1
|
Guo Y, Wang R, Lv C, Xu C, Shen G, Wang G, Zhang W, Wang Q, Zhao Y. Jak/Stat-regulated Esftz-f1 negatively regulates the antibacterial immunity of Eriocheir sinensis against Vibrio parahaemolyticus. FISH & SHELLFISH IMMUNOLOGY 2025; 162:110366. [PMID: 40273962 DOI: 10.1016/j.fsi.2025.110366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 04/18/2025] [Accepted: 04/22/2025] [Indexed: 04/26/2025]
Abstract
With the growing global demand for premium aquatic products, the expanding international market presence of Eriocheir sinensis has led to a continuous appreciation of its economic value. However, E. sinensis is threatened by various diseases during its breeding, among which bacterial diseases seriously affect its immune function and impede its growth. Ftz-f1, an orphan nuclear receptor, plays a vital role in the embryonic development, molting process, gonadal development, and immune regulation of invertebrates. This study aims to identify the ftz-f1 homolog, called Esftz-f1, in E. sinensis. The Esftz-f1 ORF spans 1770 bp, encoding a 589-amino acid protein that shares 87.84 % sequence similarity with the Litopenaeus vannamei homolog and this protein contains two conserved functional domains. It is widely expressed in the multiple tissues of E. sinensis, with particularly high expression in the hepatopancreas. Subcellular localization analysis revealed nuclear localization of EsFtz-f1. The expression level of Esftz-f1 changes significantly upon stimulation by V. parahaemolyticus. When Jak and Stat are silenced or inhibited, the expression levels of Esftz-f1 are significantly downregulated. After Esftz-f1 is silenced, the expression levels of antimicrobial peptides, the phagocytic ability of hemocytes, bacterial clearance rate and the survival rate of crabs are significantly upregulated, suggesting that EsFtz-f1 plays a negative regulatory role in the resistance of E. sinensis to V. parahaemolyticus infection. We believe our study will help broaden the research scope of orphan nuclear receptors. It may also provide useful insights that aid further study of the immune mechanism of E. sinensis and provided references for the prevention of diseases during its breeding.
Collapse
Affiliation(s)
- Yanan Guo
- School of Life Sciences, East China Normal University, Shanghai, China
| | - Rongping Wang
- School of Aquatic and Life Sciences, Shanghai Ocean University, Shanghai, China
| | - Chengyu Lv
- School of Aquatic and Life Sciences, Shanghai Ocean University, Shanghai, China
| | - Chaohui Xu
- School of Aquatic and Life Sciences, Shanghai Ocean University, Shanghai, China
| | - Guoqing Shen
- School of Life Sciences, East China Normal University, Shanghai, China
| | - Guangyu Wang
- School of Aquatic and Life Sciences, Shanghai Ocean University, Shanghai, China
| | - Wen Zhang
- School of Life Sciences, East China Normal University, Shanghai, China
| | - Qun Wang
- School of Aquatic and Life Sciences, Shanghai Ocean University, Shanghai, China.
| | - Yunlong Zhao
- School of Life Sciences, East China Normal University, Shanghai, China.
| |
Collapse
|
2
|
Meng T, Zhang Y, Wang H, Wu W, Peng W, Yue J, Huang C, Liu W, Liang C, Yang C, Chen J. Irf7 aggravates prostatitis by promoting Hif-1α-mediated glycolysis to facilitate M1 polarization. Cell Mol Life Sci 2025; 82:90. [PMID: 39985573 PMCID: PMC11846824 DOI: 10.1007/s00018-025-05608-w] [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: 10/17/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND Chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS) is a common disorder associated with voiding symptoms and pain in the pelvic or perineal area. Macrophages, particularly the pro-inflammatory M1 subtype, are crucial initiating of CP/CPPS. Interferon regulatory factor 7 (Irf7) has been implicated in promoting M1 polarization, contributing to the onset and progression of autoimmunity. However, the role of Irf7 in the etiology and progression of CP/CPPS remains unclear. METHOD We established the experimental autoimmune prostatitis (EAP) mouse model by subcutaneous injection of prostate antigen combined with complete Freund's adjuvant. Six weeks after the first immunization, we analyzed the prostates, spleen, and blood to assess the degree of prostate inflammation, Irf7 expression levels, glycolysis, and M1 polarization to evaluate whether Irf7 could exacerbate the development of EAP by enhancing Hif-1α transcription, thereby increasing glycolysis and M1 polarization. Further investigations included sh-Irf7 intervention, Dimethyloxalylglycine (a Hif-1α agonist), and in vitro M1 polarization experiments. We also employed ChIP assays, dual-luciferase reporter assays, and q-PCR to explore if Irf7 could directly interact with the Hif-1α promoter in macrophages. RESULTS In the EAP mouse and cell models, elevated Irf7 expression was observed in inflamed tissues and cells. Reducing Irf7 expression decreased M1 cell glycolysis by inhibiting the nuclear translocation of Hif-1α, thus mitigating M1 cell polarization. Additionally, Irf7 was identified as a transcription factor that regulates Hif-1α transcription by interacting with its promoter in macrophages, confirmed through ChIP and dual-luciferase assays. Co-culturing macrophage cells with 3T3 fibroblasts with reduced Irf7 levels resulted in decreased fibrosis, and a significant reduction in prostate tissue fibrosis was noted in mice with Irf7 knockdown. CONCLUSION Our findings indicate that Irf7 can contribute to the development and progression of CP/CPPS by promoting glycolysis, which can enhance both M1 polarization as well as interstitial fibrosis in the prostate. This process was found to be mediated by the upregulation of Hif-1α transcription, presenting new potential therapeutic targets for managing CP/CPPS.
Collapse
Affiliation(s)
- Tong Meng
- Department of Urology, Institute of Urology, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, People's Republic of China
- Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Yi Zhang
- Department of Urology, Institute of Urology, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, People's Republic of China
- Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Huihui Wang
- Department of Urology, Institute of Urology, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, People's Republic of China
- Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Weikang Wu
- Department of Urology, Institute of Urology, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, People's Republic of China
- Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Wei Peng
- Department of Urology, Institute of Urology, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, People's Republic of China
- Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Jiabin Yue
- Department of Urology, Institute of Urology, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, People's Republic of China
- Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Cong Huang
- Department of Urology, Institute of Urology, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, People's Republic of China
- Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Wanqing Liu
- Department of Urology, Institute of Urology, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, People's Republic of China
- Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China
| | - Chaozhao Liang
- Department of Urology, Institute of Urology, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, People's Republic of China.
- Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China.
| | - Cheng Yang
- Department of Urology, Institute of Urology, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, People's Republic of China.
- Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China.
| | - Jing Chen
- Department of Urology, Institute of Urology, Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, Anhui Province, 230022, People's Republic of China.
- Center for Scientific Research of the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People's Republic of China.
| |
Collapse
|
3
|
Wu Y, He L, Li R, Li J, Zhao Q, Shao B. A20 as a Potential Therapeutic Target for COVID-19. Immun Inflamm Dis 2025; 13:e70127. [PMID: 39853876 PMCID: PMC11760982 DOI: 10.1002/iid3.70127] [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: 04/12/2024] [Revised: 11/29/2024] [Accepted: 01/03/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a major concern due to its astonishing prevalence and high fatality rate, especially among elderly people. Patients suffering from COVID-19 may exhibit immunosuppression in the initial stage of infection, while a cytokine storm can occur when the disease progresses to a severe stage. This inopportune immune rhythm not only makes patients more susceptible to the virus but also leads to numerous complications resulting from the excessive production of inflammatory factors. A20, which is widely accepted as a pivotal regulator of inflammation, has been shown to be implicated in the processes of antiviral responses and immunosuppression. Thus, A20 may participate in regulating the pathological processes of COVID-19. METHODS This narrative literature review summarizes recent evidence on the mechanisms of A20 in regulating the pathological processes of COVID-19. We also downloaded single-cell RNA-seq data sets from healthy individuals and patients with varying severities of COVID-19 from the NCBI GEO database to further dissect A20's regulatory mechanisms of these intricate cytokine pathways that are closely associated with SARS-CoV-2 infection. RESULTS A20 might be one of the most critical anti-infectious and anti-inflammatory factors involved in the pathogenesis of COVID-19. It effectively suppresses the immune damage and inflammatory storm caused by viral infection. CONCLUSIONS Understanding the relationship between A20-regulated signaling pathways and pathological processes of COVID-19 can provide insight into potential targets for intervention. Precise regulation of A20 to induce antiviral activity and an anti-inflammatory response could mediate the pathogenesis of COVID-19 and could become an effective treatment.
Collapse
Affiliation(s)
- Yongyao Wu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Lilan He
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Rong Li
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Jiuxuan Li
- Laboratory of Radiation Biology, Laboratory Medicine Centre, Department of Blood TransfusionThe Second Affiliated HospitalArmy Military Medical UniversityChongqingChina
| | - Qing Zhao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Bin Shao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| |
Collapse
|
4
|
Chen Z, Wang W, Zeng K, Zhu J, Wang X, Huang W. Potential antiviral activity of rhamnocitrin against influenza virus H3N2 by inhibiting cGAS/STING pathway in vitro. Sci Rep 2024; 14:28287. [PMID: 39550441 PMCID: PMC11569172 DOI: 10.1038/s41598-024-79788-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 11/12/2024] [Indexed: 11/18/2024] Open
Abstract
Influenza remains a serious issue for public health and it's urgent to discover more effected drugs against influenza virus. Rhamnocitrin, as a flavonoid, its effect on influenza virus infection remains poorly explored. In this study, rhamnocitrin showed antiviral effect and anti-apoptosis on influenza virus A/Aichi/2/1968 (H3N2) in MDCK cells and A549 cells. In addition, molecular docking revealed that rhamnocitrin have good binding activity with the target proteins cGAS and STING, molecular dynamic simulation and surface plasmon resonance showed that rhamnocitrin could form a stable complex with the above proteins. Moreover, the qPCR and western blot assays further verified that rhamnocitrin could reduce type I IFN and proinflammatory cytokines production by inhibiting the cGAS/STING pathway. Taken together, the results suggest that rhamnocitrin could be a potential anti-viral agent against influenza.
Collapse
Affiliation(s)
- Zexing Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510180, China
- Institute of Integration of Traditional and Western Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wanqi Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510180, China
- Institute of Integration of Traditional and Western Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kefeng Zeng
- Institute of Integration of Traditional and Western Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jinyi Zhu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510180, China
- Institute of Integration of Traditional and Western Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xinhua Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510180, China
- Institute of Integration of Traditional and Western Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wanyi Huang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510180, China.
- Institute of Integration of Traditional and Western Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
5
|
Zhang S, Yu B, Sheng C, Yao C, Liu Y, Wang J, Zeng Q, Mao Y, Bei J, Zhu B, Chen S. SHISA3 Reprograms Tumor-Associated Macrophages Toward an Antitumoral Phenotype and Enhances Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403019. [PMID: 39054639 PMCID: PMC11423144 DOI: 10.1002/advs.202403019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/22/2024] [Indexed: 07/27/2024]
Abstract
The main challenge for immune checkpoint blockade (ICB) therapy lies in immunosuppressive tumor microenvironment (TME). Repolarizing M2-like tumor-associated macrophages (TAMs) into inflammatory M1 phenotype is a promising strategy for cancer immunotherapy. Here, this study shows that the tumor suppressive protein SHISA3 regulates the antitumor functions of TAMs. Local delivery of mRNA encoding Shisa3 enables cancer immunotherapy by reprogramming TAMs toward an antitumoral phenotype, thus enhancing the efficacy of programmed cell death 1 (PD-1) antibody. Enforced expression of Shisa3 in TAMs increases their phagocytosis and antigen presentation abilities and promotes CD8+ T cell-mediated antitumor immunity. The expression of SHISA3 is induced by damage/pathogen-associated molecular patterns (DAMPs/PAMPs) in macrophages via nuclear factor-κB (NF-κB) transcription factors. Reciprocally, SHISA3 forms a complex with heat shock protein family A member 8 (HSPA8) to activate NF-κB signaling thus maintaining M1 polarization of macrophages. Knockout Shisa3 largely abolishes the antitumor efficacy of combination immunotherapy with Toll-like receptor 4 (TLR4) agonist monophosphoryl lipid A (MPLA) and PD-1 antibody. It further found that higher expression of SHISA3 in antitumoral TAMs is associated with better overall survival in lung cancer patients. Taken together, the findings describe the role of SHISA3 in reprogramming TAMs that ameliorate cancer immunotherapy.
Collapse
Affiliation(s)
- Shimeng Zhang
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| | - Bingbing Yu
- Key Laboratory of Molecular Biophysicsthe Ministry of EducationCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanHubei430074P. R. China
| | - Chunjie Sheng
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| | - Chen Yao
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| | - Yang Liu
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| | - Jing Wang
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| | - Qi Zeng
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| | - Yizhi Mao
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| | - Jinxin Bei
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| | - Bin Zhu
- Key Laboratory of Molecular Biophysicsthe Ministry of EducationCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanHubei430074P. R. China
- Shenzhen Huazhong University of Science and Technology Research InstituteShenzhen518063P. R. China
| | - Shuai Chen
- State Key Laboratory of Oncology in South ChinaGuangdong Provincial Clinical Research Center for CancerSun Yat‐sen University Cancer CenterGuangzhou510060P. R. China
| |
Collapse
|
6
|
Joshi R, Brezani V, Mey GM, Guixé-Muntet S, Ortega-Ribera M, Zhuang Y, Zivny A, Werneburg S, Gracia-Sancho J, Szabo G. IRF3 regulates neuroinflammatory responses and the expression of genes associated with Alzheimer's disease. J Neuroinflammation 2024; 21:212. [PMID: 39215356 PMCID: PMC11363437 DOI: 10.1186/s12974-024-03203-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
The pathological role of interferon signaling is emerging in neuroinflammatory disorders, yet, the specific role of Interferon Regulatory Factor 3 (IRF3) in neuroinflammation remains poorly understood. Here, we show that global IRF3 deficiency delays TLR4-mediated signaling in microglia and attenuates the hallmark features of LPS-induced inflammation such as cytokine release, microglial reactivity, astrocyte activation, myeloid cell infiltration, and inflammasome activation. Moreover, expression of a constitutively active IRF3 (S388D/S390D: IRF3-2D) in microglia induces a transcriptional program reminiscent of the Activated Response Microglia and the expression of genes associated with Alzheimer's disease, notably apolipoprotein-e. Using bulk-RNAseq of IRF3-2D brain myeloid cells, we identified Z-DNA binding protein-1 (ZBP1) as a target of IRF3 that is relevant across various neuroinflammatory disorders. Lastly, we show IRF3 phosphorylation and IRF3-dependent ZBP1 induction in response to Aβ in primary microglia cultures. Together, our results identify IRF3 as an important regulator of LPS and Aβ -mediated neuroinflammatory responses and highlight IRF3 as a central regulator of disease-specific gene activation in different neuroinflammatory diseases.
Collapse
Affiliation(s)
- Radhika Joshi
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, USA
| | - Veronika Brezani
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, USA
| | - Gabrielle M Mey
- Department of Opthalmology and Visual Sciences, Kellogg Eye Center Michigan Neuroscience Institute, University of Michigan, Ann Arbor, USA
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Sergi Guixé-Muntet
- Liver Vascular Biology, IDIBAPS Biomedical Research Institute-CIBEREHD, Barcelona, Spain
| | - Marti Ortega-Ribera
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, USA
| | - Yuan Zhuang
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, USA
| | - Adam Zivny
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, USA
| | - Sebastian Werneburg
- Department of Opthalmology and Visual Sciences, Kellogg Eye Center Michigan Neuroscience Institute, University of Michigan, Ann Arbor, USA
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Jordi Gracia-Sancho
- Liver Vascular Biology, IDIBAPS Biomedical Research Institute-CIBEREHD, Barcelona, Spain
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Gyongyi Szabo
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, USA.
| |
Collapse
|
7
|
Yan Z, Yue J, Zhang Y, Hou Z, Li D, Yang Y, Li X, Idris A, Li H, Li S, Xie J, Feng R. Pseudorabies virus VHS protein abrogates interferon responses by blocking NF-κB and IRF3 nuclear translocation. Virol Sin 2024; 39:587-599. [PMID: 38823782 PMCID: PMC11401465 DOI: 10.1016/j.virs.2024.05.009] [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: 01/05/2024] [Accepted: 05/23/2024] [Indexed: 06/03/2024] Open
Abstract
Herpesviruses antagonize host antiviral responses through a myriad of molecular strategies culminating in the death of the host cells. Pseudorabies virus (PRV) is a significant veterinary pathogen in pigs, causing neurological sequalae that ultimately lead to the animal's demise. PRV is known to trigger apoptotic cell death during the late stages of infection. The virion host shutdown protein (VHS) encoded by UL41 plays a crucial role in the PRV infection process. In this study, we demonstrate that UL41 inhibits PRV-induced activation of inflammatory cytokine and negatively regulates the cGAS-STING-mediated antiviral activity by targeting IRF3, thereby inhibiting the translocation and phosphorylation of IRF3. Notably, mutating the conserved amino acid sites (E192, D194, and D195) in the RNase domain of UL41 or knocking down UL41 inhibits the immune evasion of PRV, suggesting that UL41 may play a crucial role in PRV's evasion of the host immune response during infection. These results enhance our understanding of how PRV structural proteins assist the virus in evading the host immune response.
Collapse
Affiliation(s)
- Zhenfang Yan
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China
| | - Jiayu Yue
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China
| | - Yaxin Zhang
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China
| | - Zhengyang Hou
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China
| | - Dianyu Li
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China
| | - Yanmei Yang
- College of Life Science and Engineering, Northwest Minzu University, Lanzhou, 730030, China
| | - Xiangrong Li
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China; Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China; Engineering Research Center of Key Technology and Industrialization of Cell-based Vaccine, Ministry of Education, Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China
| | - Adi Idris
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Kelvin Grove, Queensland, 4702, Australia
| | - Huixia Li
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China
| | - Shasha Li
- College of Life Science and Engineering, Northwest Minzu University, Lanzhou, 730030, China
| | - Jingying Xie
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China; College of Life Science and Engineering, Northwest Minzu University, Lanzhou, 730030, China.
| | - Ruofei Feng
- Key Laboratory of Biotechnology and Bioengineering of State Ethnic Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China; Gansu Tech Innovation Center of Animal Cell, Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China; Engineering Research Center of Key Technology and Industrialization of Cell-based Vaccine, Ministry of Education, Biomedical Research Center, Northwest Minzu University, Lanzhou, 730030, China.
| |
Collapse
|
8
|
Carrasco JL, Ambrós S, Gutiérrez PA, Elena SF. Adaptation of turnip mosaic virus to Arabidopsis thaliana involves rewiring of VPg-host proteome interactions. Virus Evol 2024; 10:veae055. [PMID: 39091990 PMCID: PMC11291303 DOI: 10.1093/ve/veae055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/23/2024] [Accepted: 07/16/2024] [Indexed: 08/04/2024] Open
Abstract
The outcome of a viral infection depends on a complex interplay between the host physiology and the virus, mediated through numerous protein-protein interactions. In a previous study, we used high-throughput yeast two-hybrid (HT-Y2H) to identify proteins in Arabidopsis thaliana that bind to the proteins encoded by the turnip mosaic virus (TuMV) genome. Furthermore, after experimental evolution of TuMV lineages in plants with mutations in defense-related or proviral genes, most mutations observed in the evolved viruses affected the VPg cistron. Among these mutations, D113G was a convergent mutation selected in many lineages across different plant genotypes, including cpr5-2 with constitutive expression of systemic acquired resistance. In contrast, mutation R118H specifically emerged in the jin1 mutant with affected jasmonate signaling. Using the HT-Y2H system, we analyzed the impact of these two mutations on VPg's interaction with plant proteins. Interestingly, both mutations severely compromised the interaction of VPg with the translation initiation factor eIF(iso)4E, a crucial interactor for potyvirus infection. Moreover, mutation D113G, but not R118H, adversely affected the interaction with RHD1, a zinc-finger homeodomain transcription factor involved in regulating DNA demethylation. Our results suggest that RHD1 enhances plant tolerance to TuMV infection. We also discuss our findings in a broad virus evolution context.
Collapse
Affiliation(s)
- José L Carrasco
- Instituto de Biología Integrativa de Sistemas (CSIC—Universitat de València), Catedratico Agustin Escardino 9, Paterna, València 46182, Spain
| | - Silvia Ambrós
- Instituto de Biología Integrativa de Sistemas (CSIC—Universitat de València), Catedratico Agustin Escardino 9, Paterna, València 46182, Spain
| | - Pablo A Gutiérrez
- Laboratorio de Microbiología Industrial, Facultad de Ciencias, Universidad Nacional de Colombia, Carrera 65 Nro. 59A - 110, Medellín, Antioquia 050034, Colombia
| | - Santiago F Elena
- Instituto de Biología Integrativa de Sistemas (CSIC—Universitat de València), Catedratico Agustin Escardino 9, Paterna, València 46182, Spain
- The Santa Fe Institute, 1399 Hyde Park Rd, Santa Fe, NM 87501, United States
| |
Collapse
|
9
|
Crow YJ, Casanova JL. Human life within a narrow range: The lethal ups and downs of type I interferons. Sci Immunol 2024; 9:eadm8185. [PMID: 38968338 DOI: 10.1126/sciimmunol.adm8185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 06/13/2024] [Indexed: 07/07/2024]
Abstract
The past 20 years have seen the definition of human monogenic disorders and their autoimmune phenocopies underlying either defective or enhanced type I interferon (IFN) activity. These disorders delineate the impact of type I IFNs in natural conditions and demonstrate that only a narrow window of type I IFN activity is beneficial. Insufficient type I IFN predisposes humans to life-threatening viral diseases (albeit unexpectedly few) with a central role in immunity to respiratory and cerebral viral infection. Excessive type I IFN, perhaps counterintuitively, appears to underlie a greater number of autoinflammatory and/or autoimmune conditions known as type I interferonopathies, whose study has revealed multiple molecular programs involved in the induction of type I IFN signaling. These observations suggest that the manipulation of type I IFN activity to within a physiological range may be clinically relevant for the prevention and treatment of viral and inflammatory diseases.
Collapse
Affiliation(s)
- Yanick J Crow
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- Laboratory of Neurogenetics and Neuroinflammation, Imagine Institute, INSERM UMR 1163, Paris, France
- University Paris Cité, Paris, France
| | - Jean-Laurent Casanova
- University Paris Cité, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Howard Hughes Medical Institute, New York, NY, USA
- Department of Pediatrics, Necker Hospital for Sick Children, Paris, France
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Imagine Institute, INSERM UMR 1163, Necker Hospital for Sick Children, Paris, France
| |
Collapse
|
10
|
Qing F, Sui L, He W, Chen Y, Xu L, He L, Xiao Q, Guo T, Liu Z. IRF7 Exacerbates Candida albicans Infection by Compromising CD209-Mediated Phagocytosis and Autophagy-Mediated Killing in Macrophages. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1932-1944. [PMID: 38709167 DOI: 10.4049/jimmunol.2300826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 03/20/2024] [Indexed: 05/07/2024]
Abstract
IFN regulatory factor 7 (IRF7) exerts anti-infective effects by promoting the production of IFNs in various bacterial and viral infections, but its role in highly morbid and fatal Candida albicans infections is unknown. We unexpectedly found that Irf7 gene expression levels were significantly upregulated in tissues or cells after C. albicans infection in humans and mice and that IRF7 actually exacerbates C. albicans infection in mice independent of its classical function in inducing IFNs production. Compared to controls, Irf7-/- mice showed stronger phagocytosis of fungus, upregulation of C-type lectin receptor CD209 expression, and enhanced P53-AMPK-mTOR-mediated autophagic signaling in macrophages after C. albicans infection. The administration of the CD209-neutralizing Ab significantly hindered the phagocytosis of Irf7-/- mouse macrophages, whereas the inhibition of p53 or autophagy impaired the killing function of these macrophages. Thus, IRF7 exacerbates C. albicans infection by compromising the phagocytosis and killing capacity of macrophages via regulating CD209 expression and p53-AMPK-mTOR-mediated autophagy, respectively. This finding reveals a novel function of IRF7 independent of its canonical IFNs production and its unexpected role in enhancing fungal infections, thus providing more specific and effective targets for antifungal therapy.
Collapse
Affiliation(s)
- Furong Qing
- School of Basic Medicine; Gannan Medical University, Ganzhou, Jiangxi
| | - Lina Sui
- School of Basic Medicine; Gannan Medical University, Ganzhou, Jiangxi
| | - Wenji He
- School of Basic Medicine; Gannan Medical University, Ganzhou, Jiangxi
- School of Graduate, China Medical University, Shenyang, Liaoning
| | - Yayun Chen
- School of Basic Medicine; Gannan Medical University, Ganzhou, Jiangxi
- School of Graduate, China Medical University, Shenyang, Liaoning
| | - Li Xu
- Center for Scientific Research, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Liangmei He
- School of Graduate, China Medical University, Shenyang, Liaoning
- Department of Gastroenterology, First Affiliated Hospital, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Qiuxiang Xiao
- School of Graduate, China Medical University, Shenyang, Liaoning
- Department of Pathology, First Affiliated Hospital, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Tianfu Guo
- School of Basic Medicine; Gannan Medical University, Ganzhou, Jiangxi
| | - Zhiping Liu
- School of Basic Medicine; Gannan Medical University, Ganzhou, Jiangxi
- Center for Scientific Research, Gannan Medical University, Ganzhou, Jiangxi, China
- Center for Immunology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|
11
|
MacFawn I, Farris J, Pifer P, Margaryan NV, Akhter H, Wang L, Dziadowicz S, Denvir J, Hu G, Frisch SM. Grainyhead-like-2, an epithelial master programmer, promotes interferon induction and suppresses breast cancer recurrence. Mol Immunol 2024; 170:156-169. [PMID: 38692097 PMCID: PMC11106721 DOI: 10.1016/j.molimm.2024.04.012] [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: 01/29/2024] [Revised: 04/22/2024] [Accepted: 04/22/2024] [Indexed: 05/03/2024]
Abstract
Type-I and -III interferons play a central role in immune rejection of pathogens and tumors, thus promoting immunogenicity and suppressing tumor recurrence. Double strand RNA is an important ligand that stimulates tumor immunity via interferon responses. Differentiation of embryonic stem cells to pluripotent epithelial cells activates the interferon response during development, raising the question of whether epithelial vs. mesenchymal gene signatures in cancer potentially regulate the interferon pathway as well. Here, using genomics and signaling approaches, we show that Grainyhead-like-2 (GRHL2), a master programmer of epithelial cell identity, promotes type-I and -III interferon responses to double-strand RNA. GRHL2 enhanced the activation of IRF3 and relA/NF-kB and the expression of IRF1; a functional GRHL2 binding site in the IFNL1 promoter was also identified. Moreover, time to recurrence in breast cancer correlated positively with GRHL2 protein expression, indicating that GRHL2 is a tumor recurrence suppressor, consistent with its enhancement of interferon responses. These observations demonstrate that epithelial cell identity supports interferon responses in the context of cancer.
Collapse
Affiliation(s)
- Ian MacFawn
- Department of Immunology, University of Pittsburgh, 5051 Centre Avenue, Pittsburgh, PA 15213, USA; UPMC Hillman Cancer Center, 5115 Centre Avenue, Pittsburgh, PA 15232, USA
| | - Joshua Farris
- Wake Forest University, Department of Radiation Oncology, 1 Medical Center Blvd., Winston-Salem, NC 27157, USA
| | - Phillip Pifer
- Department of Radiation Oncology, WVU Cancer Institute, 1 Medical Drive, Morgantown, WV, USA
| | - Naira V Margaryan
- WVU Cancer Institute, West Virginia University, 64 Medical Center Drive, Morgantown, WV 26506, USA
| | - Halima Akhter
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, 64 Medical Center Drive, Box 9142, Morgantown, WV 26505, USA
| | - Lei Wang
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, 64 Medical Center Drive, Box 9142, Morgantown, WV 26505, USA
| | - Sebastian Dziadowicz
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, 64 Medical Center Drive, Box 9142, Morgantown, WV 26505, USA
| | - James Denvir
- Byrd Biotechnology Center, Marshall University, One John Marshall Drive, Huntington, WV 25701, USA
| | - Gangqing Hu
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, 64 Medical Center Drive, Box 9142, Morgantown, WV 26505, USA.
| | - Steven M Frisch
- Department of Biochemistry and Molecular Medicine, 64 Medical Center Drive, Box 9142, West Virginia University, Morgantown, WV 26506.
| |
Collapse
|
12
|
Fan S, Popli S, Chakravarty S, Chakravarti R, Chattopadhyay S. Non-transcriptional IRF7 interacts with NF-κB to inhibit viral inflammation. J Biol Chem 2024; 300:107200. [PMID: 38508315 PMCID: PMC11040127 DOI: 10.1016/j.jbc.2024.107200] [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: 08/28/2023] [Revised: 02/23/2024] [Accepted: 03/10/2024] [Indexed: 03/22/2024] Open
Abstract
Interferon (IFN) regulatory factors (IRF) are key transcription factors in cellular antiviral responses. IRF7, a virus-inducible IRF, expressed primarily in myeloid cells, is required for transcriptional induction of interferon α and antiviral genes. IRF7 is activated by virus-induced phosphorylation in the cytoplasm, leading to its translocation to the nucleus for transcriptional activity. Here, we revealed a nontranscriptional activity of IRF7 contributing to its antiviral functions. IRF7 interacted with the pro-inflammatory transcription factor NF-κB-p65 and inhibited the induction of inflammatory target genes. Using knockdown, knockout, and overexpression strategies, we demonstrated that IRF7 inhibited NF-κB-dependent inflammatory target genes, induced by virus infection or toll-like receptor stimulation. A mutant IRF7, defective in transcriptional activity, interacted with NF-κB-p65 and suppressed NF-κB-induced gene expression. A single-action IRF7 mutant, active in anti-inflammatory function, but defective in transcriptional activity, efficiently suppressed Sendai virus and murine hepatitis virus replication. We, therefore, uncovered an anti-inflammatory function for IRF7, independent of transcriptional activity, contributing to the antiviral response of IRF7.
Collapse
Affiliation(s)
- Shumin Fan
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Science, Toledo, Ohio, USA
| | - Sonam Popli
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Science, Toledo, Ohio, USA
| | - Sukanya Chakravarty
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Science, Toledo, Ohio, USA; Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| | - Ritu Chakravarti
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Science, Toledo, Ohio, USA
| | - Saurabh Chattopadhyay
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Science, Toledo, Ohio, USA; Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, USA.
| |
Collapse
|
13
|
Xu X, Qiao D, Brasier AR. Cooperative interaction of interferon regulatory factor -1 and bromodomain-containing protein 4 on RNA polymerase activation for intrinsic innate immunity. Front Immunol 2024; 15:1366235. [PMID: 38601157 PMCID: PMC11004252 DOI: 10.3389/fimmu.2024.1366235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 03/14/2024] [Indexed: 04/12/2024] Open
Abstract
Introduction The human orthopneumovirus, Respiratory Syncytial Virus (RSV), is the causative agent of severe lower respiratory tract infections (LRTI) and exacerbations of chronic lung diseases. In immune competent hosts, RSV productively infects highly differentiated epithelial cells, where it elicits robust anti-viral, cytokine and remodeling programs. By contrast, basal cells are relatively resistant to RSV infection, in part, because of constitutive expression of an intrinsic innate immune response (IIR) consisting of a subgroup of interferon (IFN) responsive genes. The mechanisms controlling the intrinsic IIR are not known. Methods Here, we use human small airway epithelial cell hSAECs as a multipotent airway stem cell model to examine regulatory control of an intrinsic IIR pathway. Results We find hSAECs express patterns of intrinsic IIRs, highly conserved with pluri- and multi-potent stem cells. We demonstrate a core intrinsic IIR network consisting of Bone Marrow Stromal Cell Antigen 2 (Bst2), Interferon Induced Transmembrane Protein 1 (IFITM1) and Toll-like receptor (TLR3) expression are directly under IRF1 control. Moreover, expression of this intrinsic core is rate-limited by ambient IRF1• phospho-Ser 2 CTD RNA Polymerase II (pSer2 Pol II) complexes binding to their proximal promoters. In response to RSV infection, the abundance of IRF1 and pSer2 Pol II binding is dramatically increased, with IRF1 complexing to the BRD4 chromatin remodeling complex (CRC). Using chromatin immunoprecipitation in IRF1 KD cells, we find that the binding of BRD4 is IRF1 independent. Using a small molecule inhibitor of the BRD4 acetyl lysine binding bromodomain (BRD4i), we further find that BRD4 bromodomain interactions are required for stable BRD4 promoter binding to the intrinsic IIR core promoters, as well as for RSV-inducible pSer2 Pol II recruitment. Surprisingly, BRD4i does not disrupt IRF1-BRD4 interactions, but disrupts both RSV-induced BRD4 and IRF1 interactions with pSer2 Pol II. Conclusions We conclude that the IRF1 functions in two modes- in absence of infection, ambient IRF1 mediates constitutive expression of the intrinsic IIR, whereas in response to RSV infection, the BRD4 CRC independently activates pSer2 Pol II to mediates robust expression of the intrinsic IIR. These data provide insight into molecular control of anti-viral defenses of airway basal cells.
Collapse
Affiliation(s)
- Xiaofang Xu
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health (SMPH), Madison, WI, United States
| | - Dianhua Qiao
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health (SMPH), Madison, WI, United States
| | - Allan R. Brasier
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health (SMPH), Madison, WI, United States
- Institute for Clinical and Translational Research, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
14
|
Joshi R, Brezani V, Mey GM, Guixé-Muntet S, Ortega-Ribera M, Zhuang Y, Zivny A, Werneburg S, Gracia-Sancho J, Szabo G. IRF3 regulates neuroinflammatory responses and the expression of genes associated with Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.08.582968. [PMID: 38654824 PMCID: PMC11037866 DOI: 10.1101/2024.03.08.582968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
The pathological role of interferon signaling is emerging in neuroinflammatory disorders, yet, the specific role of Interferon Regulatory Factor 3 (IRF3) in neuroinflammation remains poorly understood. Here, we show that global IRF3 deficiency delays TLR4-mediated signaling in microglia and attenuates the hallmark features of LPS-induced inflammation such as cytokine release, microglial reactivity, astrocyte activation, myeloid cell infiltration, and inflammasome activation. Moreover, expression of a constitutively active IRF3 (S388D/S390D:IRF3-2D) in microglia induces a transcriptional program reminiscent of the Activated Response Microglia and the expression of genes associated with Alzheimer's Disease, notably apolipoprotein-e. Lastly, using bulk-RNAseq of IRF3-2D brain myeloid cells, we identified Z-DNA binding protein-1 as a target of IRF3 that is relevant across various neuroinflammatory disorders. Together, our results identify IRF3 as an important regulator of LPS-mediated neuroinflammatory responses and highlight IRF3 as a central regulator of disease-specific gene activation in different neuroinflammatory diseases.
Collapse
Affiliation(s)
- Radhika Joshi
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, USA
| | - Veronika Brezani
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, USA
| | - Gabrielle M Mey
- Department of Opthalmology and Visual Sciences, Kellogg Eye Center Michigan Neuroscience Institute, University of Michigan, Ann Arbor, USA
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Sergi Guixé-Muntet
- Liver Vascular Biology, IDIBAPS Biomedical Research Institute- CIBEREHD, Barcelona, Spain
| | - Marti Ortega-Ribera
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, USA
| | - Yuan Zhuang
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, USA
| | - Adam Zivny
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, USA
| | - Sebastian Werneburg
- Department of Opthalmology and Visual Sciences, Kellogg Eye Center Michigan Neuroscience Institute, University of Michigan, Ann Arbor, USA
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Jordi Gracia-Sancho
- Liver Vascular Biology, IDIBAPS Biomedical Research Institute- CIBEREHD, Barcelona, Spain
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Gyongyi Szabo
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, USA
| |
Collapse
|
15
|
Zhang S, Liu Y, Ma Z, Gao S, Chen L, Zhong H, Zhang C, Li T, Chen W, Zhang Y, Lin N. Osteoking promotes bone formation and bone defect repair through ZBP1-STAT1-PKR-MLKL-mediated necroptosis. Chin Med 2024; 19:13. [PMID: 38238785 PMCID: PMC10797925 DOI: 10.1186/s13020-024-00883-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 12/29/2023] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND Osteoking has been used for fracture therapy with a satisfying clinical efficacy. However, its therapeutic properties and the underlying mechanisms remain elusive. METHOD A bone defect rat model was established to evaluate the pharmacological effects of Osteoking by the dynamic observation of X-ray, micro-CT and histopathologic examination. Transcriptome profiling was performed to identify bone defect-related genes and Osteoking effective targets. Then, a "disease-related gene-drug target" interaction network was constructed and a list of key network targets were screened, which were experimentally verified. RESULTS Osteoking effectively promoted bone defect repair in rats by accelerating the repair of cortical bone and the growth of trabeculae. Histopathologically, the bone defect rats displayed lower histopathologic scores in cortical bone, cancellous bone and bone connection than normal controls. In contrast, Osteoking exerted a favorable effect with a dose-dependent manner. The abnormal serum levels of bone turnover markers, bone growth factors and bone metabolism-related biochemical indexes in bone defect rats were also reversed by Osteoking treatment. Following the transcriptome-based network investigation, we hypothesized that osteoking might attenuate the levels of ZBP1-STAT1-PKR-MLKL-mediated necroptosis involved into bone defect. Experimentally, the expression levels of ZBP1, STAT1, PKR and the hallmark inflammatory cytokines for the end of necroptosis were distinctly elevated in bone defect rats, but were all effectively reversed by Osteoking treatment, which were also suppressed the activities of RIPK1, RIPK3 and MLKL in bone tissue supernatants. CONCLUSIONS Osteoking may promote bone formation and bone defect repair by regulating ZBP1-STAT1-PKR axis, leading to inhibit RIPK1/RIPK3/MLKL activation-mediated necroptosis.
Collapse
Affiliation(s)
- Suya Zhang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou, 510405, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Beijing, 100700, China
| | - Yudong Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Beijing, 100700, China
| | - Zhaochen Ma
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Beijing, 100700, China
| | - Shuangrong Gao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Beijing, 100700, China
| | - Lin Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Beijing, 100700, China
| | - Honggang Zhong
- BioMechanics Lab, Wang Jing Hospital, China Academy of Chinese Medical Sciences, Beijing, 100010, China
| | - Chu Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Beijing, 100700, China
| | - Tao Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Beijing, 100700, China
| | - Weiheng Chen
- Third Affiliated Hospital of Beijing University of Chinese Medicine, No. 51 Anwai Xiaoguanjie, Chaoyang District, Beijing, 100029, China
| | - Yanqiong Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Beijing, 100700, China.
| | - Na Lin
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, 12 Airport Road, Baiyun District, Guangzhou, 510405, China.
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Beijing, 100700, China.
| |
Collapse
|
16
|
Shan J, Ma W, Guo Y, Chang X, Xie J, Chen Y, Hu X, Yu Q. Unveiling the immunomodulatory mechanism of polysaccharides from Polygonum cyrtonema based on RNA-seq. Food Res Int 2024; 175:113755. [PMID: 38129053 DOI: 10.1016/j.foodres.2023.113755] [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: 08/26/2023] [Revised: 11/04/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023]
Abstract
This work aimed to illuminate the mechanism of Polygonatum cyrtonema polysaccharide (PCP-80%) triggered immune activation. Results showed that PCP-80% enhanced the protein expression of COX-2 and iNOS, along with increasing the release of NO, ROS, cytokines (TNF-α, IL-6) in RAW264.7 cells. RNA-seq analysis revealed 2160 differentially expressed genes (DEGs) following PCP-80% treatment, comprising 1142 up-regulated and 1018 down-regulated genes. In addition, for investigating possible regulatory mechanisms, the NF-κB, MAPKs, and JAK-STAT signaling pathways were also chosen based on bioinformatics analysis. Furthermore, these findings were further corroborated through Western blot experiments, validating the activation of JAK-STAT (reduction of JAK1 in cells and elevation of p-STAT3 in the nucleus), MAPK (elevation of p-p38, p-ERK1/2, and p-JNK), and NF-κB (elevation of p-IκBα in cells, reduction of cytoplasmic p65, and increase of nuclear content of p-p65) in macrophage activation induced by PCP-80%. Besides, the production of NO and TNF-α was decreased by the inhibitor of the three pathways. In conclusion, these findings provide strong evidence that PCP-80% effectively modulates the immune response of macrophages, with significant involvement of the JAK-STAT, MAPKs, and NF-κB signaling pathways.
Collapse
Affiliation(s)
- Jialuo Shan
- State Key Laboratory of Food Science and Resources, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University College of Food Science and Technology, 235 Nanjing East Road, Nanchang 330047, China
| | - Wenjie Ma
- State Key Laboratory of Food Science and Resources, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University College of Food Science and Technology, 235 Nanjing East Road, Nanchang 330047, China
| | - Yue Guo
- State Key Laboratory of Food Science and Resources, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University College of Food Science and Technology, 235 Nanjing East Road, Nanchang 330047, China
| | - Xinxin Chang
- State Key Laboratory of Food Science and Resources, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University College of Food Science and Technology, 235 Nanjing East Road, Nanchang 330047, China
| | - Jianhua Xie
- State Key Laboratory of Food Science and Resources, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University College of Food Science and Technology, 235 Nanjing East Road, Nanchang 330047, China
| | - Yi Chen
- State Key Laboratory of Food Science and Resources, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University College of Food Science and Technology, 235 Nanjing East Road, Nanchang 330047, China
| | - Xiaobo Hu
- State Key Laboratory of Food Science and Resources, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University College of Food Science and Technology, 235 Nanjing East Road, Nanchang 330047, China
| | - Qiang Yu
- State Key Laboratory of Food Science and Resources, China-Canada Joint Laboratory of Food Science and Technology (Nanchang), Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University College of Food Science and Technology, 235 Nanjing East Road, Nanchang 330047, China.
| |
Collapse
|
17
|
Mazzarella L, Santoro F, Ravasio R, Fumagalli V, Massa PE, Rodighiero S, Gavilán E, Romanenghi M, Duso BA, Bonetti E, Manganaro L, Pallavi R, Trastulli D, Pallavicini I, Gentile C, Monzani S, Leonardi T, Pasqualato S, Buttinelli G, Di Martino A, Fedele G, Schiavoni I, Stefanelli P, Meroni G, de Francesco R, Steinkuhler C, Fossati G, Iannacone M, Minucci S, Pelicci PG. Inhibition of the lysine demethylase LSD1 modulates the balance between inflammatory and antiviral responses against coronaviruses. Sci Signal 2023; 16:eade0326. [PMID: 38113337 DOI: 10.1126/scisignal.ade0326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 12/01/2023] [Indexed: 12/21/2023]
Abstract
Innate immune responses to coronavirus infections are highly cell specific. Tissue-resident macrophages, which are infected by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in patients but are inconsistently infected in vitro, exert critical but conflicting effects by secreting both antiviral type I interferons (IFNs) and tissue-damaging inflammatory cytokines. Steroids, the only class of host-targeting drugs approved for the treatment of coronavirus disease 2019 (COVID-19), indiscriminately suppress both responses, possibly impairing viral clearance. Here, we established in vitro cell culture systems that enabled us to separately investigate the cell-intrinsic and cell-extrinsic proinflammatory and antiviral activities of mouse macrophages infected with the prototypical murine coronavirus MHV-A59. We showed that the nuclear factor κB-dependent inflammatory response to viral infection was selectively inhibited by loss of the lysine demethylase LSD1, which was previously implicated in innate immune responses to cancer, with negligible effects on the antiviral IFN response. LSD1 ablation also enhanced an IFN-independent antiviral response, blocking viral egress through the lysosomal pathway. The macrophage-intrinsic antiviral and anti-inflammatory activity of Lsd1 inhibition was confirmed in vitro and in a humanized mouse model of SARS-CoV-2 infection. These results suggest that LSD1 controls innate immune responses against coronaviruses at multiple levels and provide a mechanistic rationale for potentially repurposing LSD1 inhibitors for COVID-19 treatment.
Collapse
Affiliation(s)
- Luca Mazzarella
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Fabio Santoro
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Roberto Ravasio
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Valeria Fumagalli
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
- Vita-Salute San Raffaele University, Milan 20132, Italy
- Experimental Imaging Centre, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Paul E Massa
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Simona Rodighiero
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Elena Gavilán
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Mauro Romanenghi
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Bruno A Duso
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Emanuele Bonetti
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Lara Manganaro
- Virology, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi," 20122 Milan, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Rani Pallavi
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Deborah Trastulli
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Isabella Pallavicini
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Claudia Gentile
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Silvia Monzani
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Tommaso Leonardi
- Center for Genomic Science of IIT@SEMM, Istituto Italiano di Tecnologia (IIT), 20139 Milan, Italy
| | - Sebastiano Pasqualato
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Gabriele Buttinelli
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Angela Di Martino
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Giorgio Fedele
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Ilaria Schiavoni
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Paola Stefanelli
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Giuseppe Meroni
- IFOM-FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Raffaele de Francesco
- Virology, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi," 20122 Milan, Italy
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Christian Steinkuhler
- Preclinical R&D Italfarmaco SpA, Via dei Lavoratori 54, 20092 Cinisello Balsamo (Milan), Italy
| | - Gianluca Fossati
- Preclinical R&D Italfarmaco SpA, Via dei Lavoratori 54, 20092 Cinisello Balsamo (Milan), Italy
| | - Matteo Iannacone
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
- Vita-Salute San Raffaele University, Milan 20132, Italy
- Experimental Imaging Centre, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Saverio Minucci
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
- Department of Biosciences, University of Milan, Milan 20123, Italy
| | - Pier Giuseppe Pelicci
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hematology, University of Milan, Milan 20122, Italy
| |
Collapse
|
18
|
Korwek Z, Czerkies M, Jaruszewicz-Błońska J, Prus W, Kosiuk I, Kochańczyk M, Lipniacki T. Nonself RNA rewires IFN-β signaling: A mathematical model of the innate immune response. Sci Signal 2023; 16:eabq1173. [PMID: 38085817 DOI: 10.1126/scisignal.abq1173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 11/22/2023] [Indexed: 12/18/2023]
Abstract
Type I interferons (IFNs) are key coordinators of the innate immune response to viral infection, which, through activation of the transcriptional regulators STAT1 and STAT2 (STAT1/2) in bystander cells, induce the expression of IFN-stimulated genes (ISGs). Here, we showed that in cells transfected with poly(I:C), an analog of viral RNA, the transcriptional activity of STAT1/2 was terminated because of depletion of the interferon-β (IFN-β) receptor, IFNAR. Activation of RNase L and PKR, products of two ISGs, not only hindered the replenishment of IFNAR but also suppressed negative regulators of IRF3 and NF-κB, consequently promoting IFNB transcription. We incorporated these findings into a mathematical model of innate immunity. By coupling signaling through the IRF3-NF-κB and STAT1/2 pathways with the activities of RNase L and PKR, the model explains how poly(I:C) switches the transcriptional program from being STAT1/2 induced to being IRF3 and NF-κB induced, which converts IFN-β-responding cells to IFN-β-secreting cells.
Collapse
Affiliation(s)
- Zbigniew Korwek
- Department of Biosystems and Soft Matter, Institute of Fundamental Technological Research of the Polish Academy of Sciences, Warsaw 02-106, Poland
| | - Maciej Czerkies
- Department of Biosystems and Soft Matter, Institute of Fundamental Technological Research of the Polish Academy of Sciences, Warsaw 02-106, Poland
| | - Joanna Jaruszewicz-Błońska
- Department of Biosystems and Soft Matter, Institute of Fundamental Technological Research of the Polish Academy of Sciences, Warsaw 02-106, Poland
| | - Wiktor Prus
- Department of Biosystems and Soft Matter, Institute of Fundamental Technological Research of the Polish Academy of Sciences, Warsaw 02-106, Poland
| | - Ilona Kosiuk
- Department of Biosystems and Soft Matter, Institute of Fundamental Technological Research of the Polish Academy of Sciences, Warsaw 02-106, Poland
| | - Marek Kochańczyk
- Department of Biosystems and Soft Matter, Institute of Fundamental Technological Research of the Polish Academy of Sciences, Warsaw 02-106, Poland
| | - Tomasz Lipniacki
- Department of Biosystems and Soft Matter, Institute of Fundamental Technological Research of the Polish Academy of Sciences, Warsaw 02-106, Poland
| |
Collapse
|
19
|
Dai L, Chen P, Hou D, Wang Y, Zhou Y, Sun C. Pathogenicity and transcriptomic exploration of Vibrio fortis in Penaeus monodon. FISH & SHELLFISH IMMUNOLOGY 2023; 142:109097. [PMID: 37751788 DOI: 10.1016/j.fsi.2023.109097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 09/28/2023]
Abstract
In this study, a strain (recorded as Y6) was isolated from the biofloc pool, its DNA was extracted for 16S rDNA sequencing and compared in the NCBI database, and it was identified as Vibrio fortis. The V. fortis was activated, cultured, and artificially injected into Penaeus monodon to observe the symptoms and calculate the semi-lethal concentration (LC50). It was found that the symptoms of the red leg, an empty stomach, and enlarged hepatopancreas of P. monodon after infection with V. fortis. The LC50 was 4.00 × 107, 2.24 × 107, 1.82 × 107, 1.41 × 107, 7.52 × 106 and 3.31 × 106 CFU/mL at 16, 24, 32, 48, 128, and 144 hpi, respectively. The K-B disk method was used to detect the sensitivity of V. fortis to various antibiotic drugs. V. fortis resisted Ampicillin, Piperacillin, Cefazolin, Cephalothin and Cefoxitin. Highly sensitive to Polymyxin B, Tobramycin, Gentamicin, Cefepime, Cefoperazone and Streptomycin. To explore the molecular response mechanism of V. fortis infection in P. monodon, the hepatopancreas of P. monodon infected with V. fortis at 24 and 48 hpi by transcriptome sequencing, and a total of 347 DEGs were obtained (214 up-regulated DEGs and 133 down-regulated DEGs). In the KEGG pathway enrichment analysis of DEGs, significant changes were found in genes and signaling pathways related to immune system and substance metabolism, including NOD-like receptor signaling pathways, Toll and Imd signaling pathways, C-type lectin receptor signaling pathways and pyruvate metabolism. This study initially revealed the immune response of P. monodon to V. fortis infection from the molecular level and provided a reference for further understanding of the study and control of the vibriosis of shrimp.
Collapse
Affiliation(s)
- Linxin Dai
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Peixun Chen
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Danqing Hou
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Yue Wang
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China
| | - Yinhuan Zhou
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China; Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang, Guangdong, China.
| | - Chengbo Sun
- College of Fisheries, Guangdong Ocean University, Zhanjiang, Guangdong, China; Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals, Zhanjiang, Guangdong, China.
| |
Collapse
|
20
|
Ricciardi-Jorge T, da Rocha EL, Gonzalez-Kozlova E, Rodrigues-Luiz GF, Ferguson BJ, Sweeney T, Irigoyen N, Mansur DS. PKR-mediated stress response enhances dengue and Zika virus replication. mBio 2023; 14:e0093423. [PMID: 37732809 PMCID: PMC10653888 DOI: 10.1128/mbio.00934-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/01/2023] [Indexed: 09/22/2023] Open
Abstract
IMPORTANCE One of the fundamental features that make viruses intracellular parasites is the necessity to use cellular translational machinery. Hence, this is a crucial checkpoint for controlling infections. Here, we show that dengue and Zika viruses, responsible for nearly 400 million infections every year worldwide, explore such control for optimal replication. Using immunocompetent cells, we demonstrate that arrest of protein translations happens after sensing of dsRNA and that the information required to avoid this blocking is contained in viral 5'-UTR. Our work, therefore, suggests that the non-canonical translation described for these viruses is engaged when the intracellular stress response is activated.
Collapse
Affiliation(s)
- Taissa Ricciardi-Jorge
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de Santa Catarina (UFSC), Florianópolis, Brazil
- The Pirbright Institute, Woking, United Kingdom
| | - Edroaldo Lummertz da Rocha
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de Santa Catarina (UFSC), Florianópolis, Brazil
| | - Edgar Gonzalez-Kozlova
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de Santa Catarina (UFSC), Florianópolis, Brazil
- Icahn School of Medicine, New York, USA
| | - Gabriela Flavia Rodrigues-Luiz
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de Santa Catarina (UFSC), Florianópolis, Brazil
| | - Brian J. Ferguson
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | | | - Nerea Irigoyen
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Daniel Santos Mansur
- Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de Santa Catarina (UFSC), Florianópolis, Brazil
| |
Collapse
|
21
|
Xu C, Liu X, Yu L, Fang X, Yao L, Lau H, Vyas P, Pryke L, Xu B, Tang L, Jiang J, Chen X. CD147 monoclonal antibody attenuates abdominal aortic aneurysm formation in angiotensin II-Infused apoE -/- mice. Int Immunopharmacol 2023; 122:110526. [PMID: 37393837 DOI: 10.1016/j.intimp.2023.110526] [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: 04/16/2023] [Revised: 06/03/2023] [Accepted: 06/14/2023] [Indexed: 07/04/2023]
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is a life threatening vascular disease. Our previous study reported the upregulation of CD147 expression in human aortic aneurysms. OBJECTIVE In this study, we injected apoE-/- mice intraperitoneally with CD147 monoclonal antibody or IgG control antibody to observe its effect on Angiotensin II (AngII) induced AAA formation. METHODS ApoE-/- mice were randomly divided into an AngⅡ+CD147 antibody group (n = 20) and an AngⅡ+IgG antibody group (n = 20). The Alzet osmotic minipump was implanted subcutaneously into the backs of mice to infuse AngII (1000 ng/kg/min) for 28 days and subsequently treated with CD147 monoclonal antibody or control IgG mAb (10 μg/mouse/day) beginning one day after surgery. Body weight, food intake, drinking volume and blood pressure were measured weekly throughout the study. After 4 weeks of injection, routine bloodwork measuring liver function, kidney function and lipid levels were recorded. Hematoxylin and eosin (H&E), Masson's trichrome, and Elastic van Gieson (EVG) staining were used to evaluate the pathological changes in blood vessels. In addition, Immunohistochemical assay was used to detect infiltration of inflammatory cells. Tandem mass tag (TMT)-based proteomic analysis was used to define differentially expressed proteins (DEPs) using a p-value < 0.05 and fold change > 1.2 or < 0.83 as the threshold. Subsequently, we conducted protein-protein interaction (PPI) network and GO enrichment analysis to determine the core biological function altered after CD147 antibody injection. RESULTS The CD147 monoclonal antibody suppresses Ang II-induced AAA formation in apoE-/- mice and reduced aortic expansion, elastic lamina degradation, and inflammatory cells accumulation. Bioinformatics analysis showed that Ptk6, Itch, Casp3, and Oas1a were the hub DEPs. These DEPs in the two group were mainly involved in collagen fibril organization, extracellular matrix organization, and muscle contraction. These data robustly demonstrated that CD147 monoclonal antibody suppresses Ang II-induced AAA formation through reduction of inflammatory response and regulation of the above defined hub proteins and biological processes. Thus, the CD147 monoclonal antibody might be a promising target in the treatment of abdominal aortic aneurysm.
Collapse
Affiliation(s)
- Cheng Xu
- Department of Cardiology, Taizhou Hospital, Wenzhou Medical University, Taizhou 317000, PR China
| | - Xiaowei Liu
- Department of Cardiology, Zhejiang Hospital, Hangzhou 310013, PR China
| | - Lei Yu
- Department of Cardiology, Taizhou Hospital, Wenzhou Medical University, Taizhou 317000, PR China
| | - Xiaoxin Fang
- Department of Cardiology, Taizhou Hospital, Wenzhou Medical University, Taizhou 317000, PR China
| | - Lei Yao
- Department of Cardiology, Taizhou Hospital, Wenzhou Medical University, Taizhou 317000, PR China
| | - HuiChong Lau
- Department of Medicine, Crozer-Chester Medical Center, Upland, PA 19013, USA
| | - Punit Vyas
- Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Luke Pryke
- Internal medicine, Indiana University, Indianapolis, IN 46202
| | - Baohui Xu
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Lijiang Tang
- Department of Cardiology, Zhejiang Hospital, Hangzhou 310013, PR China
| | - Jianjun Jiang
- Department of Cardiology, Taizhou Hospital, Wenzhou Medical University, Taizhou 317000, PR China
| | - Xiaofeng Chen
- Department of Cardiology, Taizhou Hospital, Wenzhou Medical University, Taizhou 317000, PR China; Department of Radiation Oncology, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
22
|
Grabowski F, Kochańczyk M, Korwek Z, Czerkies M, Prus W, Lipniacki T. Antagonism between viral infection and innate immunity at the single-cell level. PLoS Pathog 2023; 19:e1011597. [PMID: 37669278 PMCID: PMC10503725 DOI: 10.1371/journal.ppat.1011597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 09/15/2023] [Accepted: 08/02/2023] [Indexed: 09/07/2023] Open
Abstract
When infected with a virus, cells may secrete interferons (IFNs) that prompt nearby cells to prepare for upcoming infection. Reciprocally, viral proteins often interfere with IFN synthesis and IFN-induced signaling. We modeled the crosstalk between the propagating virus and the innate immune response using an agent-based stochastic approach. By analyzing immunofluorescence microscopy images we observed that the mutual antagonism between the respiratory syncytial virus (RSV) and infected A549 cells leads to dichotomous responses at the single-cell level and complex spatial patterns of cell signaling states. Our analysis indicates that RSV blocks innate responses at three levels: by inhibition of IRF3 activation, inhibition of IFN synthesis, and inhibition of STAT1/2 activation. In turn, proteins coded by IFN-stimulated (STAT1/2-activated) genes inhibit the synthesis of viral RNA and viral proteins. The striking consequence of these inhibitions is a lack of coincidence of viral proteins and IFN expression within single cells. The model enables investigation of the impact of immunostimulatory defective viral particles and signaling network perturbations that could potentially facilitate containment or clearance of the viral infection.
Collapse
Affiliation(s)
- Frederic Grabowski
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw, Poland
| | - Marek Kochańczyk
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw, Poland
| | - Zbigniew Korwek
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw, Poland
| | - Maciej Czerkies
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw, Poland
| | - Wiktor Prus
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw, Poland
| | - Tomasz Lipniacki
- Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw, Poland
- Department of Statistics, Rice University, Houston, Texas, United States of America
| |
Collapse
|
23
|
Moran J, Feltham L, Bagnall J, Goldrick M, Lord E, Nettleton C, Spiller DG, Roberts I, Paszek P. Live-cell imaging reveals single-cell and population-level infection strategies of Listeria monocytogenes in macrophages. Front Immunol 2023; 14:1235675. [PMID: 37675103 PMCID: PMC10478088 DOI: 10.3389/fimmu.2023.1235675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 08/01/2023] [Indexed: 09/08/2023] Open
Abstract
Pathogens have developed intricate strategies to overcome the host's innate immune responses. In this paper we use live-cell microscopy with a single bacterium resolution to follow in real time interactions between the food-borne pathogen L. monocytogenes and host macrophages, a key event controlling the infection in vivo. We demonstrate that infection results in heterogeneous outcomes, with only a subset of bacteria able to establish a replicative invasion of macrophages. The fate of individual bacteria in the same host cell was independent from the host cell and non-cooperative, being independent from co-infecting bacteria. A higher multiplicity of infection resulted in a reduced probability of replication of the overall bacterial population. By use of internalisation assays and conditional probabilities to mathematically describe the two-stage invasion process, we demonstrate that the higher MOI compromises the ability of macrophages to phagocytose bacteria. We found that the rate of phagocytosis is mediated via the secreted Listeriolysin toxin (LLO), while the probability of replication of intracellular bacteria remained constant. Using strains expressing fluorescent reporters to follow transcription of either the LLO-encoding hly or actA genes, we show that replicative bacteria exhibited higher PrfA regulon expression in comparison to those bacteria that did not replicate, however elevated PrfA expression per se was not sufficient to increase the probability of replication. Overall, this demonstrates a new role for the population-level, but not single cell, PrfA-mediated activity to regulate outcomes of host pathogen interactions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ian Roberts
- School of Biology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Pawel Paszek
- School of Biology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| |
Collapse
|
24
|
Qiao D, Xu X, Zhang Y, Yang J, Brasier AR. RSV replication modifies the XBP1s binding complex on the IRF1 upstream enhancer to potentiate the mucosal anti-viral response. Front Immunol 2023; 14:1197356. [PMID: 37564646 PMCID: PMC10411192 DOI: 10.3389/fimmu.2023.1197356] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 07/06/2023] [Indexed: 08/12/2023] Open
Abstract
Introduction The unfolded protein response (UPR) has emerged as an important signaling pathway mediating anti-viral defenses to Respiratory Syncytial Virus (RSV) infection. Earlier we found that RSV replication predominantly activates the evolutionarily conserved Inositol Requiring Enzyme 1α (IRE1α)-X-Box Binding Protein 1 spliced (XBP1s) arm of the Unfolded Protein Response (UPR) producing inflammation, metabolic adaptation and cellular plasticity, yet the mechanisms how the UPR potentiates inflammation are not well understood. Methods To understand this process better, we examined the genomic response integrating RNA-seq and Cleavage Under Targets and Release Using Nuclease (CUT&RUN) analyses. These data were integrated with an RNA-seq analysis conducted on RSV-infected small airway cells ± an IRE1α RNAse inhibitor. Results We identified RSV induced expression changes in ~3.2K genes; of these, 279 required IRE1α and were enriched in IL-10/cytokine signaling pathways. From this data set, we identify those genes directly under XBP1s control by CUT&RUN. Although XBP1s binds to ~4.2 K high-confidence genomic binding sites, surprisingly only a small subset of IL10/cytokine signaling genes are directly bound. We further apply CUT&RUN to find that RSV infection enhances XBP1s loading on 786 genomic sites enriched in AP1/Fra-1, RELA and SP1 binding sites. These control a subset of cytokine regulatory factor genes including IFN response factor 1 (IRF1), CSF2, NFKB1A and DUSP10. Focusing on the downstream role of IRF1, selective knockdown (KD) and overexpression experiments demonstrate IRF1 induction controls type I and -III interferon (IFN) and IFN-stimulated gene (ISG) expression, demonstrating that ISG are indirectly regulated by XBP1 through IRF1 transactivation. Examining the mechanism of IRF1 activation, we observe that XBP1s directly binds a 5' enhancer sequence whose XBP1s loading is increased by RSV. The functional requirement for the enhancer is demonstrated by targeting a dCas9-KRAB silencer, reducing IRF1 activation. Chromatin immunoprecipitation shows that XBP1 is required, but not sufficient, for RSV-induced recruitment of activated phospho-Ser2 Pol II to the enhancer. Discussion We conclude that XBP1s is a direct activator of a core subset of IFN and cytokine regulatory genes in response to RSV. Of these IRF1 is upstream of the type III IFN and ISG response. We find that RSV modulates the XBP1s binding complex on the IRF1 5' enhancer whose activation is required for IRF1 expression. These findings provide novel insight into how the IRE1α-XBP1s pathway potentiates airway mucosal anti-viral responses.
Collapse
Affiliation(s)
- Dianhua Qiao
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health (SMPH), Madison, WI, United States
| | - Xiaofang Xu
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health (SMPH), Madison, WI, United States
| | - Yueqing Zhang
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Jun Yang
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, United States
| | - Allan R. Brasier
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health (SMPH), Madison, WI, United States
- Institute for Clinical and Translational Research (ICTR), University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
25
|
Jaruszewicz-Błońska J, Kosiuk I, Prus W, Lipniacki T. A plausible identifiable model of the canonical NF-κB signaling pathway. PLoS One 2023; 18:e0286416. [PMID: 37267242 DOI: 10.1371/journal.pone.0286416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/15/2023] [Indexed: 06/04/2023] Open
Abstract
An overwhelming majority of mathematical models of regulatory pathways, including the intensively studied NF-κB pathway, remains non-identifiable, meaning that their parameters may not be determined by existing data. The existing NF-κB models that are capable of reproducing experimental data contain non-identifiable parameters, whereas simplified models with a smaller number of parameters exhibit dynamics that differs from that observed in experiments. Here, we reduced an existing model of the canonical NF-κB pathway by decreasing the number of equations from 15 to 6. The reduced model retains two negative feedback loops mediated by IκBα and A20, and in response to both tonic and pulsatile TNF stimulation exhibits dynamics that closely follow that of the original model. We carried out the sensitivity-based linear analysis and Monte Carlo-based analysis to demonstrate that the resulting model is both structurally and practically identifiable given measurements of 5 model variables from a simple TNF stimulation protocol. The reduced model is capable of reproducing different types of responses that are characteristic to regulatory motifs controlled by negative feedback loops: nearly-perfect adaptation as well as damped and sustained oscillations. It can serve as a building block of more comprehensive models of the immune response and cancer, where NF-κB plays a decisive role. Our approach, although may not be automatically generalized, suggests that models of other regulatory pathways can be transformed to identifiable, while retaining their dynamical features.
Collapse
Affiliation(s)
| | - Ilona Kosiuk
- Institute of Fundamental Technological Research of the Polish Academy of Sciences, Warsaw, Poland
| | - Wiktor Prus
- Institute of Fundamental Technological Research of the Polish Academy of Sciences, Warsaw, Poland
| | - Tomasz Lipniacki
- Institute of Fundamental Technological Research of the Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
26
|
Groten SA, Smit ER, Janssen EFJ, van den Eshof BL, van Alphen FPJ, van der Zwaan C, Meijer AB, Hoogendijk AJ, Biggelaar MVD. Multi-omics delineation of cytokine-induced endothelial inflammatory states. Commun Biol 2023; 6:525. [PMID: 37188730 PMCID: PMC10184633 DOI: 10.1038/s42003-023-04897-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 05/02/2023] [Indexed: 05/17/2023] Open
Abstract
Vascular endothelial cells (ECs) form a dynamic interface between blood and tissue and play a crucial role in the progression of vascular inflammation. Here, we aim to dissect the system-wide molecular mechanisms of inflammatory endothelial-cytokine responses. Applying an unbiased cytokine library, we determined that TNFα and IFNγ induced the largest EC response resulting in distinct proteomic inflammatory signatures. Notably, combined TNFα + IFNγ stimulation induced an additional synergetic inflammatory signature. We employed a multi-omics approach to dissect these inflammatory states, combining (phospho-) proteome, transcriptome and secretome and found, depending on the stimulus, a wide-array of altered immune-modulating processes, including complement proteins, MHC complexes and distinct secretory cytokines. Synergy resulted in cooperative activation of transcript induction. This resource describes the intricate molecular mechanisms that are at the basis of endothelial inflammation and supports the adaptive immunomodulatory role of the endothelium in host defense and vascular inflammation.
Collapse
Affiliation(s)
- Stijn A Groten
- Department of Molecular Hematology, Sanquin Research, Amsterdam, 1066 CX, The Netherlands
| | - Eva R Smit
- Department of Molecular Hematology, Sanquin Research, Amsterdam, 1066 CX, The Netherlands
| | - Esmée F J Janssen
- Department of Molecular Hematology, Sanquin Research, Amsterdam, 1066 CX, The Netherlands
| | - Bart L van den Eshof
- Department of Molecular Hematology, Sanquin Research, Amsterdam, 1066 CX, The Netherlands
| | - Floris P J van Alphen
- Department of Molecular Hematology, Sanquin Research, Amsterdam, 1066 CX, The Netherlands
| | - Carmen van der Zwaan
- Department of Molecular Hematology, Sanquin Research, Amsterdam, 1066 CX, The Netherlands
| | - Alexander B Meijer
- Department of Molecular Hematology, Sanquin Research, Amsterdam, 1066 CX, The Netherlands
- Department of Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, 3584 CS, The Netherlands
| | - Arie J Hoogendijk
- Department of Molecular Hematology, Sanquin Research, Amsterdam, 1066 CX, The Netherlands
| | | |
Collapse
|
27
|
Qing F, Liu Z. Interferon regulatory factor 7 in inflammation, cancer and infection. Front Immunol 2023; 14:1190841. [PMID: 37251373 PMCID: PMC10213216 DOI: 10.3389/fimmu.2023.1190841] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 04/26/2023] [Indexed: 05/31/2023] Open
Abstract
Interferon regulatory factor 7 (IRF7), a member of the interferon regulatory factors (IRFs) family, is located downstream of the pattern recognition receptors (PRRs)-mediated signaling pathway and is essential for the production of type I interferon (IFN-I). Activation of IRF7 inhibits various viral and bacterial infections and suppresses the growth and metastasis of some cancers, but it may also affect the tumor microenvironment and promote the development of other cancers. Here, we summarize recent advances in the role of IRF7 as a multifunctional transcription factor in inflammation, cancer and infection by regulating IFN-I production or IFN-I-independent signaling pathways.
Collapse
|
28
|
Altea-Manzano P, Doglioni G, Liu Y, Cuadros AM, Nolan E, Fernández-García J, Wu Q, Planque M, Laue KJ, Cidre-Aranaz F, Liu XZ, Marin-Bejar O, Van Elsen J, Vermeire I, Broekaert D, Demeyer S, Spotbeen X, Idkowiak J, Montagne A, Demicco M, Alkan HF, Rabas N, Riera-Domingo C, Richard F, Geukens T, De Schepper M, Leduc S, Hatse S, Lambrechts Y, Kay EJ, Lilla S, Alekseenko A, Geldhof V, Boeckx B, de la Calle Arregui C, Floris G, Swinnen JV, Marine JC, Lambrechts D, Pelechano V, Mazzone M, Zanivan S, Cools J, Wildiers H, Baud V, Grünewald TGP, Ben-David U, Desmedt C, Malanchi I, Fendt SM. A palmitate-rich metastatic niche enables metastasis growth via p65 acetylation resulting in pro-metastatic NF-κB signaling. NATURE CANCER 2023; 4:344-364. [PMID: 36732635 PMCID: PMC7615234 DOI: 10.1038/s43018-023-00513-2] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 01/03/2023] [Indexed: 02/04/2023]
Abstract
Metabolic rewiring is often considered an adaptive pressure limiting metastasis formation; however, some nutrients available at distant organs may inherently promote metastatic growth. We find that the lung and liver are lipid-rich environments. Moreover, we observe that pre-metastatic niche formation increases palmitate availability only in the lung, whereas a high-fat diet increases it in both organs. In line with this, targeting palmitate processing inhibits breast cancer-derived lung metastasis formation. Mechanistically, breast cancer cells use palmitate to synthesize acetyl-CoA in a carnitine palmitoyltransferase 1a-dependent manner. Concomitantly, lysine acetyltransferase 2a expression is promoted by palmitate, linking the available acetyl-CoA to the acetylation of the nuclear factor-kappaB subunit p65. Deletion of lysine acetyltransferase 2a or carnitine palmitoyltransferase 1a reduces metastasis formation in lean and high-fat diet mice, and lung and liver metastases from patients with breast cancer show coexpression of both proteins. In conclusion, palmitate-rich environments foster metastases growth by increasing p65 acetylation, resulting in a pro-metastatic nuclear factor-kappaB signaling.
Collapse
Affiliation(s)
- Patricia Altea-Manzano
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Ginevra Doglioni
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Yawen Liu
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Alejandro M Cuadros
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | | | - Juan Fernández-García
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Qi Wu
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Mélanie Planque
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Kathrin Julia Laue
- Department of Human Molecular Genetics & Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Florencia Cidre-Aranaz
- Hopp-Children's Cancer Center (KiTZ), Heidelberg, Germany
- Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Xiao-Zheng Liu
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Oskar Marin-Bejar
- Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| | - Joke Van Elsen
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Ines Vermeire
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Dorien Broekaert
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Sofie Demeyer
- Laboratory for Molecular Biology of Leukemia, VIB-KU Leuven, Leuven, Belgium
| | - Xander Spotbeen
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Jakub Idkowiak
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven, Leuven, Belgium
- Department of Analytical Chemistry, Faculty of Chemical Technology, University of Pardubice, Pardubice, Czech Republic
| | - Aurélie Montagne
- Université Paris Cité, NF-kappaB, Différenciation et Cancer, Paris, France
| | - Margherita Demicco
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - H Furkan Alkan
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | | | - Carla Riera-Domingo
- Laboratory of Tumor Inflammation and Angiogenesis, VIB Center for Cancer Biology, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - François Richard
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Tatjana Geukens
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Maxim De Schepper
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Sophia Leduc
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Sigrid Hatse
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Yentl Lambrechts
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, Leuven, Belgium
| | | | - Sergio Lilla
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Alisa Alekseenko
- SciLifeLab, Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Solna, Sweden
| | - Vincent Geldhof
- Laboratory for Angiogenesis and Vascular Metabolism, VIB-KU Leuven, Leuven, Belgium
| | - Bram Boeckx
- Laboratory of Translational Genetics, VIB Center for Cancer Biology, Leuven, Belgium
- Laboratory of Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Celia de la Calle Arregui
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Giuseppe Floris
- Department of Imaging and Pathology, Laboratory of Translational Cell & Tissue Research, KU Leuven, Leuven, Belgium
- Department of Pathology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Johannes V Swinnen
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Jean-Christophe Marine
- Laboratory for Molecular Cancer Biology, VIB Center for Cancer Biology, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| | - Diether Lambrechts
- Laboratory of Translational Genetics, VIB Center for Cancer Biology, Leuven, Belgium
- Laboratory of Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Vicent Pelechano
- SciLifeLab, Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Solna, Sweden
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, VIB Center for Cancer Biology, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Sara Zanivan
- Cancer Research UK Beatson Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Jan Cools
- Laboratory for Molecular Biology of Leukemia, VIB-KU Leuven, Leuven, Belgium
| | - Hans Wildiers
- Laboratory of Experimental Oncology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Véronique Baud
- Université Paris Cité, NF-kappaB, Différenciation et Cancer, Paris, France
| | - Thomas G P Grünewald
- Hopp-Children's Cancer Center (KiTZ), Heidelberg, Germany
- Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Uri Ben-David
- Department of Human Molecular Genetics & Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Christine Desmedt
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | | | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium.
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium.
| |
Collapse
|
29
|
Zhang L, Xia Q, Li W, Liu Q, Zhang L, Tian X, Ye L, Wang G, Peng Q. Immunoproteasome subunit β5i promotes perifascicular muscle atrophy in dermatomyositis by upregulating RIG-I. RMD Open 2023; 9:rmdopen-2022-002818. [PMID: 36854567 PMCID: PMC9980316 DOI: 10.1136/rmdopen-2022-002818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 01/04/2023] [Indexed: 03/02/2023] Open
Abstract
BACKGROUND Perifascicular atrophy is a unique pathological hallmark in dermatomyositis (DM)-affected muscles; however, the mechanism underlying this process remains unclear. In this study, we aimed to investigate the potential role of the immunoproteasome subunit β5i and retinoic acid-inducible gene-I (RIG-I) in DM-associated muscle atrophy. METHODS The expression of β5i and RIG-I in the muscles of 16 patients with DM was examined by PCR, western blotting and immunohistochemistry. The associations between β5i and RIG-I expression levels and muscle disease severity were evaluated. Lentivirus transduction was used to overexpress β5i in human skeletal muscle myoblasts (HSMMs) and consequent cell functional changes were studied in vitro. RESULTS β5i and RIG-I expression in the muscle of patients with DM was significantly increased and closely associated with muscle disease severity. Immunohistochemistry and immunofluorescence analyses showed the marked colocalised expression of β5i and RIG-I in perifascicular myofibres. β5i overexpression in HSMMs significantly upregulated RIG-I, the muscle atrophy marker MuRF1, type I IFN-related proteins (MxA and IFNβ) and NF-κB pathway-related proteins (pIκBα, pIRF3 and pNF-κBp65). In addition, the viability of HSMMs decreased significantly after β5i overexpression and was partly recovered by treatment with a β5i inhibitor (PR957). Moreover, activation of RIG-I by pppRNA upregulated IFNβ and MuRF1 and reduced the cell viability of HSMMs. CONCLUSION The immunoproteasome subunit β5i promotes perifascicular muscle atrophy in DM via RIG-I upregulation; our findings suggest a pathomechanistic role of β5i and RIG-I in DM-associated muscle damage, highlighting these components as potential therapeutic targets for the treatment of DM.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Rheumatology, Key Myositis Laboratories, China-Japan Friendship Hospital, Beijing, China
| | - Qisheng Xia
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Wenli Li
- Department of Rheumatology, Key Myositis Laboratories, China-Japan Friendship Hospital, Beijing, China
| | - Qingyan Liu
- Department of Rheumatology, Key Myositis Laboratories, China-Japan Friendship Hospital, Beijing, China
| | - Lining Zhang
- Department of Rheumatology, Key Myositis Laboratories, China-Japan Friendship Hospital, Beijing, China
| | - Xiaolan Tian
- Department of Rheumatology, Key Myositis Laboratories, China-Japan Friendship Hospital, Beijing, China
| | - Lifang Ye
- Department of Rheumatology, Key Myositis Laboratories, China-Japan Friendship Hospital, Beijing, China
| | - Guochun Wang
- Department of Rheumatology, Key Myositis Laboratories, China-Japan Friendship Hospital, Beijing, China
| | - Qinglin Peng
- Department of Rheumatology, Key Myositis Laboratories, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
30
|
Bao J, He Y, Yang C, Lu N, Li A, Gao S, Hosyanto FF, Tang J, Si J, Tang X, Fu H, Xu L. Inhibition of mycobacteria proliferation in macrophages by low cisplatin concentration through phosphorylated p53-related apoptosis pathway. PLoS One 2023; 18:e0281170. [PMID: 36719870 PMCID: PMC9888694 DOI: 10.1371/journal.pone.0281170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 01/16/2023] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Drug resistance is a prominent problem in the treatment of tuberculosis, so it is urgent to develop new anti- tuberculosis drugs. Here, we investigated the effects and mechanisms of cisplatin (DDP) on intracellular Mycobacterium smegmatis to tap the therapeutic potential of DDP in mycobacterial infection. RESULTS Macrophages infected with Mycobacterium smegmatis were treated with DDP alone or combined with isoniazid or rifampicin. The results showed that the bacterial count in macrophages decreased significantly after DDP (≤ 6 μg/mL) treatment. When isoniazid or rifampicin was combined with DDP, the number of intracellular mycobacteria was also significantly lower than that of isoniazid or rifampicin alone. Apoptosis of infected cells increased after 24 h of DDP treatment, as shown by flow cytometry and transmission electron microscopy detection. Transcriptome sequencing showed that there were 1161 upregulated and 645 downregulated differentially expressed genes (DEGs) between the control group and DDP treatment group. A Trp53-centered protein interaction network was found based on the top 100 significant DEGs through STRING and Cytoscape software. The expression of phosphorylated p53, Bax, JAK, p38 MAPK and PI3K increased after DDP treatment, as shown by Western blot analysis. Inhibitors of JAK, PI3K or p38 MAPK inhibited the increase in cell apoptosis and the reduction in the intracellular bacterial count induced by DDP. The p53 promoter Kevetrin hydrochloride scavenges intracellular mycobacteria. If combined with DDP, Kevetrin hydrochloride could increase the effect of DDP on the elimination of intracellular mycobacteria. In conclusion, DDP at low concentrations could activate the JAK, p38 MAPK and PI3K pathways in infected macrophages, promote the phosphorylation of p53 protein, and increase the ratio of Bax to Bcl-2, leading to cell apoptosis, thus eliminating intracellular bacteria and reducing the spread of mycobacteria. CONCLUSION DDP may be a new host-directed therapy for tuberculosis treatment, as well as the p53 promoter Kevetrin hydrochloride.
Collapse
Affiliation(s)
- Jiajia Bao
- Department of Pathogenic Biology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
- Hospital-Acquired Infection Control Department, First People’s Hospital of Jintang County, Chengdu, China
| | - Yonglin He
- Department of Pathogenic Biology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Chun Yang
- Department of Pathogenic Biology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Nan Lu
- Department of Pathogenic Biology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Anlong Li
- Department of Pathogenic Biology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Sijia Gao
- Department of Pathogenic Biology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | | | - Jialing Tang
- Department of Pathogenic Biology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Junzhuo Si
- Department of Pathogenic Biology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Xia Tang
- Clinical laboratory, People’s Hospital of Rongchang District, Chongqing, China
| | - Huichao Fu
- Department of Pathogenic Biology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Lei Xu
- Department of Pathogenic Biology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
- * E-mail:
| |
Collapse
|
31
|
Brasier AR. Innate Immunity, Epithelial Plasticity, and Remodeling in Asthma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1426:265-285. [PMID: 37464126 DOI: 10.1007/978-3-031-32259-4_13] [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: 07/20/2023]
Abstract
Innate immune responses (IIR) of the epithelium play a critical role in the initiation and progression of asthma. The core of the IIR is an intracellular signaling pathway activated by pattern recognition receptors (PRRs) to limit the spread of infectious organisms. This chapter will focus on the epithelium as the major innate sentinel cell and its role in acute exacerbations (AEs). Although the pathways of how the IIR activates the NFκB transcription factor, triggering cytokine secretion, dendritic cell activation, and Th2 polarization are well-described, recent exciting work has developed mechanistic insights into how chronic activation of the IIR is linked to mucosal adaptive responses. These adaptations include changes in cell state, now called epithelial-mesenchymal plasticity (EMP). EMP is a coordinated, genomic response to airway injury disrupting epithelial barrier function, expanding the basal lamina, and producing airway remodeling. EMP is driven by activation of the unfolded protein response (UPR), a transcriptional response producing metabolic shunting of glucose through the hexosamine biosynthetic pathway (HBP) to protein N-glycosylation. NFκB signaling and UPR activation pathways potentiate each other in remodeling the basement membrane. Understanding of injury-repair process of epithelium provides new therapeutic targets for precision approaches to the treatment of asthma exacerbations and their sequelae.
Collapse
Affiliation(s)
- Allan R Brasier
- Department of Medicine and Institute for Clinical and Translational Research (ICTR), School of Medicine and Public Health (SMPH), University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
32
|
Respiratory Syncytial Virus Protects Bystander Cells against Influenza A Virus Infection by Triggering Secretion of Type I and Type III Interferons. J Virol 2022; 96:e0134122. [DOI: 10.1128/jvi.01341-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Influenza A virus (IAV) and respiratory syncytial virus (RSV) are common recurrent respiratory infectants that show a relatively high coincidence. We demonstrated that preinfection with RSV partitions the cell population into a subpopulation susceptible to subsequent infection with IAV and an IAV-proof subpopulation.
Collapse
|
33
|
A balance between vector survival and virus transmission is achieved through JAK/STAT signaling inhibition by a plant virus. Proc Natl Acad Sci U S A 2022; 119:e2122099119. [PMID: 36191206 PMCID: PMC9564230 DOI: 10.1073/pnas.2122099119] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Viruses pose a great threat to animal and plant health worldwide, with many being dependent on insect vectors for transmission between hosts. While the virus-host arms race has been well established, how viruses and insect vectors adapt to each other remains poorly understood. Begomoviruses comprise the largest genus of plant-infecting DNA viruses and are exclusively transmitted by the whitefly Bemisia tabaci. Here, we show that the vector Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway plays an important role in mediating the adaptation between the begomovirus tomato yellow leaf curl virus (TYLCV) and whiteflies. We found that the JAK/STAT pathway in B. tabaci functions as an antiviral mechanism against TYLCV infection in whiteflies as evidenced by the increase in viral DNA and coat protein (CP) levels after inhibiting JAK/STAT signaling. Two STAT-activated effector genes, BtCD109-2 and BtCD109-3, mediate this anti-TYLCV activity. To counteract this vector immunity, TYLCV has evolved strategies that impair the whitefly JAK/STAT pathway. Infection of TYLCV is associated with a reduction of JAK/STAT pathway activity in whiteflies. Moreover, TYLCV CP binds to STAT and blocks its nuclear translocation, thus, abrogating the STAT-dependent transactivation of target genes. We further show that inhibition of the whitefly JAK/STAT pathway facilitates TYLCV transmission but reduces whitefly survival and fecundity, indicating that this JAK/STAT-dependent TYLCV-whitefly interaction plays an important role in keeping a balance between whitefly fitness and TYLCV transmission. This study reveals a mechanism of plant virus-insect vector coadaptation in relation to vector survival and virus transmission.
Collapse
|
34
|
Wei M, Yan X, Xin X, Chen H, Hou L, Zhang J. Hepatocyte-Specific Smad4 Deficiency Alleviates Liver Fibrosis via the p38/p65 Pathway. Int J Mol Sci 2022; 23:ijms231911696. [PMID: 36232998 PMCID: PMC9570188 DOI: 10.3390/ijms231911696] [Citation(s) in RCA: 3] [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: 07/16/2022] [Revised: 09/17/2022] [Accepted: 09/29/2022] [Indexed: 11/20/2022] Open
Abstract
Liver fibrosis is a wound-healing response caused by the abnormal accumulation of extracellular matrix, which is produced by activated hepatic stellate cells (HSCs). Most studies have focused on the activated HSCs themselves in liver fibrosis, and whether hepatocytes can modulate the process of fibrosis is still unclear. Sma mothers against decapentaplegic homologue 4 (Smad4) is a key intracellular transcription mediator of transforming growth factor-β (TGF-β) during the development and progression of liver fibrosis. However, the role of hepatocyte Smad4 in the development of fibrosis is poorly elucidated. Here, to explore the functional role of hepatocyte Smad4 and the molecular mechanism in liver fibrosis, a CCl4-induced liver fibrosis model was established in mice with hepatocyte-specific Smad4 deletion (Smad4Δhep). We found that hepatocyte-specific Smad4 deficiency reduced liver inflammation and fibrosis, alleviated epithelial-mesenchymal transition, and inhibited hepatocyte proliferation and migration. Molecularly, Smad4 deletion in hepatocytes suppressed the expression of inhibitor of differentiation 1 (ID1) and the secretion of connective tissue growth factor (CTGF) of hepatocytes, which subsequently activated the p38 and p65 signaling pathways of HSCs in an epidermal growth factor receptor-dependent manner. Taken together, our results clearly demonstrate that the Smad4 expression in hepatocytes plays an important role in promoting liver fibrosis and could therefore be a promising target for future anti-fibrotic therapy.
Collapse
Affiliation(s)
- Miaomiao Wei
- The College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China
| | - Xinlong Yan
- Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing 100124, China
| | - Xin Xin
- The College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China
| | - Haiqiang Chen
- The College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China
| | - Lingling Hou
- The College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China
| | - Jinhua Zhang
- The College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China
| |
Collapse
|
35
|
Maier BD, Aguilera LU, Sahle S, Mutz P, Kalra P, Dächert C, Bartenschlager R, Binder M, Kummer U. Stochastic dynamics of Type-I interferon responses. PLoS Comput Biol 2022; 18:e1010623. [PMID: 36269758 PMCID: PMC9629604 DOI: 10.1371/journal.pcbi.1010623] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 11/02/2022] [Accepted: 09/30/2022] [Indexed: 11/07/2022] Open
Abstract
Interferon (IFN) activates the transcription of several hundred of IFN stimulated genes (ISGs) that constitute a highly effective antiviral defense program. Cell-to-cell variability in the induction of ISGs is well documented, but its source and effects are not completely understood. The molecular mechanisms behind this heterogeneity have been related to randomness in molecular events taking place during the JAK-STAT signaling pathway. Here, we study the sources of variability in the induction of the IFN-alpha response by using MxA and IFIT1 activation as read-out. To this end, we integrate time-resolved flow cytometry data and stochastic modeling of the JAK-STAT signaling pathway. The complexity of the IFN response was matched by fitting probability distributions to time-course flow cytometry snapshots. Both, experimental data and simulations confirmed that the MxA and IFIT1 induction circuits generate graded responses rather than all-or-none responses. Subsequently, we quantify the size of the intrinsic variability at different steps in the pathway. We found that stochastic effects are transiently strong during the ligand-receptor activation steps and the formation of the ISGF3 complex, but negligible for the final induction of the studied ISGs. We conclude that the JAK-STAT signaling pathway is a robust biological circuit that efficiently transmits information under stochastic environments. We investigate the impact of intrinsic and extrinsic noise on the reliability of interferon signaling. Information must be transduced robustly despite existing biochemical variability and at the same time the system has to allow for cellular variability to tune it against changing environments. Getting insights into stochasticity in signaling networks is crucial to understand cellular dynamics and decision-making processes. To this end, we developed a detailed stochastic computational model based on single cell data. We are able to show that reliability is achieved despite high noise at the receptor level.
Collapse
Affiliation(s)
- Benjamin D. Maier
- Department of Modeling of Biological Processes, COS Heidelberg / Bioquant, Heidelberg University, Heidelberg, Germany
| | - Luis U. Aguilera
- Department of Modeling of Biological Processes, COS Heidelberg / Bioquant, Heidelberg University, Heidelberg, Germany
| | - Sven Sahle
- Department of Modeling of Biological Processes, COS Heidelberg / Bioquant, Heidelberg University, Heidelberg, Germany
| | - Pascal Mutz
- Division Virus-Associated Carcinogenesis, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department for Infectious Diseases, Molecular Virology, Medical Faculty, Heidelberg University, Heidelberg, Germany
| | - Priyata Kalra
- Department of Modeling of Biological Processes, COS Heidelberg / Bioquant, Heidelberg University, Heidelberg, Germany
| | - Christopher Dächert
- Research Group “Dynamics of early viral infection and the innate antiviral response”, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department for Infectious Diseases, Molecular Virology, Medical Faculty, Heidelberg University, Heidelberg, Germany
| | - Ralf Bartenschlager
- Division Virus-Associated Carcinogenesis, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department for Infectious Diseases, Molecular Virology, Medical Faculty, Heidelberg University, Heidelberg, Germany
| | - Marco Binder
- Research Group “Dynamics of early viral infection and the innate antiviral response”, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ursula Kummer
- Department of Modeling of Biological Processes, COS Heidelberg / Bioquant, Heidelberg University, Heidelberg, Germany
- * E-mail:
| |
Collapse
|
36
|
IRF3 inhibits nuclear translocation of NF-κB to prevent viral inflammation. Proc Natl Acad Sci U S A 2022; 119:e2121385119. [PMID: 36067309 PMCID: PMC9478676 DOI: 10.1073/pnas.2121385119] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Interferon (IFN) regulatory factor 3 (IRF3) is a transcription factor activated by phosphorylation in the cytoplasm of a virus-infected cell; by translocating to the nucleus, it induces transcription of IFN-β and other antiviral genes. We have previously reported IRF3 can also be activated, as a proapoptotic factor, by its linear polyubiquitination mediated by the RIG-I pathway. Both transcriptional and apoptotic functions of IRF3 contribute to its antiviral effect. Here, we report a nontranscriptional function of IRF3, namely, the repression of IRF3-mediated NF-κB activity (RIKA), which attenuated viral activation of NF-κB and the resultant inflammatory gene induction. In Irf3-/- mice, consequently, Sendai virus infection caused enhanced inflammation in the lungs. Mechanistically, RIKA was mediated by the direct binding of IRF3 to the p65 subunit of NF-κB in the cytoplasm, which prevented its nuclear import. A mutant IRF3 defective in both the transcriptional and the apoptotic activities was active in RIKA and inhibited virus replication. Our results demonstrated IRF3 deployed a three-pronged attack on virus replication and the accompanying inflammation.
Collapse
|
37
|
Son M, Frank T, Holst-Hansen T, Wang AG, Junkin M, Kashaf SS, Trusina A, Tay S. Spatiotemporal NF-κB dynamics encodes the position, amplitude, and duration of local immune inputs. SCIENCE ADVANCES 2022; 8:eabn6240. [PMID: 36044569 PMCID: PMC9432835 DOI: 10.1126/sciadv.abn6240] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 07/19/2022] [Indexed: 05/31/2023]
Abstract
Infected cells communicate through secreted signaling molecules like cytokines, which carry information about pathogens. How differences in cytokine secretion affect inflammatory signaling over space and how responding cells decode information from propagating cytokines are not understood. By computationally and experimentally studying NF-κB dynamics in cocultures of signal-sending cells (macrophages) and signal-receiving cells (fibroblasts), we find that cytokine signals are transmitted by wave-like propagation of NF-κB activity and create well-defined activation zones in responding cells. NF-κB dynamics in responding cells can simultaneously encode information about cytokine dose, duration, and distance to the cytokine source. Spatially resolved transcriptional analysis reveals that responding cells transmit local cytokine information to distance-specific proinflammatory gene expression patterns, creating "gene expression zones." Despite single-cell variability, the size and duration of the signaling zone are tightly controlled by the macrophage secretion profile. Our results highlight how macrophages tune cytokine secretion to control signal transmission distance and how inflammatory signaling interprets these signals in space and time.
Collapse
Affiliation(s)
- Minjun Son
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
- Institute for Genomics and Systems Biology, University of Chicago, Chicago, IL 60637, USA
| | - Tino Frank
- Department of Biosystems Science and Engineering, ETH Zürich, Basel 4058, Switzerland
| | | | - Andrew G. Wang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Michael Junkin
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
- Department of Biosystems Science and Engineering, ETH Zürich, Basel 4058, Switzerland
| | - Sara S. Kashaf
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Ala Trusina
- Niels Bohr Institute, University of Copenhagen, Copenhagen 2100, Denmark
| | - Savaş Tay
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
- Institute for Genomics and Systems Biology, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
38
|
Mechanistic Interrogation of Cell Transformation In Vitro: The Transformics Assay as an Exemplar of Oncotransformation. Int J Mol Sci 2022; 23:ijms23147603. [PMID: 35886950 PMCID: PMC9321586 DOI: 10.3390/ijms23147603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/21/2022] [Accepted: 07/03/2022] [Indexed: 12/19/2022] Open
Abstract
The Transformics Assay is an in vitro test which combines the BALB/c 3T3 Cell Transformation Assay (CTA) with microarray transcriptomics. It has been shown to improve upon the mechanistic understanding of the CTA, helping to identify mechanisms of action leading to chemical-induced transformation thanks to RNA extractions in specific time points along the process of in vitro transformation. In this study, the lowest transforming concentration of the carcinogenic benzo(a)pyrene (B(a)P) has been tested in order to find molecular signatures of initial events relevant for oncotransformation. Application of Enrichment Analysis (Metacore) to the analyses of the results facilitated key biological interpretations. After 72 h of exposure, as a consequence of the molecular initiating event of aryl hydrocarbon receptor (AhR) activation, there is a cascade of cellular events and microenvironment modification, and the immune and inflammatory responses are the main processes involved in cell response. Furthermore, pathways and processes related to cell cycle regulation, cytoskeletal adhesion and remodeling processes, cell differentiation and transformation were observed.
Collapse
|
39
|
The Peptide/Antibody-Based Surface Decoration of Calcium Phosphate Nanoparticles Carrying siRNA Influences the p65 NF-κB Protein Expression in Inflamed Cells In Vitro. Biomedicines 2022; 10:biomedicines10071571. [PMID: 35884877 PMCID: PMC9313450 DOI: 10.3390/biomedicines10071571] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 06/28/2022] [Accepted: 06/28/2022] [Indexed: 12/22/2022] Open
Abstract
Earlier studies with nanoparticles carrying siRNA were restricted to investigating the inhibition of target-specific protein expression, while almost ignoring effects related to the nanoparticle composition. Here, we demonstrate how the design and surface decoration of nanoparticles impact the p65 nuclear factor-kappa B (NF-κB) protein expression in inflamed leucocytes and endothelial cells in vitro. We prepared silica-coated calcium phosphate nanoparticles carrying encapsulated siRNA against p65 NF-κB and surface-decorated with peptides or antibodies. We show that RGD-decorated nanoparticles are efficient in down-regulating p65 NF-κB protein expression in endothelial cells as a result of an enhanced specific cellular binding and subsequent uptake of nanoparticles. In contrast, nanoparticles decorated with IgG (whether specific or not for CD69) are efficient in down-regulating p65 NF-κB protein expression in T-cells, but not in B-cells. Thus, an optimized nanoparticle decoration with xenogenic IgG may stimulate a specific cellular uptake. In summary, the composition of siRNA-loaded calcium phosphate nanoparticles can either weaken or stimulate p65 NF-κB protein expression in targeted inflamed leucocytes and endothelial cells. In general, unveiling such interactions may be very useful for the future design of anti-p65 siRNA-based nanomedicines for treatment of inflammation-associated diseases.
Collapse
|
40
|
Urolithin A Inactivation of TLR3/TRIF Signaling to Block the NF-κB/STAT1 Axis Reduces Inflammation and Enhances Antioxidant Defense in Poly(I:C)-Induced RAW264.7 Cells. Int J Mol Sci 2022; 23:ijms23094697. [PMID: 35563088 PMCID: PMC9101441 DOI: 10.3390/ijms23094697] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/18/2022] [Accepted: 04/20/2022] [Indexed: 02/04/2023] Open
Abstract
Urolithin A is an active compound of gut-microbiota-derived metabolites of polyphenol ellagic acid that has anti-aging, antioxidative, and anti-inflammatory effects. However, the effects of urolithin A on polyinosinic acid-polycytidylic acid (poly(I:C))-induced inflammation remain unclear. Poly(I:C) is a double-stranded RNA (dsRNA) similar to a virus and is recognized by Toll-like receptor-3 (TLR3), inducing an inflammatory response in immune cells, such as macrophages. Inflammation is a natural defense process of the innate immune system. Therefore, we used poly(I:C)-induced RAW264.7 cells and attenuated the inflammation induced by urolithin A. First, our data suggested that 1–30 μM urolithin A does not reduce RAW264.7 cell viability, whereas 1 μM urolithin A is sufficient for antioxidation and the decreased production of tumor necrosis factor-α (TNF-α), monocyte chemoattractant protein-1 (MCP-1), and C-C chemokine ligand 5. The inflammation-related proteins cyclooxygenase-2 and inducible nitric oxide synthase were also downregulated by urolithin A. Next, 1 μM urolithin A inhibited the levels of interferon (INF)-α and INF-β. Urolithin A was applied to investigate the blockade of the TLR3 signaling pathway in poly(I:C)-induced RAW264.7 cells. Moreover, the TLR3 signaling pathway, subsequent inflammatory-related pathways, and antioxidation pathways showed changes in nuclear factor-κB (NF-κB) signaling and blocked ERK/mitogen-activated protein kinase (MAPK) signaling. Urolithin A enhanced catalase (CAT) and superoxide dismutase (SOD) activities, but decreased malondialdehyde (MDA) levels in poly(I:C)-induced RAW264.7 cells. Thus, our results suggest that urolithin A inhibits TLR3-activated inflammatory and oxidative-associated pathways in macrophages, and that this inhibition is induced by poly(I:C). Therefore, urolithin A may have antiviral effects and could be used to treat viral-infection-related diseases.
Collapse
|
41
|
Al Hamrashdi M, Brady G. Regulation of IRF3 activation in Human Antiviral Signalling Pathways. Biochem Pharmacol 2022; 200:115026. [PMID: 35367198 DOI: 10.1016/j.bcp.2022.115026] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 12/24/2022]
Abstract
The interferon regulatory factor (IRF) family of transcription factors play a vital role in the human innate antiviral immune responses with production of interferons (IFNs) as a hallmark outcome of activation. In recent years, IRF3 has been considered a principal early regulator of type I IFNs (TI-IFNs) directly downstream of intracellular virus sensing. Despite decades of research on IRF-activating pathways, many questions remain on the regulation of IRF3 activation. The kinases IκB kinase epsilon (IKKε) and TANK-binding kinase-1 (TBK1) and the scaffold proteins TRAF family member-associated NF-kappa-B activator (TANK), NF-kappa-B-activating kinase-associated protein 1 (NAP1) and TANK-binding kinase 1-binding protein 1 (TBKBP1)/similar to NAP1 TBK1 adaptor (SINTBAD) are believed to be core components of an IRF3-activation complex yet their contextual involvement and complex composition are still unclear. This review will give an overview of antiviral signaling pathways leading to the activation of IRF3 and discuss recent developments in our understanding of its proximal regulation.
Collapse
Affiliation(s)
- Mariya Al Hamrashdi
- Trinity Translational Medicine Institute, Trinity College Dublin, St. James' Hospital Campus, Dublin, Ireland.
| | - Gareth Brady
- Trinity Translational Medicine Institute, Trinity College Dublin, St. James' Hospital Campus, Dublin, Ireland.
| |
Collapse
|
42
|
Chen Y, Zhong W, Xie Z, Li B, Li H, Gao K, Ning Z. Suppressor of cytokine signaling 1 (SOCS1) inhibits antiviral responses to facilitate Senecavirus A infection by regulating the NF-κB signaling pathway. Virus Res 2022; 313:198748. [PMID: 35304133 DOI: 10.1016/j.virusres.2022.198748] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/12/2022] [Accepted: 03/14/2022] [Indexed: 11/19/2022]
Abstract
Senecavirus A (SVA) is a new virus inducing porcine idiopathic vesicular disease that causes significant economic losses. Although some progress has been made in etiological research, the role of host factors in SVA infection remains unclear. This study investigated the role of the host factor, suppressor of cytokine signaling 1 (SOCS1), in SVA infection. The expression of SOCS1 was significantly upregulated with infection of SVA in a dose-dependent manner, and SOCS1 inhibited the expression of type I interferons (IFN-α, IFN-β) and the production of interferon stimulating genes (ISGs) (ISG56, ISG54, PKR), thereby facilitating viral replication. Further results showed that inhibition of antiviral responses of SOCS1 was achieved by regulating the NF-κB signaling pathway, which attenuates the production of IFNs and pro-inflammatory cytokines. These findings provide a new perspective of SVA pathogenesis and may partially explain the persistence of this infection. Moreover, the data indicate that targeting SOCS1 can help in developing new agents against SVA infection.
Collapse
Affiliation(s)
- Yongjie Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Wenxia Zhong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Zhenxin Xie
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Baojian Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Huizi Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Kuipeng Gao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Zhangyong Ning
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Maoming Branch, Maoming 525000, China.
| |
Collapse
|
43
|
The RIG-I-NRF2 axis regulates the mesenchymal stromal niche for bone marrow transplantation. Blood 2022; 139:3204-3221. [PMID: 35259210 DOI: 10.1182/blood.2021013048] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 02/18/2022] [Indexed: 11/20/2022] Open
Abstract
Bone marrow-derived mesenchymal stem cells (BMSCs) support bone formation and constitute the stromal niche in regulating hematopoietic stem cells (HSCs). Stromal niche dysfunction affects HSC engraftment during transplantation; however, the underlying mechanisms remain elusive. In the present study, we found that all-trans retinoic acid (ATRA) and inflammation stress upregulated retinoic acid-inducible gene I (RIG-I) in BMSCs. Excess RIG-I expression damaged the clonogenicity, bone-forming ability of BMSCs and, particularly, their stromal niche function that supports HSC expansion in vitro and engraftment in vivo. Mechanistically, RIG-I elevation promoted the degradation of NRF2, a checkpoint for antioxidant cellular response, by altering the RIG-I-Trim25-Keap1-NRF2 complex, leading to reactive oxygen species (ROS) accumulation and BMSC damage. Genetic inhibition of RIG-I sustained NRF2 protein levels and reduced ROS levels in ATRA-treated BMSCs, thus preserving their clonogenicity, bone-forming ability, and stromal niche function in supporting HSC engraftment in mice. More importantly, RIG-I inhibition recovered the ATRA-treated stromal niche function, to enhance HSC engraftment and emergency myelopoiesis for innate immunity against the bacterium Listeria monocytogenes during transplantation. Overall, we identified a non-canonical role of RIG-I in the regulation of the stromal niche for HSC transplantation.
Collapse
|
44
|
Akaishi T, Yamamoto S, Abe K. 3',4',7-Trihydroxyflavone Downregulates NO Production in LPS- or IFN-γ-Activated MG6 Microglial Cells by Attenuating the JNK-STAT1 Pathway. Biol Pharm Bull 2022; 45:301-308. [PMID: 35228396 DOI: 10.1248/bpb.b21-00841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Neuroinflammation induced by activated microglia is a key feature of neurodegenerative diseases such as Alzheimer's disease. The natural flavonoid 3',4',7-trihydroxyflavone protects nerve cells from oxidative stress-mediated apoptosis and inhibits the aggregation of amyloid β protein in vitro. However, little is known about its effects on microglial activation. In this study, we investigated the effects of 3',4',7-trihydroxyflavone on lipopolysaccharide (LPS)- or interferon-γ (IFN-γ)-induced neuroinflammatory responses in MG6 microglial cells. 3',4',7-Trihydroxyflavone inhibited LPS- or IFN-γ-mediated nitric oxide (NO) generation and the upregulation of inducible NO synthase (iNOS) in MG6 cells. 3',4',7-Trihydroxyflavone also suppressed LPS- or IFN-γ-mediated phosphorylation of signal transducer and activator of transcription 1 (STAT1), which is crucial for iNOS expression. LPS stimulation induced rapid phosphorylation of c-Jun N-terminal kinase (JNK), p38 mitogen-activated protein kinase (MAPK), and extracellular signal-regulated kinase (ERK) in MG6 cells. 3',4',7-Trihydroxyflavone significantly inhibited the LPS-mediated phosphorylation of JNK, but not that of ERK and p38 MAPK. The inhibitory effect of 3',4',7-trihydroxyflavone on NO generation was mimicked by pharmacological inhibition of the JNK signaling pathway with SP600125. Furthermore, SP600125 significantly inhibited LPS- or IFN-γ-mediated phosphorylation of STAT1 in MG6 cells. These results suggest that 3',4',7-trihydroxyflavone exerts anti-neuroinflammatory effects via inhibition of the JNK-STAT1 pathway in microglia.
Collapse
Affiliation(s)
- Tatsuhiro Akaishi
- Laboratory of Pharmacology, Faculty of Pharmacy and Research Institute of Pharmaceutical Sciences, Musashino University
| | - Shohei Yamamoto
- Laboratory of Pharmacology, Faculty of Pharmacy and Research Institute of Pharmaceutical Sciences, Musashino University
| | - Kazuho Abe
- Laboratory of Pharmacology, Faculty of Pharmacy and Research Institute of Pharmaceutical Sciences, Musashino University
| |
Collapse
|
45
|
Abstract
A new model of viral infection spreading in cell cultures is proposed taking into account virus mutation. This model represents a reaction-diffusion system of equations with time delay for the concentrations of uninfected cells, infected cells and viral load. Infection progression is characterized by the virus replication number Rv, which determines the total viral load. Analytical formulas for the speed of propagation and for the viral load are obtained and confirmed by numerical simulations. It is shown that virus mutation leads to the emergence of a new virus variant. Conditions of the coexistence of the two variants or competitive exclusion of one of them are found, and different stages of infection progression are identified.
Collapse
|
46
|
Brasier AR, Qiao D, Zhao Y. The Hexosamine Biosynthetic Pathway Links Innate Inflammation With Epithelial-Mesenchymal Plasticity in Airway Remodeling. Front Pharmacol 2021; 12:808735. [PMID: 35002741 PMCID: PMC8727908 DOI: 10.3389/fphar.2021.808735] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 12/07/2021] [Indexed: 01/15/2023] Open
Abstract
Disruption of the lower airway epithelial barrier plays a major role in the initiation and progression of chronic lung disease. Here, repetitive environmental insults produced by viral and allergens triggers metabolic adaptations, epithelial-mesenchymal plasticity (EMP) and airway remodeling. Epithelial plasticity disrupts epithelial barrier function, stimulates release of fibroblastic growth factors, and remodels the extracellular matrix (ECM). This review will focus on recent work demonstrating how the hexosamine biosynthetic pathway (HBP) links innate inflammation to airway remodeling. The HBP is a core metabolic pathway of the unfolded protein response (UPR) responsible for protein N-glycosylation, relief of proteotoxic stress and secretion of ECM modifiers. We will overview findings that the IκB kinase (IKK)-NFκB pathway directly activates expression of the SNAI-ZEB1 mesenchymal transcription factor module through regulation of the Bromodomain Containing Protein 4 (BRD4) chromatin modifier. BRD4 mediates transcriptional elongation of SNAI1-ZEB as well as enhancing chromatin accessibility and transcription of fibroblast growth factors, ECM and matrix metalloproteinases (MMPs). In addition, recent exciting findings that IKK cross-talks with the UPR by controlling phosphorylation and nuclear translocation of the autoregulatory XBP1s transcription factor are presented. HBP is required for N glycosylation and secretion of ECM components that play an important signaling role in airway remodeling. This interplay between innate inflammation, metabolic reprogramming and lower airway plasticity expands a population of subepithelial myofibroblasts by secreting fibroblastic growth factors, producing changes in ECM tensile strength, and fibroblast stimulation by MMP binding. Through these actions on myofibroblasts, EMP in lower airway cells produces expansion of the lamina reticularis and promotes airway remodeling. In this manner, metabolic reprogramming by the HBP mediates environmental insult-induced inflammation with remodeling in chronic airway diseases.
Collapse
Affiliation(s)
- Allan R. Brasier
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health (SMPH), Madison, WI, United States
- Institute for Clinical and Translational Research (ICTR), University of Wisconsin-Madison, Madison, WI, United States
| | - Dianhua Qiao
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health (SMPH), Madison, WI, United States
| | - Yingxin Zhao
- Department of Internal Medicine, University of Texas Medical Branch Galveston, Galveston, TX, United States
| |
Collapse
|
47
|
AlDaif BA, Mercer AA, Fleming SB. The parapoxvirus Orf virus ORF116 gene encodes an antagonist of the interferon response. J Gen Virol 2021; 102. [PMID: 34890310 DOI: 10.1099/jgv.0.001695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Orf virus (ORFV) is the type species of the Parapoxvirus genus of the Poxviridae family. Genetic and functional studies have revealed ORFV has multiple immunomodulatory genes that manipulate innate immune responses, during the early stage of infection. ORF116 is a novel gene of ORFV with hitherto unknown function. Characterization of an ORF116 deletion mutant showed that it replicated in primary lamb testis cells with reduced levels compared to the wild-type and produced a smaller plaque phenotype. ORF116 was shown to be expressed prior to DNA replication. The potential function of ORF116 was investigated by gene-expression microarray analysis in HeLa cells infected with wild-type ORFV or the ORF116 deletion mutant. The analysis of differential cellular gene expression revealed a number of interferon-stimulated genes (ISGs) differentially expressed at either 4 or 6 h post infection. IFI44 showed the greatest differential expression (4.17-fold) between wild-type and knockout virus. Other ISGs that were upregulated in the knockout included RIG-I, IFIT2, MDA5, OAS1, OASL, DDX60, ISG20 and IFIT1 and in addition the inflammatory cytokine IL-8. These findings were validated by infecting HeLa cells with an ORF116 revertant recombinant virus and analysis of transcript expression by quantitative real time-PCR (qRT-PCR). These observations suggested a role for the ORFV gene ORF116 in modulating the IFN response and inflammatory cytokines. This study represents the first functional analysis of ORF116.
Collapse
Affiliation(s)
- Basheer A AlDaif
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Andrew A Mercer
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Stephen B Fleming
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
48
|
Wang C, Li J, Yang X, Wang Q, Zhong H, Liu Y, Yan W, He Y, Deng Z, Xiao J, Feng H. Black carp IKKε collaborates with IRF3 in the antiviral signaling. FISH & SHELLFISH IMMUNOLOGY 2021; 118:160-168. [PMID: 34500054 DOI: 10.1016/j.fsi.2021.08.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 08/25/2021] [Accepted: 08/27/2021] [Indexed: 06/13/2023]
Abstract
Interferon regulatory factor 3 (IRF3) is activated by IκB kinase ε (IKKε) and Tank-binding kinase 1 (TBK1), which plays a crucial role in the interferon signaling in vertebrates. However, the regulation of teleost IRF3 by IKKε remains largely unknown. In this study, the IRF3 homologue (bcIRF3) of black carp (Mylopharyngodon piceus) has been cloned and characterized. The transcription of bcIRF3 was detected to increase in host cells in response to different stimuli. bcIRF3 distributed predominantly in the cytosolic area; however, translocated into nuclei after virus infection. bcIRF3 showed IFN-inducing ability in reporter assay and EPC cells expressing bcIRF3 showed enhanced antiviral ability against both grass carp reovirus (GCRV) and spring viremia of carp virus (SVCV). Moreover, knockdown of bcIRF3 reduced the antiviral ability of the host cells, and the transcription of antiviral-related cytokines was obviously lower in bcIRF3-deficient host cells than that of control cells. The data of reporter assay and plaque assay demonstrated that bcIKKε obviously enhanced bcIRF3-mediated IFN production and antiviral activity. Immunofluorescent staining and co-immunoprecipitation assay revealed that bcIKKε interacted with bcIRF3. It was interesting that the nuclear translocation of bcIRF3 and bcIKKε was enhanced by each other when these two molecules were co-expressed in the cells, however, the protein levels of bcIRF3 and bcIKKε were decreased mutually. Thus, our data support the conclusion that bcIKKε interacts with bcIRF3 and enhances bcIRF3-mediated antiviral signaling during host innate immune activation.
Collapse
Affiliation(s)
- Chanyuan Wang
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Jun Li
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China; Key Laboratory of Hunan Province for Study and Utilization of Ethnic Medicinal Plant Resources, College of Biological and Food Engineering, Huaihua University, Huaihua, 418008, China
| | - Xiao Yang
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Qun Wang
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Huijuan Zhong
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Yankai Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Weiyi Yan
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Yunfan He
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Zhuoyi Deng
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Jun Xiao
- 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
|
49
|
Kell AM. Innate Immunity to Orthohantaviruses: Could Divergent Immune Interactions Explain Host-specific Disease Outcomes? J Mol Biol 2021; 434:167230. [PMID: 34487792 PMCID: PMC8894506 DOI: 10.1016/j.jmb.2021.167230] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 10/20/2022]
Abstract
The genus Orthohantavirus (family Hantaviridae, order Bunyavirales) consists of numerous genetic and pathologically distinct viral species found within rodent and mammalian insectivore populations world-wide. Although reservoir hosts experience persistent asymptomatic infection, numerous rodent-borne orthohantaviruses cause severe disease when transmitted to humans, with case-fatality rates up to 40%. The first isolation of an orthohantavirus occurred in 1976 and, since then, the field has made significant progress in understanding the immune correlates of disease, viral interactions with the human innate immune response, and the immune kinetics of reservoir hosts. Much still remains elusive regarding the molecular mechanisms of orthohantavirus recognition by the innate immune response and viral antagonism within the reservoir host, however. This review provides a summary of the last 45 years of research into orthohantavirus interaction with the host innate immune response. This summary includes discussion of current knowledge involving human, non-reservoir rodent, and reservoir innate immune responses to viruses which cause hemorrhagic fever with renal syndrome and hantavirus cardio-pulmonary syndrome. Review of the literature concludes with a brief proposition for the development of novel tools needed to drive forward investigations into the molecular mechanisms of innate immune activation and consequences for disease outcomes in the various hosts for orthohantaviruses.
Collapse
Affiliation(s)
- Alison M Kell
- Department of Molecular Genetics and Microbiology, University of New Mexico, 915 Camino de Salud, Albuquerque, NM 87131, United States.
| |
Collapse
|
50
|
Van Eyndhoven LC, Singh A, Tel J. Decoding the dynamics of multilayered stochastic antiviral IFN-I responses. Trends Immunol 2021; 42:824-839. [PMID: 34364820 DOI: 10.1016/j.it.2021.07.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/11/2021] [Accepted: 07/11/2021] [Indexed: 12/11/2022]
Abstract
Type I Interferon (IFN-I) responses were first recognized for their role in antiviral immunity, but it is now widely appreciated that IFN-Is have many immunomodulatory functions, influencing antitumor responses, autoimmune manifestations, and antimicrobial defenses. Given these pivotal roles, it may be surprising that multilayered stochastic events create highly heterogeneous, but tightly regulated, all-or-nothing cellular decisions. Recently, mathematical models have provided crucial insights into the stochastic nature of antiviral IFN-I responses, which we critically evaluate in this review. In this context, we emphasize the need for innovative single-cell technologies combined with mathematical models to further reveal, understand, and predict the complexity of the IFN-I system in physiological and pathological conditions that may be relevant to a plethora of diseases.
Collapse
Affiliation(s)
- Laura C Van Eyndhoven
- Laboratory of Immunoengineering, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands; Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Abhyudai Singh
- Department of Electrical and Computer Engineering, University of Delaware, Newark, Delaware, USA
| | - Jurjen Tel
- Laboratory of Immunoengineering, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands; Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, The Netherlands.
| |
Collapse
|