders and cancer share similarities. In neurodevelopmental disorders, immunity, and cancer, there appears an almost invariable involvement of small GTPases (e.g., Ras, RhoA, and Rac) and their pathways. TLRs, IL-1, GIT1, and FGFR signaling pathways, all can be dysregulated in neurodevelopmental disorders and cancer. Although there are signaling similarities, decisive differentiating factors are timing windows, and cell type specific perturbation levels, pointing to chromatin reorganization. Finally, we discuss drug discovery.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD 21702, USA
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
- Corresponding author
| | - Chung-Jung Tsai
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD 21702, USA
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD 21702, USA
| |
Collapse
|
12
|
Chen F, Chen L, Li Y, Sang H, Zhang C, Yuan S, Yang J. TRAF3 Positively Regulates Host Innate Immune Resistance to Influenza A Virus Infection. Front Cell Infect Microbiol 2022; 12:839625. [PMID: 35573779 PMCID: PMC9093644 DOI: 10.3389/fcimb.2022.839625] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/28/2022] [Indexed: 11/13/2022] Open
Abstract
Tumor necrosis factor receptor-associated factor 3 (TRAF3) is one of the intracellular adaptor proteins for the innate immune response, which is involved in signaling regulation in various cellular processes, including the immune responses defending against invading pathogens. However, the defense mechanism of TRAF3 against influenza virus infection remains elusive. In this study, we found that TRAF3 could positively regulate innate antiviral response. Overexpression of TRAF3 significantly enhanced virus-induced IRF3 activation, IFN-β production, and antiviral response, while TRAF3 knockdown promoted influenza A virus replication. Moreover, we clarified that inhibiting ubiquitinated degradation of TRAF3 was associated with anti-influenza effect, thereby facilitating antiviral immunity upon influenza A virus infection. We further demonstrated the key domains of TRAF3 involved in anti-influenza effect. Taken together, these results suggested that TRAF3 performs a vital role in host defense against influenza A virus infection by the type-I IFN signaling pathway. Our findings provide insights into the development of drugs to prevent TRAF3 degradation, which could be a novel therapeutic approach for treatment of influenza A virus infection.
Collapse
Affiliation(s)
- Fangzhao Chen
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
- Department of Pharmacy, Shenzhen Children’s Hospital, Shenzhen, China
| | - Liurong Chen
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Yinyan Li
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Huiting Sang
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Chunyu Zhang
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Shuofeng Yuan
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Jie Yang
- National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
13
|
Geronikolou SA, Takan I, Pavlopoulou A, Mantzourani M, Chrousos GP. Thrombocytopenia in COVID‑19 and vaccine‑induced thrombotic thrombocytopenia. Int J Mol Med 2022; 49:35. [PMID: 35059730 PMCID: PMC8815408 DOI: 10.3892/ijmm.2022.5090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/28/2021] [Indexed: 12/16/2022] Open
Abstract
The highly heterogeneous symptomatology and unpredictable progress of COVID-19 triggered unprecedented intensive biomedical research and a number of clinical research projects. Although the pathophysiology of the disease is being progressively clarified, its complexity remains vast. Moreover, some extremely infrequent cases of thrombotic thrombocytopenia following vaccination against SARS-CoV-2 infection have been observed. The present study aimed to map the signaling pathways of thrombocytopenia implicated in COVID-19, as well as in vaccine-induced thrombotic thrombocytopenia (VITT). The biomedical literature database, MEDLINE/PubMed, was thoroughly searched using artificial intelligence techniques for the semantic relations among the top 50 similar words (>0.9) implicated in COVID-19-mediated human infection or VITT. Additionally, STRING, a database of primary and predicted associations among genes and proteins (collected from diverse resources, such as documented pathway knowledge, high-throughput experimental studies, cross-species extrapolated information, automated text mining results, computationally predicted interactions, etc.), was employed, with the confidence threshold set at 0.7. In addition, two interactomes were constructed: i) A network including 119 and 56 nodes relevant to COVID-19 and thrombocytopenia, respectively; and ii) a second network containing 60 nodes relevant to VITT. Although thrombocytopenia is a dominant morbidity in both entities, three nodes were observed that corresponded to genes (AURKA, CD46 and CD19) expressed only in VITT, whilst ADAM10, CDC20, SHC1 and STXBP2 are silenced in VITT, but are commonly expressed in both COVID-19 and thrombocytopenia. The calculated average node degree was immense (11.9 in COVID-19 and 6.43 in VITT), illustrating the complexity of COVID-19 and VITT pathologies and confirming the importance of cytokines, as well as of pathways activated following hypoxic events. In addition, PYCARD, NLP3 and P2RX7 are key potential therapeutic targets for all three morbid entities, meriting further research. This interactome was based on wild-type genes, revealing the predisposition of the body to hypoxia-induced thrombosis, leading to the acute COVID-19 phenotype, the 'long-COVID syndrome', and/or VITT. Thus, common nodes appear to be key players in illness prevention, progression and treatment.
Collapse
Affiliation(s)
- Styliani A Geronikolou
- Clinical, Translational and Experimental Surgery Research Centre, Biomedical Research Foundation Academy of Athens, 11527 Athens, Greece
| | - Işil Takan
- Izmir Biomedicine and Genome Center (IBG), 35340 Izmir, Turkey
| | | | - Marina Mantzourani
- First Department of Internal Medicine, Laiko Hospital, National and Kapodistrian University of Athens Medical School, 11527 Athens, Greece
| | - George P Chrousos
- Clinical, Translational and Experimental Surgery Research Centre, Biomedical Research Foundation Academy of Athens, 11527 Athens, Greece
| |
Collapse
|
14
|
Xu YP, Zhou YL, Xiao Y, Gu WB, Li B, Cheng YX, Li BW, Chen DY, Zhao XF, Dong WR, Shu MA. Functional differences in the products of two TRAF3 genes in antiviral responses in the Chinese giant salamander, Andrias davidianus. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 119:104015. [PMID: 33460679 DOI: 10.1016/j.dci.2021.104015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 12/20/2020] [Accepted: 12/20/2020] [Indexed: 06/12/2023]
Abstract
Tumour necrosis factor receptor associated factor 3 (TRAF3) is a crucial transducing protein for linking upstream receptor signals and downstream antiviral signalling pathways. Previous studies mostly clarified the functions of TRAF3 in mammals, birds and fish, but little is known about the characterization and function of TRAF3 in amphibians. In this study, the molecular and functional identification of two TRAF3 genes, AdTRAF3A and AdTRAF3B, were investigated in the Chinese giant salamander Andrias davidianus. The complete open reading frames (ORFs) of AdTRAF3A and AdTRAF3B were 1698 bp and 1743 bp in length, encoding 565 and 580 amino acids, respectively. Both AdTRAF3A and AdTRAF3B deduced proteins contained a RING finger, two TRAF-type zinc fingers, a coiled-coil and a MATH domain. Phylogenetic analysis showed that the AdTRAF3 protein clustered together with other known TRAF3 proteins. Gene expression analysis showed that AdTRAF3s were broadly distributed in all examined tissues with similar distribution patterns. AdTRAF3s in the blood or spleen positively responded to Giant salamander iridovirus (GSIV) and poly (I:C) induction but exhibited distinct response patterns. Silencing AdTRAF3A/B remarkably suppressed the expression of IFN signalling pathway-related genes when leukocytes were treated with DNA virus and the viral RNA analogue. Moreover, overexpression of AdTRAF3A may induce the activation of the IFN-β promoter, and the zinc finger, coiled coil and MATH domains of AdTRAF3A were essential for IFN-β promoter activation. However, the overexpression of AdTRAF3B significantly suppressed IFN-β promoter activity, and its inhibitory effect was enhanced when the RING finger or MATH domain was deleted. Furthermore, AdTRAF3A rather than AdTRAF3B significantly induced NF-κB activation, implying that AdTRAF3A may function as an enhancer in both the IFN and NF-κB signalling pathways. Taken together, our results suggest that the two TRAF3 genes play different crucial regulatory roles in innate antiviral immunity in Chinese giant salamanders.
Collapse
Affiliation(s)
- Ya-Ping Xu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yi-Lian Zhou
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yi Xiao
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Wen-Bin Gu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Bo Li
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yuan-Xin Cheng
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Bing-Wu Li
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Da-Yong Chen
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xiao-Feng Zhao
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 310036, China
| | - Wei-Ren Dong
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Miao-An Shu
- College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
15
|
Wang F, Han S, Yang J, Yan W, Hu G. Knowledge-Guided "Community Network" Analysis Reveals the Functional Modules and Candidate Targets in Non-Small-Cell Lung Cancer. Cells 2021; 10:cells10020402. [PMID: 33669233 PMCID: PMC7919838 DOI: 10.3390/cells10020402] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/06/2021] [Accepted: 02/15/2021] [Indexed: 12/24/2022] Open
Abstract
Non-small-cell lung cancer (NSCLC) represents a heterogeneous group of malignancies that are the leading cause of cancer-related death worldwide. Although many NSCLC-related genes and pathways have been identified, there remains an urgent need to mechanistically understand how these genes and pathways drive NSCLC. Here, we propose a knowledge-guided and network-based integration method, called the node and edge Prioritization-based Community Analysis, to identify functional modules and their candidate targets in NSCLC. The protein–protein interaction network was prioritized by performing a random walk with restart algorithm based on NSCLC seed genes and the integrating edge weights, and then a “community network” was constructed by combining Girvan–Newman and Label Propagation algorithms. This systems biology analysis revealed that the CCNB1-mediated network in the largest community provides a modular biomarker, the second community serves as a drug regulatory module, and the two are connected by some contextual signaling motifs. Moreover, integrating structural information into the signaling network suggested novel protein–protein interactions with therapeutic significance, such as interactions between GNG11 and CXCR2, CXCL3, and PPBP. This study provides new mechanistic insights into the landscape of cellular functions in the context of modular networks and will help in developing therapeutic targets for NSCLC.
Collapse
Affiliation(s)
- Fan Wang
- Center for Systems Biology, Department of Bioinformatics, School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, China; (F.W.); (S.H.); (J.Y.)
| | - Shuqing Han
- Center for Systems Biology, Department of Bioinformatics, School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, China; (F.W.); (S.H.); (J.Y.)
| | - Ji Yang
- Center for Systems Biology, Department of Bioinformatics, School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, China; (F.W.); (S.H.); (J.Y.)
| | - Wenying Yan
- Center for Systems Biology, Department of Bioinformatics, School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, China; (F.W.); (S.H.); (J.Y.)
- Correspondence: (W.Y.); (G.H.)
| | - Guang Hu
- Center for Systems Biology, Department of Bioinformatics, School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, China; (F.W.); (S.H.); (J.Y.)
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
- Correspondence: (W.Y.); (G.H.)
| |
Collapse
|
16
|
Lv Y, Xu Q, Mao Y, Xu Y, Zhang R, Zhong H, Zhou Y, Xiao J, Du M, Song H, Liang Y, Yan J. TRAF3 of blunt snout bream participates in host innate immune response to pathogenic bacteria via NF-κB signaling pathway. FISH & SHELLFISH IMMUNOLOGY 2020; 104:592-604. [PMID: 32589928 DOI: 10.1016/j.fsi.2020.06.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 06/08/2020] [Accepted: 06/10/2020] [Indexed: 06/11/2023]
Abstract
Tumor necrosis factor receptor-associated factor 3 (TRAF3) is a multifunctional adaptor protein primarily involved in both bacterial defense and antiviral immunity in living organisms. However, the knowledge on TRAF3 in blunt snout bream (Megalobrama amblycephala), a freshwater fish with economic values, remained unclear. In the present study, we identified and characterized successfully Traf3 gene from M. amblycephala (maTraf3). The maTraf3 cDNA contained a 1722 bp open reading frame that encoded a protein of 573 amino acid residues. The deduced amino acid sequence comprised of a RING finger domain, two zinc finger motifs, a coiled-coil region and a MATH domain. Analysis of the transcriptional patterns of maTraf3 revealed that it was ubiquitously distributed in various tissues tested from M. amblycephala, with the abundance of expression in spleen and muscle. Following a challenge with Aeromonas hydrophila and lipopolysaccharide stimulation, the expression of maTraf3 was strongly enhanced at different time points in vitro and in vivo. MaTRAF3 was identified as a cytosolic protein and suggested to form aggregates or be associated with vesicles scattering in the cytoplasm. NF-κB transcription was activated by maTraf3 in reporter assay. The overexpression of maTraf3 produced high levels of pro-inflammatory cytokines such as IL-1β, IL-6, IL-8 and TNF-α, implying its immune-regulatory role in M. amblycephala. Taken together, our results obtained in this study demonstrated the crucial role of maTraf3 in mediating host innate immune response to pathogen invasion via NF-κB signaling pathway, which might indicate a novel therapeutic approach to combat bacterial infection in fish.
Collapse
Affiliation(s)
- Yina Lv
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, 410017, China; Department of Biology, Zhaoqing University, Zhaoqing, 526061, China
| | - Qian Xu
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, 410017, China
| | - Ying Mao
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, 410017, China
| | - Yandong Xu
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, 410017, China
| | - Ru Zhang
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, 410017, China
| | - Huan Zhong
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, Guangxi, 530021, China
| | - Yi Zhou
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, Guangxi, 530021, China
| | - Jun Xiao
- Guangxi Key Laboratory of Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, Guangxi, 530021, China
| | - Mengke Du
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, 410017, China
| | - Huiyang Song
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, 410017, China
| | - Yinhua Liang
- Department of Operation, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Jinpeng Yan
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, 410017, China.
| |
Collapse
|
17
|
Sun H, Tian J, Li J. MiR-92b-3p ameliorates inflammation and autophagy by targeting TRAF3 and suppressing MKK3-p38 pathway in caerulein-induced AR42J cells. Int Immunopharmacol 2020; 88:106691. [PMID: 32822908 DOI: 10.1016/j.intimp.2020.106691] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/20/2020] [Accepted: 06/08/2020] [Indexed: 02/08/2023]
Abstract
Acute pancreatitis (AP) is an inflammatory disease with high morbidity and mortality. Dysregulation of microRNAs (miRNAs) was involved in human diseases, including AP. However, the effects of miR-92b-3p on AP process and its mechanism remain not been fully clarified. The expression levels of miR-92b-3p and tumor necrosis factor receptor-associated factor-3 (TRAF3) were measured by quantitative real-time polymerase chain reaction (qRT-PCR). The protein levels of TRAF3, tumor necrosis factor α (TNF-α) TNF-α, interleukin-6 (IL-6), phosphorylated mitogen-activated protein kinase kinase 3 (p-MKK3), MKK3, p38 and phosphorylated p38 (p-p38) were detected by western blot. The concentration of TNF-α and IL-6 in the medium was measured using ELISA kits. The possible binding sites of miR-92b-3p and TRAF3 were predicted by TargetScan and verified by dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay. The expression level of miR-92b-3p was decreased and TRAF3 expression was increased in AR42J cells stimulated with caerulein. Moreover, the protein levels of pro-inflammatory cytokines (TNF-α and IL-6) were markedly elevated, and the expression levels of autophagy-related markers Beclin1 as well as the ratio of LC3-II/I were obviously increased in AR42J cells treated with caerulein. In addition, overexpression of miR-92b-3p or knockdown of TRAF3 significantly suppressed the release of pro-inflammatory cytokines and autophagy in caerulein-induced AR42J cells. Furthermore, TRAF3 was a direct target of miR-92b-3p and its upregulation reversed the effects of miR-92b-3p overexpression on inflammatory response and autophagy. Besides, overexpression of miR-92b-3p inhibited the activation of the MKK3-p38 pathway by affecting TRAF3 expression. In conclusion, miR-92b-3p attenuated inflammatory response and autophagy by downregulating TRAF3 and suppressing MKK3-p38 pathway in caerulein-induced AR42J cells, providing a novel avenue for treatment of AP.
Collapse
Affiliation(s)
- Hongzhi Sun
- Department of Critical Care Medicine, The Second Hospital of Jilin University, No 218 Ziqiang Street, Nanguan District 130041, Changchun, China
| | - Jiakun Tian
- Department of Critical Care Medicine, The Second Hospital of Jilin University, No 218 Ziqiang Street, Nanguan District 130041, Changchun, China
| | - Jinliang Li
- Department of Critical Care Medicine, The Second Hospital of Jilin University, No 218 Ziqiang Street, Nanguan District 130041, Changchun, China.
| |
Collapse
|
18
|
Shen Y, Liu WW, Zhang X, Shi JG, Jiang S, Zheng L, Qin Y, Liu B, Shi JH. TRAF3 promotes ROS production and pyroptosis by targeting ULK1 ubiquitination in macrophages. FASEB J 2020; 34:7144-7159. [PMID: 32275117 DOI: 10.1096/fj.201903073r] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 03/18/2020] [Accepted: 03/23/2020] [Indexed: 12/11/2022]
Abstract
Disrupted mitochondrial function and reactive oxygen species (ROS) generation cause cellular damage and oxidative stress-induced macrophage inflammatory cell death. It remains unclear how mitochondrial dysfunction relates to inflammasome activation and pyroptotic cell death. In this study, we demonstrated that tumor necrosis factor receptor-associated factor 3 (TRAF3) regulates mitochondrial ROS production and promotes TLR agonist LPS plus nigericin (LPS/Ng)-induced inflammasome and pyroptosis in mouse primary macrophages and human monocyte THP-1 cells. Co-IP assays confirmed that TRAF3 forms a complex with TRAF2 and cIAP1 and mediates ubiquitin and degradation of Unc-51 like autophagy activating kinase 1 (ULK1). Moreover, knockdown of ULK1 in THP-1 cells significantly promoted LPS/Ng-induced inflammasome by activating caspase 1 and mature IL-1β. Apoptosis inducing factor (AIF) translocation from mitochondrial to nuclear was observed in ULK1-deficient THP-1 cells under LPS/Ng stimulation, which mediates LPS/Ng-induced cell death in ULK1 deficient macrophages. In conclusion, this study identified a novel role of TRAF3 in regulation of ULK1 ubiquitination and inflammasome signaling and provided molecular mechanisms by which ubiquitination of ULK1 controls mitochondrial ROS production, inflammasome activity, and AIF-dependent pyroptosis.
Collapse
Affiliation(s)
- Yang Shen
- Central Laboratory, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Affiliated Hospital of Hebei University, Baoding, China
| | - Wen-Wen Liu
- Central Laboratory, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Affiliated Hospital of Hebei University, Baoding, China
| | - Xiu Zhang
- Central Laboratory, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Affiliated Hospital of Hebei University, Baoding, China
| | - Jian-Guo Shi
- Department of Urinary Surgery, The 82nd Army Hospital of Chinese People's Liberation Army, Baoding, China
| | - Shan Jiang
- Central Laboratory, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Affiliated Hospital of Hebei University, Baoding, China
| | - Lishuang Zheng
- Central Laboratory, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Affiliated Hospital of Hebei University, Baoding, China
| | - Yan Qin
- Central Laboratory, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Affiliated Hospital of Hebei University, Baoding, China
| | - Bin Liu
- Central Laboratory, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Affiliated Hospital of Hebei University, Baoding, China
| | - Jian-Hong Shi
- Central Laboratory, Hebei Key Laboratory of Cancer Radiotherapy and Chemotherapy, Affiliated Hospital of Hebei University, Baoding, China
| |
Collapse
|
19
|
Guven-Maiorov E, Tsai CJ, Nussinov R. Oncoviruses Can Drive Cancer by Rewiring Signaling Pathways Through Interface Mimicry. Front Oncol 2019; 9:1236. [PMID: 31803618 PMCID: PMC6872517 DOI: 10.3389/fonc.2019.01236] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 10/28/2019] [Indexed: 01/17/2023] Open
Abstract
Oncoviruses rewire host pathways to subvert host immunity and promote their survival and proliferation. However, exactly how is challenging to understand. Here, by employing the first and to date only interface-based host-microbe interaction (HMI) prediction method, we explore a pivotal strategy oncoviruses use to drive cancer: mimicking binding surfaces-interfaces-of human proteins. We show that oncoviruses can target key human network proteins and transform cells by acquisition of cancer hallmarks. Experimental large-scale mapping of HMIs is difficult and individual HMIs do not permit in-depth grasp of tumorigenic virulence mechanisms. Our computational approach is tractable and 3D structural HMI models can help elucidate pathogenesis mechanisms and facilitate drug design. We observe that many host proteins are unique targets for certain oncoviruses, whereas others are common to several, suggesting similar infectious strategies. A rough estimation of our false discovery rate based on the tissue expression of oncovirus-targeted human proteins is 25%.
Collapse
Affiliation(s)
- Emine Guven-Maiorov
- Computational Structural Biology Section, Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Chung-Jung Tsai
- Computational Structural Biology Section, Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Ruth Nussinov
- Computational Structural Biology Section, Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
- Department of Human Genetics and Molecular Medicine, Sackler Institute of Molecular Medicine, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
20
|
Zhou Y, Zhou Y, Kang X, Meng C, Zhang R, Guo Y, Xiong D, Song L, Jiao X, Pan Z. Molecular cloning and functional characterisation of duck ( Anas platyrhynchos) tumour necrosis factor receptor-associated factor 3. Br Poult Sci 2019; 60:357-365. [PMID: 31046421 DOI: 10.1080/00071668.2019.1614528] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
1. Tumour necrosis factor receptor-associated factor 3 (TRAF3) is a key regulator of innate immunity and acquired immunity, and has a salient anti-viral role. 2. In this experiment, the duck TRAF3 (DuTRAF3) gene was cloned according to the Anas platyrhynchos TRAF3 sequence to explore its function. The TRAF3 open reading frame contains 1704 bp that encode a protein of 567 amino acids, which contain a RING finger domain, two zinc finger motifs, a coiled-coil region, and a MATH domain. 3. Reverse transcription-polymerase chain reaction showed that DuTRAF3 was expressed in all the examined tissues, with a comparatively higher expression in the spleen and brain tissues. 4. In HEK293T cells, DuTRAF3 overexpression resulted in a significantly increased NF-κB activity and interferon (IFN)-β promoter activation. 5. Following resiquimod (R848) and poly(I:C) stimulation of duck peripheral blood mononuclear cells (PBMCs), the expressions of TRAF3 and IFN-β were significantly upregulated; in addition, following R848 stimulation, the mRNA levels of IL-6, IL-8 and IL-10 were also significantly upregulated. After infection with the Newcastle Disease Virus LaSota vaccine strain, the mRNA levels of IL-6 and IL-10 were significantly upregulated, while that of TRAF3 was downregulated. 6. These results suggest that DuTRAF3 has an important role to play in innate antiviral immune responses.
Collapse
Affiliation(s)
- Y Zhou
- a Jiangsu Key Laboratory of Zoonosis , Yangzhou University , Yangzhou , Jiangsu , China.,b Jiangsu Co-innovation Centre for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses , Yangzhou University , Yangzhou , Jiangsu , China.,c Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture and Rural Affairs , Yangzhou University , Yangzhou , Jiangsu , China.,d Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education , Yangzhou University , Yangzhou , Jiangsu , China
| | - Y Zhou
- a Jiangsu Key Laboratory of Zoonosis , Yangzhou University , Yangzhou , Jiangsu , China.,b Jiangsu Co-innovation Centre for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses , Yangzhou University , Yangzhou , Jiangsu , China.,c Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture and Rural Affairs , Yangzhou University , Yangzhou , Jiangsu , China.,d Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education , Yangzhou University , Yangzhou , Jiangsu , China
| | - X Kang
- a Jiangsu Key Laboratory of Zoonosis , Yangzhou University , Yangzhou , Jiangsu , China.,b Jiangsu Co-innovation Centre for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses , Yangzhou University , Yangzhou , Jiangsu , China.,c Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture and Rural Affairs , Yangzhou University , Yangzhou , Jiangsu , China.,d Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education , Yangzhou University , Yangzhou , Jiangsu , China
| | - C Meng
- a Jiangsu Key Laboratory of Zoonosis , Yangzhou University , Yangzhou , Jiangsu , China.,b Jiangsu Co-innovation Centre for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses , Yangzhou University , Yangzhou , Jiangsu , China.,c Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture and Rural Affairs , Yangzhou University , Yangzhou , Jiangsu , China.,d Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education , Yangzhou University , Yangzhou , Jiangsu , China
| | - R Zhang
- a Jiangsu Key Laboratory of Zoonosis , Yangzhou University , Yangzhou , Jiangsu , China.,b Jiangsu Co-innovation Centre for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses , Yangzhou University , Yangzhou , Jiangsu , China.,c Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture and Rural Affairs , Yangzhou University , Yangzhou , Jiangsu , China.,d Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education , Yangzhou University , Yangzhou , Jiangsu , China
| | - Y Guo
- a Jiangsu Key Laboratory of Zoonosis , Yangzhou University , Yangzhou , Jiangsu , China.,b Jiangsu Co-innovation Centre for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses , Yangzhou University , Yangzhou , Jiangsu , China.,c Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture and Rural Affairs , Yangzhou University , Yangzhou , Jiangsu , China.,d Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education , Yangzhou University , Yangzhou , Jiangsu , China
| | - D Xiong
- a Jiangsu Key Laboratory of Zoonosis , Yangzhou University , Yangzhou , Jiangsu , China.,b Jiangsu Co-innovation Centre for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses , Yangzhou University , Yangzhou , Jiangsu , China.,c Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture and Rural Affairs , Yangzhou University , Yangzhou , Jiangsu , China.,d Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education , Yangzhou University , Yangzhou , Jiangsu , China
| | - L Song
- a Jiangsu Key Laboratory of Zoonosis , Yangzhou University , Yangzhou , Jiangsu , China.,b Jiangsu Co-innovation Centre for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses , Yangzhou University , Yangzhou , Jiangsu , China.,c Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture and Rural Affairs , Yangzhou University , Yangzhou , Jiangsu , China.,d Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education , Yangzhou University , Yangzhou , Jiangsu , China
| | - X Jiao
- a Jiangsu Key Laboratory of Zoonosis , Yangzhou University , Yangzhou , Jiangsu , China.,b Jiangsu Co-innovation Centre for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses , Yangzhou University , Yangzhou , Jiangsu , China.,c Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture and Rural Affairs , Yangzhou University , Yangzhou , Jiangsu , China.,d Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education , Yangzhou University , Yangzhou , Jiangsu , China
| | - Z Pan
- a Jiangsu Key Laboratory of Zoonosis , Yangzhou University , Yangzhou , Jiangsu , China.,b Jiangsu Co-innovation Centre for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses , Yangzhou University , Yangzhou , Jiangsu , China.,c Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture and Rural Affairs , Yangzhou University , Yangzhou , Jiangsu , China.,d Joint International Research Laboratory of Agriculture and Agri-product Safety of the Ministry of Education , Yangzhou University , Yangzhou , Jiangsu , China
| |
Collapse
|
21
|
Swaidani S, Liu C, Zhao J, Bulek K, Li X. TRAF Regulation of IL-17 Cytokine Signaling. Front Immunol 2019; 10:1293. [PMID: 31316496 PMCID: PMC6610456 DOI: 10.3389/fimmu.2019.01293] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 05/21/2019] [Indexed: 01/23/2023] Open
Abstract
Tumor necrosis factor receptor (TNFR)-associated factors or (TRAFs) are important mediators of Interleukin-17 (IL-17) cytokine signaling and contribute to driving tissue responses that are crucial for protective immunity but are often implicated in immunopathology. By amplifying tissue immune activity, IL-17 cytokine pathways contribute to maintaining barrier function as well as activation of innate and adaptive immunity necessary for host defense. IL-17 receptors signaling is orchestrated in part, by the engagement of TRAFs and the subsequent unlocking of downstream cellular machinery that can promote pathogen clearance or contribute to immune dysregulation, chronic inflammation, and disease. Originally identified as signaling adaptors for TNFR superfamily, TRAF proteins can mediate the signaling of a variety of intercellular and extracellular stimuli and have been shown to regulate the downstream activity of many cytokine receptors including receptors for IL-1β, IL-2, IL-6, IL-17, IL-18, IL-33, type I IFNs, type III IFNs, GM-CSF, M-CSF, and TGF-β Toll-like receptors (TLRs), NOD-like receptors (NLRs), RIG-I- like receptors, and C-type lectin receptors. This review will focus on discussing studies that reveal our current understanding of how TRAFs mediate and regulate biochemical activities downstream of the IL-17 cytokines signaling.
Collapse
Affiliation(s)
- Shadi Swaidani
- Department of Inflammation and Immunity, Cleveland Clinic, Lerner Research Institute, Cleveland, OH, United States.,Department of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Lerner Research Institute, Cleveland, OH, United States
| | - Caini Liu
- Department of Inflammation and Immunity, Cleveland Clinic, Lerner Research Institute, Cleveland, OH, United States
| | - Junjie Zhao
- Department of Inflammation and Immunity, Cleveland Clinic, Lerner Research Institute, Cleveland, OH, United States
| | - Katarzyna Bulek
- Department of Inflammation and Immunity, Cleveland Clinic, Lerner Research Institute, Cleveland, OH, United States
| | - Xiaoxia Li
- Department of Inflammation and Immunity, Cleveland Clinic, Lerner Research Institute, Cleveland, OH, United States
| |
Collapse
|
22
|
Perez-Chacon G, Adrados M, Vallejo-Cremades MT, Lefebvre S, Reed JC, Zapata JM. Dysregulated TRAF3 and BCL2 Expression Promotes Multiple Classes of Mature Non-hodgkin B Cell Lymphoma in Mice. Front Immunol 2019; 9:3114. [PMID: 30687320 PMCID: PMC6338067 DOI: 10.3389/fimmu.2018.03114] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 12/17/2018] [Indexed: 11/13/2022] Open
Abstract
TNF-Receptor Associated Factor (TRAF)-3 is a master regulator of B cell homeostasis and function. TRAF3 has been shown to bind and regulate various proteins involved in the control of innate and adaptive immune responses. Previous studies showed that TRAF3 overexpression renders B cells hyper-reactive to antigens and Toll-like receptor (TLR) agonists, while TRAF3 deficiency has been implicated in the development of a variety of B cell neoplasms. In this report, we show that transgenic mice overexpressing TRAF3 and BCL2 in B cells develop with high incidence severe lymphadenopathy, splenomegaly and lymphoid infiltrations into tissues and organs, which is the result of the growth of monoclonal and oligoclonal B cell neoplasms, as demonstrated by analysis of VHDJH gene rearrangement. FACS and immunohistochemical analyses show that different types of mature B cell neoplasms arise in TRAF3/BCL2 double-transgenic (tg) mice, all of which are characterized by the loss of surface IgM and IgD expression. However, two types of lymphomas are predominant: (1) mature B cell neoplasms consistent with diffuse large B cell lymphoma and (2) plasma cell neoplasms. The Ig isotypes expressed by the expanded B-cell clones included IgA, IgG, and IgM, with most having undergone somatic hypermutation. In contrast, mouse littermates representing all the other genotypes (TRAF3-/BCL2-; TRAF3+/BCL2-, and TRAF3-/BCL2+) did not develop significant lymphadenopathy or clonal B cell expansions within the observation period of 20 months. Interestingly, a large representation of the HCDR3 sequences expressed in the TRAF3-tg and TRAF3/BCL2-double-tg B cells are highly similar to those recognizing pathogen-associated molecular patterns and damage-associated molecular patterns, strongly suggesting a role for TRAF3 in promoting B cell differentiation in response to these antigens. Finally, allotransplantation of either splenocytes or cell-containing ascites from lymphoma-bearing TRAF3/BCL2 mice into SCID/NOD immunodeficient mice showed efficient transfer of the parental expanded B-cell clones. Altogether, these results indicate that TRAF3, perhaps by promoting exacerbated B cell responses to certain antigens, and BCL2, presumably by supporting survival of these clones, cooperate to induce mature B cell neoplasms in transgenic mice.
Collapse
Affiliation(s)
- Gema Perez-Chacon
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, Madrid, Spain.,Instituto de Investigación Hospital Universitario La Paz, Madrid, Spain
| | - Magdalena Adrados
- Instituto de Investigación del Hospital Universitario de La Princesa, Madrid, Spain
| | | | - Sophie Lefebvre
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - John C Reed
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, United States
| | - Juan M Zapata
- Instituto de Investigaciones Biomédicas "Alberto Sols", CSIC-UAM, Madrid, Spain.,Instituto de Investigación Hospital Universitario La Paz, Madrid, Spain
| |
Collapse
|
23
|
Gillison ML, Akagi K, Xiao W, Jiang B, Pickard RKL, Li J, Swanson BJ, Agrawal AD, Zucker M, Stache-Crain B, Emde AK, Geiger HM, Robine N, Coombes KR, Symer DE. Human papillomavirus and the landscape of secondary genetic alterations in oral cancers. Genome Res 2018; 29:1-17. [PMID: 30563911 PMCID: PMC6314162 DOI: 10.1101/gr.241141.118] [Citation(s) in RCA: 163] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 11/30/2018] [Indexed: 12/15/2022]
Abstract
Human papillomavirus (HPV) is a necessary but insufficient cause of a subset of oral squamous cell carcinomas (OSCCs) that is increasing markedly in frequency. To identify contributory, secondary genetic alterations in these cancers, we used comprehensive genomics methods to compare 149 HPV-positive and 335 HPV-negative OSCC tumor/normal pairs. Different behavioral risk factors underlying the two OSCC types were reflected in distinctive genomic mutational signatures. In HPV-positive OSCCs, the signatures of APOBEC cytosine deaminase editing, associated with anti-viral immunity, were strongly linked to overall mutational burden. In contrast, in HPV-negative OSCCs, T>C substitutions in the sequence context 5'-ATN-3' correlated with tobacco exposure. Universal expression of HPV E6*1 and E7 oncogenes was a sine qua non of HPV-positive OSCCs. Significant enrichment of somatic mutations was confirmed or newly identified in PIK3CA, KMT2D, FGFR3, FBXW7, DDX3X, PTEN, TRAF3, RB1, CYLD, RIPK4, ZNF750, EP300, CASZ1, TAF5, RBL1, IFNGR1, and NFKBIA Of these, many affect host pathways already targeted by HPV oncoproteins, including the p53 and pRB pathways, or disrupt host defenses against viral infections, including interferon (IFN) and nuclear factor kappa B signaling. Frequent copy number changes were associated with concordant changes in gene expression. Chr 11q (including CCND1) and 14q (including DICER1 and AKT1) were recurrently lost in HPV-positive OSCCs, in contrast to their gains in HPV-negative OSCCs. High-ranking variant allele fractions implicated ZNF750, PIK3CA, and EP300 mutations as candidate driver events in HPV-positive cancers. We conclude that virus-host interactions cooperatively shape the unique genetic features of these cancers, distinguishing them from their HPV-negative counterparts.
Collapse
Affiliation(s)
- Maura L Gillison
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Keiko Akagi
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Weihong Xiao
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Bo Jiang
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Robert K L Pickard
- Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus, Ohio 43210, USA
| | - Jingfeng Li
- Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus, Ohio 43210, USA
| | - Benjamin J Swanson
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | - Amit D Agrawal
- Department of Otolaryngology - Head and Neck Surgery, Ohio State University Comprehensive Cancer Center, Columbus, Ohio 43210, USA
| | - Mark Zucker
- Department of Biomedical Informatics, Ohio State University Comprehensive Cancer Center, Columbus, Ohio 43210, USA
| | | | | | | | | | - Kevin R Coombes
- Department of Biomedical Informatics, Ohio State University Comprehensive Cancer Center, Columbus, Ohio 43210, USA
| | - David E Symer
- Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
24
|
Wang X, Song X, Xie X, Li W, Lu L, Chen S, Wu H, Feng H. TRAF3 enhances STING-mediated antiviral signaling during the innate immune activation of black carp. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 88:83-93. [PMID: 30009929 PMCID: PMC7124771 DOI: 10.1016/j.dci.2018.07.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/10/2018] [Accepted: 07/10/2018] [Indexed: 05/07/2023]
Abstract
Tumor necrosis factor receptor-associated factor 3 (TRAF3) is a main regulator of antiviral and anti-inflammatory pathways in mammals, which is considered to induce type I interferon (IFN) activation and negatively regulate the activation of the canonical and non-canonical NF-κB pathways. To elucidate its function in teleost fish, TRAF3 homologue of black carp (Mylopharyngodon piceus) has been cloned and characterized in this study. The open reading frame (ORF) of black carp TRAF3 (bcTRAF3) consists of 1722 nucleotides and bcTRAF3 contains 574 amino acids. bcTRAF3 protein migrated around 65 KDa in immunoblot analysis of both EPC and HEK293T cells. bcTRAF3 was identified as a cytosolic protein and suggested to form aggregates or be associated with vesicles scattering in the cytoplasm. It was interesting that both NF-κB and IFN transcription was activated by bcTRAF3 in reporter assay. When co-expressed with black carp STING (bcSTING), bcTRAF3 was redistributed in the cytoplasm and its subcellular location overlapped with that of bcSTING no matter what the cells was infected with GCRV or not, which suggested the association between these two molecules. bcSTING-mediated IFN production was up-regulated by bcTRAF3 in a dose dependent manner in reporter assay. Accordingly, EPC cells transfected with both bcSTING and bcTRAF3 showed enhanced antiviral activity comparing EPC cells expressing bcSTING alone. Taken together, the data generated in this paper supported the conclusion that bcTRAF3 was recruited into host innate immune activation and positively regulated bcSTING-mediated antiviral signaling.
Collapse
Affiliation(s)
- Xu Wang
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Xuejiao Song
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Xinchi Xie
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Wanzhen Li
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Liang Lu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Song Chen
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Hui Wu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China
| | - Hao Feng
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha 410081, China.
| |
Collapse
|
25
|
Abstract
Hundreds of different species colonize multicellular organisms making them "metaorganisms". A growing body of data supports the role of microbiota in health and in disease. Grasping the principles of host-microbiota interactions (HMIs) at the molecular level is important since it may provide insights into the mechanisms of infections. The crosstalk between the host and the microbiota may help resolve puzzling questions such as how a microorganism can contribute to both health and disease. Integrated superorganism networks that consider host and microbiota as a whole-may uncover their code, clarifying perhaps the most fundamental question: how they modulate immune surveillance. Within this framework, structural HMI networks can uniquely identify potential microbial effectors that target distinct host nodes or interfere with endogenous host interactions, as well as how mutations on either host or microbial proteins affect the interaction. Furthermore, structural HMIs can help identify master host cell regulator nodes and modules whose tweaking by the microbes promote aberrant activity. Collectively, these data can delineate pathogenic mechanisms and thereby help maximize beneficial therapeutics. To date, challenges in experimental techniques limit large-scale characterization of HMIs. Here we highlight an area in its infancy which we believe will increasingly engage the computational community: predicting interactions across kingdoms, and mapping these on the host cellular networks to figure out how commensal and pathogenic microbiota modulate the host signaling and broadly cross-species consequences.
Collapse
Affiliation(s)
- Emine Guven-Maiorov
- Cancer and Inflammation Program, Leidos Biomedical Research, Inc. Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, United States of America
| | - Chung-Jung Tsai
- Cancer and Inflammation Program, Leidos Biomedical Research, Inc. Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, United States of America
| | - Ruth Nussinov
- Cancer and Inflammation Program, Leidos Biomedical Research, Inc. Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, United States of America
- Sackler Inst. of Molecular Medicine, Department of Human Genetics and Molecular Medicine, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
26
|
Hajek M, Sewell A, Kaech S, Burtness B, Yarbrough WG, Issaeva N. TRAF3/CYLD mutations identify a distinct subset of human papillomavirus-associated head and neck squamous cell carcinoma. Cancer 2017; 123:1778-1790. [PMID: 28295222 PMCID: PMC5419871 DOI: 10.1002/cncr.30570] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 12/18/2016] [Accepted: 12/21/2016] [Indexed: 12/27/2022]
Abstract
BACKGROUND The incidence of human papillomavirus (HPV)‐associated (HPV‐positive) head and neck squamous cell carcinoma (HNSCC) of the oropharynx has dramatically increased over the last decade and continues to rise. Newly diagnosed HPV‐positive HNSCCs in the United States currently outnumber any other HPV‐associated cancers, including cervical cancer. Despite introduction of the HPV vaccine, the epidemic of HPV‐positive HNSCC is expected to continue for approximately 60 years. Compared with patients who have tobacco‐associated HNSCC, those who have HPV‐positive HNSCC have better overall survival and response to treatment. Current treatment, including chemotherapy and radiation therapy, is associated with lifelong morbidity, and there are limited treatments and no curative options for patients who develop recurrent metastatic disease. Therapeutic de‐escalation (decreased radiation dose) is being tested through clinical trials; however, those studies select patients based solely on tumor and patient smoking characteristics. Mechanisms of HPV‐driven carcinogenesis in HNSCC are not well understood, which limits new therapeutic strategies and hinders the appropriate selection of patients for de‐escalation therapy. METHODS The authors analyzed HNSCC data from The Cancer Genome Atlas to identify molecular characteristics that correlate with outcomes and integration status of the HPV genome. RESULTS The current investigations identified a subset of HPV‐positive HNSCCs with mutations in the genes TRAF3 (tumor necrosis factor receptor‐associated factor 3) and CYLD (cylindromatosis lysine 63 deubiquitinase). Defects in TRAF3 and CYLD correlated with the activation of transcriptional factor nuclear factor κB, episomal HPV status of tumors, and improved patient survival. CONCLUSIONS Defects in TRAF3/CYLD were accompanied with the activation of nuclear factor κB signaling and maintenance of episomal HPV in tumors, suggesting that these mutations may support an alternative mechanism of HPV tumorigenesis in head and neck tumors. Cancer 2017;123:1778–1790. © 2017 The Authors. Cancer published by Wiley Periodicals, Inc. on behalf of American Cancer Society. This is an open access article under the terms of the Creative Commons Attribution NonCommercial License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited and is not used for commercial purposes. Human papillomavirus‐associated head and neck cancer tends to respond better to treatment compared with tobacco‐associated tumors; however, patients suffer severe and long‐lasting side effects. Somatic mutations in the genes TRAF3 and CYLD identified in The Cancer Genome Atlas data set are correlated with the activation of nuclear factor‐κB, define a distinct etiologic subset of head and neck cancers, and will be useful as biomarkers for predicting improved prognosis and selecting patients with human papillomavirus‐positive head and neck cancer who may be successfully treated with de‐escalating therapy. See also pages 1695‐98.
Collapse
Affiliation(s)
- Michael Hajek
- Division of Otolaryngology, Department of Surgery, Yale University, New Haven, Connecticut
| | - Andrew Sewell
- Division of Otolaryngology, Department of Surgery, Yale University, New Haven, Connecticut
| | - Susan Kaech
- Department of Immunobiology, Yale University, New Haven, Connecticut
| | - Barbara Burtness
- Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut
| | - Wendell G Yarbrough
- Division of Otolaryngology, Department of Surgery, Yale University, New Haven, Connecticut.,Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut.,Department of Pathology, Yale School of Medicine, New Haven, Connecticut
| | - Natalia Issaeva
- Division of Otolaryngology, Department of Surgery, Yale University, New Haven, Connecticut.,Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
27
|
Mann JE, Hoesli R, Michmerhuizen NL, Devenport SN, Ludwig ML, Vandenberg TR, Matovina C, Jawad N, Mierzwa M, Shuman AG, Spector ME, Brenner JC. Surveilling the Potential for Precision Medicine-driven PD-1/PD-L1-targeted Therapy in HNSCC. J Cancer 2017; 8:332-344. [PMID: 28261333 PMCID: PMC5332883 DOI: 10.7150/jca.17547] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 11/14/2016] [Indexed: 12/20/2022] Open
Abstract
Immunotherapy is becoming an accepted treatment modality for many patients with cancer and is now approved for use in platinum-refractory recurrent or metastatic head and neck squamous cell carcinoma (HNSCC). Despite these successes, a minority of patients with HNSCC receiving immunotherapy respond to treatment, and few undergo a complete response. Thus, there is a critical need to identify mechanisms regulating immune checkpoints in HNSCC such that one can predict who will benefit, and so novel combination strategies can be developed for non-responders. Here, we review the immunotherapy and molecular genetics literature to describe what is known about immune checkpoints in common genetic subsets of HNSCC. We highlight several highly recurrent genetic lesions that may serve as biomarkers or targets for combination immunotherapy in HNSCC.
Collapse
Affiliation(s)
- J E Mann
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, MI.; Department of Pathology, University of Michigan Medical School, Ann Arbor, MI
| | - R Hoesli
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, MI
| | - N L Michmerhuizen
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, MI.; Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI
| | - S N Devenport
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, MI.; Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI
| | - M L Ludwig
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, MI.; Program in Cellular and Molecular Biology, University of Michigan Medical School, Ann Arbor, MI
| | - T R Vandenberg
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, MI
| | - C Matovina
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, MI
| | - N Jawad
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, MI
| | - M Mierzwa
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI.; Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI
| | - A G Shuman
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, MI.; Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI
| | - M E Spector
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, MI.; Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI
| | - J C Brenner
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, MI.; Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI
| |
Collapse
|
28
|
Cai Y, Huang T. Systems genetics - deciphering the complex disease with a systems approach. Biochim Biophys Acta Gen Subj 2016; 1860:2611-2. [DOI: 10.1016/j.bbagen.2016.07.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
29
|
Guven-Maiorov E, Tsai CJ, Nussinov R. Pathogen mimicry of host protein-protein interfaces modulates immunity. Semin Cell Dev Biol 2016; 58:136-45. [DOI: 10.1016/j.semcdb.2016.06.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 06/02/2016] [Accepted: 06/06/2016] [Indexed: 12/21/2022]
|
30
|
Csermely P, Korcsmáros T, Nussinov R. Intracellular and intercellular signaling networks in cancer initiation, development and precision anti-cancer therapy: RAS acts as contextual signaling hub. Semin Cell Dev Biol 2016; 58:55-9. [PMID: 27395026 PMCID: PMC5028272 DOI: 10.1016/j.semcdb.2016.07.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 07/05/2016] [Indexed: 12/31/2022]
Abstract
Cancer initiation and development are increasingly perceived as systems-level phenomena, where intra- and inter-cellular signaling networks of the ecosystem of cancer and stromal cells offer efficient methodologies for outcome prediction and intervention design. Within this framework, RAS emerges as a 'contextual signaling hub', i.e. the final result of RAS activation or inhibition is determined by the signaling network context. Current therapies often 'train' cancer cells shifting them to a novel attractor, which has increased metastatic potential and drug resistance. The few therapy-surviving cancer cells are surrounded by massive cell death triggering a primordial adaptive and reparative general wound healing response. Overall, dynamic analysis of patient- and disease-stage specific intracellular and intercellular signaling networks may open new areas of anticancer therapy using multitarget drugs, drugs combinations, edgetic drugs, as well as help design 'gentler', differentiation and maintenance therapies.
Collapse
Affiliation(s)
- Peter Csermely
- Department of Medical Chemistry, Semmelweis University, P.O. Box 2, H-1428 Budapest, Hungary.
| | - Tamás Korcsmáros
- Gut Health and Food Safety Programme, Institute of Food Research, Norwich Research Park, Norwich NR4 7UA, UK; Earlham Institute/TGAC, The Genome Analysis Centre, Norwich Research Park, Norwich NR4 7UH, UK
| | - Ruth Nussinov
- Cancer and Inflammation Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA; Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
31
|
Affiliation(s)
- Ruth Nussinov
- Basic Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Cancer and Inflammation Program, National Cancer Institute at Frederick, Frederick, MD 21702, U.S.A
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Chung-Jung Tsai
- Basic Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Cancer and Inflammation Program, National Cancer Institute at Frederick, Frederick, MD 21702, U.S.A
| | - Hyunbum Jang
- Basic Science Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Cancer and Inflammation Program, National Cancer Institute at Frederick, Frederick, MD 21702, U.S.A
| |
Collapse
|