1
|
Siff TE, Allen PE, Armistead DL, Hunt JR, Rolland SJ, Agaisse H, Carlyon JA. Orientia tsutsugamushi Modulates RIPK3 Cellular Levels but Does Not Inhibit Necroptosis. Pathogens 2025; 14:478. [PMID: 40430799 PMCID: PMC12114526 DOI: 10.3390/pathogens14050478] [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: 04/09/2025] [Revised: 05/06/2025] [Accepted: 05/11/2025] [Indexed: 05/29/2025] Open
Abstract
Scrub typhus is an emerging chigger-borne disease caused by the obligate intracellular bacterium Orientia tsutsugamushi. Necroptosis is a form of programmed cell death (PCD) mediated by RIPK3 (serine/threonine kinase receptor interacting protein 3) and its downstream effector MLKL (mixed-lineage kinase domain-like). While O. tsutsugamushi modulates apoptosis, another form of PCD, its interplay with necroptosis is unknown. Much of Orientia pathobiology is linked to its ankyrin repeat (AR)-containing effectors (Anks). Two of these, Ank1 and Ank6, share similarities with the cowpox AR protein, vIRD (viral inducer of RIPK3 degradation) that prevents necroptosis. Here, we show that Ank1 and Ank6 reduce RIPK3 cellular levels although not as robustly as and mechanistically distinct from vIRD. Orientia infection lowers RIPK3 amounts and does not elicit necroptosis in endothelial cells. In HeLa cells ectopically expressing RIPK3, Orientia fails to inhibit RIPK3 and MLKL phosphorylation as well as cell death. MLKL colocalization with Orientia or Listeria monocytogenes, another intracytoplasmic pathogen, was not observed. Thus, O. tsutsugamushi reduces cellular levels of RIPK3 and does not elicit necroptosis but cannot inhibit this PCD pathway once it is induced. This study is a first step toward understanding how the relationship between Orientia and necroptosis contributes to scrub typhus pathogenesis.
Collapse
Affiliation(s)
- Thomas E. Siff
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, VA 23298, USA; (T.E.S.); (P.E.A.); (D.L.A.); (J.R.H.)
| | - Paige E. Allen
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, VA 23298, USA; (T.E.S.); (P.E.A.); (D.L.A.); (J.R.H.)
| | - David L. Armistead
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, VA 23298, USA; (T.E.S.); (P.E.A.); (D.L.A.); (J.R.H.)
| | - Jason R. Hunt
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, VA 23298, USA; (T.E.S.); (P.E.A.); (D.L.A.); (J.R.H.)
| | - Steven J. Rolland
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; (S.J.R.); (H.A.)
| | - Hervé Agaisse
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; (S.J.R.); (H.A.)
| | - Jason A. Carlyon
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University Medical Center, Richmond, VA 23298, USA; (T.E.S.); (P.E.A.); (D.L.A.); (J.R.H.)
| |
Collapse
|
2
|
Chakraborty R, Chronopoulos J, Sun R, Morozan A, Joy S, Divangahi M, Lauzon AM, Martin JG. Anti-ST2 antibody reduces airway hyperresponsiveness mediated by monocyte-derived macrophages during influenza A infection. Mucosal Immunol 2025:S1933-0219(25)00046-7. [PMID: 40319941 DOI: 10.1016/j.mucimm.2025.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 03/23/2025] [Accepted: 04/27/2025] [Indexed: 05/07/2025]
Abstract
Influenza A virus (IAV) infections trigger asthma attacks and cause airway hyperresponsiveness (AHR) in murine models. However, the mechanism by which AHR is induced remains to be fully elucidated. Here, we show that targeting the interleukin (IL)-33 suppression of tumorigenicity 2 (ST2) receptor complex with an anti-ST2 antibody during acute IAV infection of C57BL/6 mice reduced AHR, without affecting expansion of ILC2s and independently of IL-13. Among the lung inflammatory cells, the anti-ST2 antibody selectively reduced the monocyte-derived macrophages (MMs). Furthermore, AHR was reduced in C-C chemokine receptor 2 (CCR2)-knockout mice that have deficient MM recruitment. Depletion of MMs achieved by anti-Ly6C antibody administration also reduced AHR. The treatment of airway smooth muscle (ASM) with conditioned medium from IL-33-treated human THP-1-derived macrophages enhanced potassium chloride-induced ASM contraction. These findings suggest that MMs contribute to acute AHR following IAV infection in an IL-33-dependent manner, but independent of the ILC2/IL-13 axis. Additionally, IL-33 stimulates the release of macrophage-derived mediators that enhance airway smooth muscle contraction, offering a potential mechanistic basis for IAV-induced AHR.
Collapse
Affiliation(s)
- Rohin Chakraborty
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.
| | - Julia Chronopoulos
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.
| | - Rui Sun
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.
| | - Arina Morozan
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.
| | - Sydney Joy
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.
| | - Maziar Divangahi
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.
| | - Anne-Marie Lauzon
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.
| | - James G Martin
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.
| |
Collapse
|
3
|
Boyd DF, Jordan SV, Balachandran S. ZBP1-driven cell death in severe influenza. Trends Microbiol 2025; 33:521-532. [PMID: 39809680 DOI: 10.1016/j.tim.2024.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025]
Abstract
Influenza A virus (IAV) infections can cause life-threatening illness in humans. The severity of disease is directly linked to virus replication in the alveoli of the lower respiratory tract. In particular, the lytic death of infected alveolar epithelial cells (AECs) is a major driver of influenza severity. Recent studies have begun to define the molecular mechanisms by which IAV triggers lytic cell death. Z-form nucleic-acid-binding protein 1 (ZBP1) senses replicating IAV and drives programmed cell death (PCD) in infected cells, including apoptosis and necroptosis in AECs and pyroptosis in myeloid cells. Necroptosis and pyroptosis, both lytic forms of death, contribute to pathogenesis during severe infections. Pharmacological blockade of necroptosis shows strong therapeutic potential in mouse models of lethal influenza. We suggest that targeting ZBP1-initiated necroinflammatory cell lysis, either alone or in combination antiviral drugs, will provide clinical benefit in severe influenza.
Collapse
Affiliation(s)
- David F Boyd
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, CA, USA.
| | - Summer Vaughn Jordan
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, CA, USA
| | | |
Collapse
|
4
|
Li Y, Qi Q, Chen Y, Ding M, Huang M, Huang C, Liu P, Gao X, Guo X, Zheng Z. RIPK3 activation of CaMKII triggers mitochondrial apoptosis in NIBV-infected renal tubular epithelial cells. Vet Microbiol 2025; 302:110375. [PMID: 39808936 DOI: 10.1016/j.vetmic.2025.110375] [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: 10/02/2024] [Revised: 01/04/2025] [Accepted: 01/05/2025] [Indexed: 01/16/2025]
Abstract
The purpose of this study was to investigate whether RIPK3-mediated programmed cell death can promote the replication and transmission of renal infectious bronchitis virus in renal tubular epithelial cells. Primary renal tubular epithelial cells were extracted from 1 to 7 day old Hy-Line Brown chicks, cultured in vitro by type I collagenase digestion, and infected with 1MOI SX9 strain. Cell samples were collected at 12 hpi, 24 hpi, 36 hpi and 48 hpi for experimental exploration. Our results showed that NIBV infection could lead to programmed necrosis and mitochondrial apoptosis, and the expression levels of programmed necrosis-related genes TNFR1, TRADD, FADD, RIPK1, RIPK3 and MLKL increased significantly with the extension of infection time, the expression levels of mitochondrial apoptosis-related genes Cyt-C, APAF-1, Caspase-9 and Csapase-3 were significantly increased at 36 hpi. While, after 36 hpi of virus infection, apoptosis decreased and necrosis increased, and virus replication peaked. In order to further explore the effect of necroptosis on the amplification of renal infectious virus, the RIPK3 was inhibited at 36 hpi. Inhibition of necroptosis could reduce viral replication and cell death, programmed necrosis occurred in the cells, and cell membrane perforation led to virus diffusion and replication. NIBV-induced necroptosis depends on RIPK3, and RIPK3 activates CAMKII and interacts to cause abnormal opening of mitochondrial membrane permeability transition pore, promotes Ca2 + influx into mitochondria, initiates mitochondrial apoptosis. While, inhibition of RIPK3 significantly inhibited the programmed necrosis of cells caused by NIBV infection, making excessive necrosis into moderate necrosis, thereby inhibiting the replication of the virus in cells.
Collapse
Affiliation(s)
- Ying Li
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China.
| | - Qiurong Qi
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China.
| | - Yifei Chen
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China.
| | - Mengbing Ding
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China.
| | - Manzi Huang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China.
| | - Cheng Huang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China.
| | - Ping Liu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China.
| | - Xiaona Gao
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China.
| | - Xiaoquan Guo
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China.
| | - Zhanhong Zheng
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China.
| |
Collapse
|
5
|
Xu H, Li H, Sun B, Sun L. An intracellular bacterial pathogen triggers RIG-I/MDA5-dependent necroptosis. CURRENT RESEARCH IN MICROBIAL SCIENCES 2024; 7:100318. [PMID: 39726972 PMCID: PMC11670418 DOI: 10.1016/j.crmicr.2024.100318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2024] Open
Abstract
RIG-I and MDA5 are members of RIG-I-like receptors (RLRs) that detect viral RNA within the cytoplasm and subsequently initiate antiviral immune responses. Necroptosis is a form of programmed cell death (PCD) executed by mixed lineage kinase domain-like (MLKL), which, upon phosphorylation by receptor-interacting protein kinase 3 (RIPK3), causes necrotic cell death. To date, no link between RLRs and necroptosis has been observed during bacterial infection. Edwardsiella tarda is a zoonotic bacterial pathogen that can thrive in host macrophages. In a previous study, we identified RIG-I and MDA5 as two hub factors of RAW264.7 cells responsive to E. tarda infection. The present study aimed to determine the specific form of cell death triggered by E. tarda and explore the association between RIG-I/MDA5 and PCD in the context of bacterial infection. Our results showed that E. tarda infection induced RIPK3-MLKL-mediated necroptosis, rather than pyroptosis or apoptosis, in RAW264.7 cells. Meanwhile, E. tarda promoted RIG-I/MDA5 production and activated the RIG-I/MDA5 pathways that led to IRF3 phosphorylation, IFN-β secretion, and interferon-stimulated gene (ISG) and cytokine expression. Both RIG-I and MDA5 were essential for E. tarda-triggered necroptosis and required for effective inhibition of intracellular bacterial replication. Furthermore, the regulatory effect of RIG-I/MDA5 on necroptosis was not affected by type I IFN or TNF-α signaling blockage. Together these results revealed that necroptosis could be triggered by intracellular bacterial infection through the RIG-I/MDA5 pathways, and that there existed intricate interplays between PCD and RLRs induced by bacterial pathogen.
Collapse
Affiliation(s)
- Hang Xu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology; CAS Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
- College of Marine Sciences, University of Chinese Academy of Sciences, Qingdao, China
| | - Huili Li
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology; CAS Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
- College of Marine Sciences, University of Chinese Academy of Sciences, Qingdao, China
| | - Boguang Sun
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology; CAS Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
- College of Marine Sciences, University of Chinese Academy of Sciences, Qingdao, China
| | - Li Sun
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology; CAS Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
- College of Marine Sciences, University of Chinese Academy of Sciences, Qingdao, China
| |
Collapse
|
6
|
Liu X, Chen W, Li K, Sheng J. RNA N6-methyladenosine methylation in influenza A virus infection. Front Microbiol 2024; 15:1401997. [PMID: 38957616 PMCID: PMC11217485 DOI: 10.3389/fmicb.2024.1401997] [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: 03/16/2024] [Accepted: 05/30/2024] [Indexed: 07/04/2024] Open
Abstract
Influenza A virus (IAV) is a negative-sense single-stranded RNA virus that causes acute lung injury and acute respiratory distress syndrome, posing a serious threat to both animal and human health. N6-methyladenosine (m6A), a prevalent and abundant post-transcriptional methylation of RNA in eukaryotes, plays a crucial regulatory role in IAV infection by altering viral RNA and cellular transcripts to affect viral infection and the host immune response. This review focuses on the molecular mechanisms underlying m6A modification and its regulatory function in the context of IAV infection and the host immune response. This will provide a better understanding of virus-host interactions and offer insights into potential anti-IAV strategies.
Collapse
Affiliation(s)
- Xueer Liu
- Department of Microbiology and Immunology, Guangdong Provincial Key Laboratory of Infectious Disease and Molecular Immunopathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Weiqiang Chen
- Department of Neurosurgery, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Kangsheng Li
- Department of Microbiology and Immunology, Guangdong Provincial Key Laboratory of Infectious Disease and Molecular Immunopathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Jiangtao Sheng
- Department of Microbiology and Immunology, Guangdong Provincial Key Laboratory of Infectious Disease and Molecular Immunopathology, Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
7
|
Pernet E, Poschmann J, Divangahi M. A complex immune communication between eicosanoids and pulmonary macrophages. Curr Opin Virol 2024; 66:101399. [PMID: 38547562 DOI: 10.1016/j.coviro.2024.101399] [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: 01/23/2024] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 06/07/2024]
Abstract
Respiratory viral infections represent a constant threat for human health and urge for a better understanding of the pulmonary immune response to prevent disease severity. Macrophages are at the center of pulmonary immunity, where they play a pivotal role in orchestrating beneficial and/or pathological outcomes during infection. Eicosanoids, the host bioactive lipid mediators, have re-emerged as important regulators of pulmonary immunity during respiratory viral infections. In this review, we summarize the current knowledge linking eicosanoids' and pulmonary macrophages' homeostatic and antimicrobial functions and discuss eicosanoids as emerging targets for immunotherapy in viral infection.
Collapse
Affiliation(s)
- Erwan Pernet
- Department of Medical Biology, Université du Québec à Trois-Rivières, Québec, Canada.
| | - Jeremie Poschmann
- INSERM, Nantes Université, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, ITUN, Nantes, France
| | - Maziar Divangahi
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada.
| |
Collapse
|
8
|
Tran KA, Pernet E, Sadeghi M, Downey J, Chronopoulos J, Lapshina E, Tsai O, Kaufmann E, Ding J, Divangahi M. BCG immunization induces CX3CR1 hi effector memory T cells to provide cross-protection via IFN-γ-mediated trained immunity. Nat Immunol 2024; 25:418-431. [PMID: 38225437 DOI: 10.1038/s41590-023-01739-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 12/20/2023] [Indexed: 01/17/2024]
Abstract
After a century of using the Bacillus Calmette-Guérin (BCG) vaccine, our understanding of its ability to provide protection against homologous (Mycobacterium tuberculosis) or heterologous (for example, influenza virus) infections remains limited. Here we show that systemic (intravenous) BCG vaccination provides significant protection against subsequent influenza A virus infection in mice. We further demonstrate that the BCG-mediated cross-protection against influenza A virus is largely due to the enrichment of conventional CD4+ effector CX3CR1hi memory αβ T cells in the circulation and lung parenchyma. Importantly, pulmonary CX3CR1hi T cells limit early viral infection in an antigen-independent manner via potent interferon-γ production, which subsequently enhances long-term antimicrobial activity of alveolar macrophages. These results offer insight into the unknown mechanism by which BCG has persistently displayed broad protection against non-tuberculosis infections via cross-talk between adaptive and innate memory responses.
Collapse
Affiliation(s)
- Kim A Tran
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, Research Institute of the McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
| | - Erwan Pernet
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, Research Institute of the McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
- Department of Medical Biology, Université du Québec à Trois-Rivières, Quebec, Quebec, Canada
| | - Mina Sadeghi
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, Research Institute of the McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
| | - Jeffrey Downey
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, Research Institute of the McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
| | - Julia Chronopoulos
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, Research Institute of the McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
| | - Elizabeth Lapshina
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, Research Institute of the McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
| | - Oscar Tsai
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, Research Institute of the McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
| | - Eva Kaufmann
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, Research Institute of the McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Jun Ding
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, Research Institute of the McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
| | - Maziar Divangahi
- Department of Medicine, Department of Pathology, Department of Microbiology & Immunology, Research Institute of the McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
9
|
Casalino-Matsuda SM, Chen F, Gonzalez-Gonzalez FJ, Matsuda H, Nair A, Abdala-Valencia H, Budinger GS, Dong JT, Beitel GJ, Sporn PH. Myeloid Zfhx3 deficiency protects against hypercapnia-induced suppression of host defense against influenza A virus. JCI Insight 2024; 9:e170316. [PMID: 38227369 PMCID: PMC11143927 DOI: 10.1172/jci.insight.170316] [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: 03/10/2023] [Accepted: 01/10/2024] [Indexed: 01/17/2024] Open
Abstract
Hypercapnia, elevation of the partial pressure of CO2 in blood and tissues, is a risk factor for mortality in patients with severe acute and chronic lung diseases. We previously showed that hypercapnia inhibits multiple macrophage and neutrophil antimicrobial functions and that elevated CO2 increases the mortality of bacterial and viral pneumonia in mice. Here, we show that normoxic hypercapnia downregulates innate immune and antiviral gene programs in alveolar macrophages (AMØs). We also show that zinc finger homeobox 3 (Zfhx3) - a mammalian ortholog of zfh2, which mediates hypercapnic immune suppression in Drosophila - is expressed in mouse and human macrophages. Deletion of Zfhx3 in the myeloid lineage blocked the suppressive effect of hypercapnia on immune gene expression in AMØs and decreased viral replication, inflammatory lung injury, and mortality in hypercapnic mice infected with influenza A virus. To our knowledge, our results establish Zfhx3 as the first known mammalian mediator of CO2 effects on immune gene expression and lay the basis for future studies to identify therapeutic targets to interrupt hypercapnic immunosuppression in patients with advanced lung disease.
Collapse
Affiliation(s)
- S. Marina Casalino-Matsuda
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Fei Chen
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Francisco J. Gonzalez-Gonzalez
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Hiroaki Matsuda
- Department of Physical Sciences and Engineering, Wilbur Wright College, Chicago, Illinois, USA
| | - Aisha Nair
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Hiam Abdala-Valencia
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - G.R. Scott Budinger
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Research Service, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| | - Jin-Tang Dong
- Department of Human Cell Biology and Genetics, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Greg J. Beitel
- Department of Molecular Biosciences, Weinberg College of Arts and Sciences, Northwestern University, Evanston, Illinois, USA
| | - Peter H.S. Sporn
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Research Service, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| |
Collapse
|
10
|
Lindman M, Angel JP, Estevez I, Chang NP, Chou TW, McCourt M, Atkins C, Daniels BP. RIPK3 promotes brain region-specific interferon signaling and restriction of tick-borne flavivirus infection. PLoS Pathog 2023; 19:e1011813. [PMID: 38011306 PMCID: PMC10703404 DOI: 10.1371/journal.ppat.1011813] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 12/07/2023] [Accepted: 11/09/2023] [Indexed: 11/29/2023] Open
Abstract
Innate immune signaling in the central nervous system (CNS) exhibits many remarkable specializations that vary across cell types and CNS regions. In the setting of neuroinvasive flavivirus infection, neurons employ the immunologic kinase receptor-interacting kinase 3 (RIPK3) to promote an antiviral transcriptional program, independently of the traditional function of this enzyme in promoting necroptotic cell death. However, while recent work has established roles for neuronal RIPK3 signaling in controlling mosquito-borne flavivirus infections, including West Nile virus and Zika virus, functions for RIPK3 signaling in the CNS during tick-borne flavivirus infection have not yet been explored. Here, we use a model of Langat virus (LGTV) encephalitis to show that RIPK3 signaling is specifically required in neurons of the cerebellum to control LGTV replication and restrict disease pathogenesis. This effect did not require the necroptotic executioner molecule mixed lineage kinase domain like protein (MLKL), a finding similar to previous observations in models of mosquito-borne flavivirus infection. However, control of LGTV infection required a unique, region-specific dependence on RIPK3 to promote expression of key antiviral interferon-stimulated genes (ISG) in the cerebellum. This RIPK3-mediated potentiation of ISG expression was associated with robust cell-intrinsic restriction of LGTV replication in cerebellar granule cell neurons. These findings further illuminate the complex roles of RIPK3 signaling in the coordination of neuroimmune responses to viral infection, as well as provide new insight into the mechanisms of region-specific innate immune signaling in the CNS.
Collapse
Affiliation(s)
- Marissa Lindman
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey, United States of America
| | - Juan P. Angel
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey, United States of America
| | - Irving Estevez
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey, United States of America
| | - Nydia P. Chang
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey, United States of America
| | - Tsui-Wen Chou
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey, United States of America
| | - Micheal McCourt
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey, United States of America
| | - Colm Atkins
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey, United States of America
| | - Brian P. Daniels
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey, United States of America
| |
Collapse
|
11
|
Si Y, Zhang H, Zhou Z, Zhu X, Yang Y, Liu H, Zhang L, Cheng L, Wang K, Ye W, Lv X, Zhang X, Hou W, Zhao G, Lei Y, Zhang F, Ma H. RIPK3 promotes hantaviral replication by restricting JAK-STAT signaling without triggering necroptosis. Virol Sin 2023; 38:741-754. [PMID: 37633447 PMCID: PMC10590702 DOI: 10.1016/j.virs.2023.08.006] [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: 02/22/2023] [Accepted: 08/21/2023] [Indexed: 08/28/2023] Open
Abstract
Hantaan virus (HTNV) is a rodent-borne virus that causes hemorrhagic fever with renal syndrome (HFRS), resulting in a high mortality rate of 15%. Interferons (IFNs) play a critical role in the anti-hantaviral immune response, and IFN pretreatment efficiently restricts HTNV infection by triggering the expression of a series of IFN-stimulated genes (ISGs) through the Janus kinase-signal transducer and activator of transcription 1 (JAK-STAT) pathway. However, the tremendous amount of IFNs produced during late infection could not restrain HTNV replication, and the mechanism remains unclear. Here, we demonstrated that receptor-interacting protein kinase 3 (RIPK3), a crucial molecule that mediates necroptosis, was activated by HTNV and contributed to hantavirus evasion of IFN responses by inhibiting STAT1 phosphorylation. RNA-seq analysis revealed the upregulation of multiple cell death-related genes after HTNV infection, with RIPK3 identified as a key modulator of viral replication. RIPK3 ablation significantly enhanced ISGs expression and restrained HTNV replication, without affecting the expression of pattern recognition receptors (PRRs) or the production of type I IFNs. Conversely, exogenously expressed RIPK3 compromised the host's antiviral response and facilitated HTNV replication. RIPK3-/- mice also maintained a robust ability to clear HTNV with enhanced innate immune responses. Mechanistically, we found that RIPK3 could bind STAT1 and inhibit STAT1 phosphorylation dependent on the protein kinase domain (PKD) of RIPK3 but not its kinase activity. Overall, these observations demonstrated a noncanonical function of RIPK3 during viral infection and have elucidated a novel host innate immunity evasion strategy utilized by HTNV.
Collapse
Affiliation(s)
- Yue Si
- Department of Microbiology, School of Basic Medicine, Air Force Medical University, Xi'an, 710032, China
| | - Haijun Zhang
- Department of Neurology, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China; Center of Clinical Aerospace Medicine, School of Aerospace Medicine, Key Laboratory of Aerospace Medicine of Ministry of Education, Air Force Medical University, Xi'an, 710032, China
| | - Ziqing Zhou
- Department of Microbiology, School of Basic Medicine, Air Force Medical University, Xi'an, 710032, China
| | - Xudong Zhu
- Department of Microbiology, School of Basic Medicine, Air Force Medical University, Xi'an, 710032, China
| | - Yongheng Yang
- Department of Microbiology, School of Basic Medicine, Air Force Medical University, Xi'an, 710032, China
| | - He Liu
- Department of Microbiology, School of Basic Medicine, Air Force Medical University, Xi'an, 710032, China
| | - Liang Zhang
- Department of Microbiology, School of Basic Medicine, Air Force Medical University, Xi'an, 710032, China
| | - Linfeng Cheng
- Department of Microbiology, School of Basic Medicine, Air Force Medical University, Xi'an, 710032, China
| | - Kerong Wang
- Department of Microbiology, School of Basic Medicine, Air Force Medical University, Xi'an, 710032, China
| | - Wei Ye
- Department of Microbiology, School of Basic Medicine, Air Force Medical University, Xi'an, 710032, China
| | - Xin Lv
- Department of Microbiology, School of Basic Medicine, Air Force Medical University, Xi'an, 710032, China
| | - Xijing Zhang
- Department of Anesthesiology & Critical Care Medicine, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Wugang Hou
- Department of Anesthesiology & Critical Care Medicine, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China
| | - Gang Zhao
- Department of Neurology, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China; The College of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China
| | - Yingfeng Lei
- Department of Microbiology, School of Basic Medicine, Air Force Medical University, Xi'an, 710032, China.
| | - Fanglin Zhang
- Department of Microbiology, School of Basic Medicine, Air Force Medical University, Xi'an, 710032, China.
| | - Hongwei Ma
- Department of Microbiology, School of Basic Medicine, Air Force Medical University, Xi'an, 710032, China; Department of Anesthesiology & Critical Care Medicine, Xijing Hospital, Air Force Medical University, Xi'an, 710032, China.
| |
Collapse
|
12
|
Kwon EB, Kim SG, Kim YS, Kim B, Han SM, Lee HJ, Choi HM, Choi JG. Castanea crenata honey reduces influenza infection by activating the innate immune response. Front Immunol 2023; 14:1157506. [PMID: 37711616 PMCID: PMC10497975 DOI: 10.3389/fimmu.2023.1157506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 07/31/2023] [Indexed: 09/16/2023] Open
Abstract
Influenza is an acute respiratory disorder caused by the influenza virus and is associated with prolonged hospitalization and high mortality rates in older individuals and chronically ill patients. Vaccination is the most effective preventive strategy for ameliorating seasonal influenza. However, the vaccine is not fully effective in cases of antigenic mismatch with the viral strains circulating in the community. The emergence of resistance to antiviral drugs aggravates the situation. Therefore, developing new vaccines and antiviral drugs is essential. Castanea crenata honey (CH) is an extensively cultivated food worldwide and has been used as a nutritional supplement or herbal medicine. However, the potential anti-influenza properties of CH remain unexplored. In this study, the in vitro and in vivo antiviral effects of CH were assessed. CH significantly prevented influenza virus infection in mouse Raw264.7 macrophages. CH pretreatment inhibited the expression of the viral proteins M2, PA, and PB1 and enhanced the secretion of proinflammatory cytokines and type-I interferon (IFN)-related proteins in vitro. CH increased the expression of RIG-1, mitochondrial antiviral signaling (MAVS) protein, and IFN-inducible transmembrane protein, which interferes with virus replication. CH reduced body weight loss by 20.9%, increased survival by 60%, and decreased viral replication and inflammatory response in the lungs of influenza A virus-infected mice. Therefore, CH stimulates an antiviral response in murine macrophages and mice by preventing viral infection through the RIG-1-mediated MAVS pathway. Further investigation is warranted to understand the molecular mechanisms involved in the protective effects of CH on influenza virus infection.
Collapse
Affiliation(s)
- Eun-Bin Kwon
- Korean Medicine Application Center, Korea Institute of Oriental Medicine, Daegu, Republic of Korea
| | - Se-Gun Kim
- Department of Agricultural Biology, National Institute of Agricultural Sciences, Rural Development Administration, Wanju, Republic of Korea
| | - Young Soo Kim
- Korean Medicine Application Center, Korea Institute of Oriental Medicine, Daegu, Republic of Korea
| | - Buyun Kim
- Korean Medicine Application Center, Korea Institute of Oriental Medicine, Daegu, Republic of Korea
| | - Sang Mi Han
- Department of Agricultural Biology, National Institute of Agricultural Sciences, Rural Development Administration, Wanju, Republic of Korea
| | - Hye Jin Lee
- Department of Agricultural Biology, National Institute of Agricultural Sciences, Rural Development Administration, Wanju, Republic of Korea
| | - Hong Min Choi
- Department of Agricultural Biology, National Institute of Agricultural Sciences, Rural Development Administration, Wanju, Republic of Korea
| | - Jang-Gi Choi
- Korean Medicine Application Center, Korea Institute of Oriental Medicine, Daegu, Republic of Korea
| |
Collapse
|
13
|
Lindman M, Angel JP, Estevez I, Chang NP, Chou TW, McCourt M, Atkins C, Daniels BP. RIPK3 promotes brain region-specific interferon signaling and restriction of tick-borne flavivirus infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.23.525284. [PMID: 36747672 PMCID: PMC9900788 DOI: 10.1101/2023.01.23.525284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Innate immune signaling in the central nervous system (CNS) exhibits many remarkable specializations that vary across cell types and CNS regions. In the setting of neuroinvasive flavivirus infection, neurons employ the immunologic kinase receptor-interacting kinase 3 (RIPK3) to promote an antiviral transcriptional program, independently of the traditional function of this enzyme in promoting necroptotic cell death. However, while recent work has established roles for neuronal RIPK3 signaling in controlling mosquito-borne flavivirus infections, including West Nile virus and Zika virus, functions for RIPK3 signaling in the CNS during tick-borne flavivirus infection have not yet been explored. Here, we use a model of Langat virus (LGTV) encephalitis to show that RIPK3 signaling is specifically required in neurons of the cerebellum to control LGTV replication and restrict disease pathogenesis. This effect did not require the necroptotic executioner molecule mixed lineage kinase domain like protein (MLKL), a finding similar to previous observations in models of mosquito-borne flavivirus infection. However, control of LGTV infection required a unique, region-specific dependence on RIPK3 to promote expression of key antiviral interferon-stimulated genes (ISG) in the cerebellum. This RIPK3-mediated potentiation of ISG expression was associated with robust cell-intrinsic restriction of LGTV replication in cerebellar granule cell neurons. These findings further illuminate the complex roles of RIPK3 signaling in the coordination of neuroimmune responses to viral infection, as well as provide new insight into the mechanisms of region-specific innate immune signaling in the CNS.
Collapse
Affiliation(s)
- Marissa Lindman
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Juan P Angel
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Irving Estevez
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Nydia P Chang
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Tsui-Wen Chou
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Micheal McCourt
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Colm Atkins
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Brian P. Daniels
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
14
|
Sai K, Nakanishi A, Scofield KM, Tokarz DA, Linder KE, Cohen TJ, Ninomiya-Tsuji J. Aberrantly activated TAK1 links neuroinflammation and neuronal loss in Alzheimer's disease mouse models. J Cell Sci 2023; 136:jcs260102. [PMID: 36912451 PMCID: PMC10112982 DOI: 10.1242/jcs.260102] [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: 04/08/2022] [Accepted: 01/27/2023] [Indexed: 03/14/2023] Open
Abstract
Neuroinflammation is causally associated with Alzheimer's disease (AD) pathology. Reactive glia cells secrete various neurotoxic factors that impair neuronal homeostasis eventually leading to neuronal loss. Although the glial activation mechanism in AD has been relatively well studied, how it perturbs intraneuronal signaling, which ultimately leads to neuronal cell death, remains poorly understood. Here, we report that compound stimulation with the neurotoxic factors TNF and glutamate aberrantly activates neuronal TAK1 (also known as MAP3K7), which promotes the pathogenesis of AD in mouse models. Glutamate-induced Ca2+ influx shifts TNF signaling to hyper-activate TAK1 enzymatic activity through Ca2+/calmodulin-dependent protein kinase II, which leads to necroptotic cellular damage. Genetic ablation and pharmacological inhibition of TAK1 ameliorated AD-associated neuronal loss and cognitive impairment in the AD model mice. Our findings provide a molecular mechanism linking cytokines, Ca2+ signaling and neuronal necroptosis in AD.
Collapse
Affiliation(s)
- Kazuhito Sai
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695-7633, USA
| | - Aoi Nakanishi
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695-7633, USA
| | - Kimberly M. Scofield
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695-7633, USA
| | - Debra A. Tokarz
- Center for Human Health and the Environment, Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
| | - Keith E. Linder
- Center for Human Health and the Environment, Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
| | - Todd J. Cohen
- Department of Neurology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jun Ninomiya-Tsuji
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695-7633, USA
| |
Collapse
|
15
|
MLKL-Driven Inflammasome Activation and Caspase-8 Mediate Inflammatory Cell Death in Influenza A Virus Infection. mBio 2023; 14:e0011023. [PMID: 36852999 PMCID: PMC10127685 DOI: 10.1128/mbio.00110-23] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023] Open
Abstract
Influenza A virus (IAV) triggers multiple programmed cell death pathways, including MLKL-dependent necroptosis, caspase-8-dependent apoptosis, and caspase-1-dependent pyroptosis in myeloid cells. All three pathways share common upstream regulators, namely, ZBP1 and RIPK3. Yet, the molecular mechanism underlying IAV-induced inflammasome activation remains unclear. Here, we demonstrate that MLKL promotes inflammasome activation and IL-1β processing in IAV-infected macrophages. MLKL drives NLRP3 inflammasome activation through potassium efflux. In the absence of the MLKL-inflammasome axis, caspase-8 coordinates the maturation and secretion of IL-1β. MLKL alone is dispensable for host inflammatory responses to IAV in vivo. Taken together, MLKL and caspase-8 serve as redundant mechanisms by which to drive an inflammatory form of cell death in response to an IAV infection. IMPORTANCE Influenza A virus (IAV) induces multiple types of cell death, which play important roles in the host antiviral responses but can also cause unwanted inflammation and tissue damage. In this study, we dissect the interplay of cell death pathways and demonstrate that macrophages utilize redundant mechanisms to drive an inflammatory form of cell death upon IAV infection. MLKL, the executor of necroptosis, promotes inflammasome activation and pyroptotic cell death. When the MLKL-inflammasome axis is inhibited, cells divert to caspase-8-dependent inflammatory cell death. Our findings advance the current understanding of the innate immune response to IAV infection as well as broader contexts involving multifaceted cell death.
Collapse
|
16
|
Preston SP, Allison CC, Schaefer J, Clow W, Bader SM, Collard S, Forsyth WO, Clark MP, Garnham AL, Li-Wai-Suen CSN, Peiris T, Teale J, Mackiewicz L, Davidson S, Doerflinger M, Pellegrini M. A necroptosis-independent function of RIPK3 promotes immune dysfunction and prevents control of chronic LCMV infection. Cell Death Dis 2023; 14:123. [PMID: 36792599 PMCID: PMC9931694 DOI: 10.1038/s41419-023-05635-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/13/2022] [Accepted: 01/31/2023] [Indexed: 02/17/2023]
Abstract
Necroptosis is a lytic and inflammatory form of cell death that is highly constrained to mitigate detrimental collateral tissue damage and impaired immunity. These constraints make it difficult to define the relevance of necroptosis in diseases such as chronic and persistent viral infections and within individual organ systems. The role of necroptotic signalling is further complicated because proteins essential to this pathway, such as receptor interacting protein kinase 3 (RIPK3) and mixed lineage kinase domain-like (MLKL), have been implicated in roles outside of necroptotic signalling. We sought to address this issue by individually defining the role of RIPK3 and MLKL in chronic lymphocytic choriomeningitis virus (LCMV) infection. We investigated if necroptosis contributes to the death of LCMV-specific CD8+ T cells or virally infected target cells during infection. We provide evidence showing that necroptosis was redundant in the pathogenesis of acute forms of LCMV (Armstrong strain) and the early stages of chronic (Docile strain) LCMV infection in vivo. The number of immune cells, their specificity and reactivity towards viral antigens and viral loads are not altered in the absence of either MLKL or RIPK3 during acute and during the early stages of chronic LCMV infection. However, we identified that RIPK3 promotes immune dysfunction and prevents control of infection at later stages of chronic LCMV disease. This was not phenocopied by the loss of MLKL indicating that the phenotype was driven by a necroptosis-independent function of RIPK3. We provide evidence that RIPK3 signaling evoked a dysregulated type 1 interferone response which we linked to an impaired antiviral immune response and abrogated clearance of chronic LCMV infection.
Collapse
Affiliation(s)
- Simon P. Preston
- grid.1042.70000 0004 0432 4889Walter and Eliza Hall Institute of Medical Research, Parkville, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, The University of Melbourne, Parkville, VIC Australia ,SYNthesis Research, Bio21 Institute, Parkville, VIC Australia
| | - Cody C. Allison
- grid.1042.70000 0004 0432 4889Walter and Eliza Hall Institute of Medical Research, Parkville, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, The University of Melbourne, Parkville, VIC Australia
| | - Jan Schaefer
- grid.1042.70000 0004 0432 4889Walter and Eliza Hall Institute of Medical Research, Parkville, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, The University of Melbourne, Parkville, VIC Australia
| | - William Clow
- grid.1042.70000 0004 0432 4889Walter and Eliza Hall Institute of Medical Research, Parkville, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, The University of Melbourne, Parkville, VIC Australia
| | - Stefanie M. Bader
- grid.1042.70000 0004 0432 4889Walter and Eliza Hall Institute of Medical Research, Parkville, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, The University of Melbourne, Parkville, VIC Australia
| | - Sophie Collard
- grid.1042.70000 0004 0432 4889Walter and Eliza Hall Institute of Medical Research, Parkville, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, The University of Melbourne, Parkville, VIC Australia
| | - Wasan O. Forsyth
- grid.1042.70000 0004 0432 4889Walter and Eliza Hall Institute of Medical Research, Parkville, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, The University of Melbourne, Parkville, VIC Australia
| | - Michelle P. Clark
- grid.1042.70000 0004 0432 4889Walter and Eliza Hall Institute of Medical Research, Parkville, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, The University of Melbourne, Parkville, VIC Australia
| | - Alexandra L. Garnham
- grid.1042.70000 0004 0432 4889Walter and Eliza Hall Institute of Medical Research, Parkville, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, The University of Melbourne, Parkville, VIC Australia
| | - Connie S. N. Li-Wai-Suen
- grid.1042.70000 0004 0432 4889Walter and Eliza Hall Institute of Medical Research, Parkville, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, The University of Melbourne, Parkville, VIC Australia
| | - Thanushi Peiris
- grid.1042.70000 0004 0432 4889Walter and Eliza Hall Institute of Medical Research, Parkville, VIC Australia
| | - Jack Teale
- grid.1042.70000 0004 0432 4889Walter and Eliza Hall Institute of Medical Research, Parkville, VIC Australia
| | - Liana Mackiewicz
- grid.1042.70000 0004 0432 4889Walter and Eliza Hall Institute of Medical Research, Parkville, VIC Australia
| | - Sophia Davidson
- grid.1042.70000 0004 0432 4889Walter and Eliza Hall Institute of Medical Research, Parkville, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, The University of Melbourne, Parkville, VIC Australia
| | - Marcel Doerflinger
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia. .,Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia.
| | - Marc Pellegrini
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia. .,Department of Medical Biology, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
17
|
Pernet E, Sun S, Sarden N, Gona S, Nguyen A, Khan N, Mawhinney M, Tran KA, Chronopoulos J, Amberkar D, Sadeghi M, Grant A, Wali S, Prevel R, Ding J, Martin JG, Thanabalasuriar A, Yipp BG, Barreiro LB, Divangahi M. Neonatal imprinting of alveolar macrophages via neutrophil-derived 12-HETE. Nature 2023; 614:530-538. [PMID: 36599368 PMCID: PMC9945843 DOI: 10.1038/s41586-022-05660-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 12/14/2022] [Indexed: 01/06/2023]
Abstract
Resident-tissue macrophages (RTMs) arise from embryonic precursors1,2, yet the developmental signals that shape their longevity remain largely unknown. Here we demonstrate in mice genetically deficient in 12-lipoxygenase and 15-lipoxygenase (Alox15-/- mice) that neonatal neutrophil-derived 12-HETE is required for self-renewal and maintenance of alveolar macrophages (AMs) during lung development. Although the seeding and differentiation of AM progenitors remained intact, the absence of 12-HETE led to a significant reduction in AMs in adult lungs and enhanced senescence owing to increased prostaglandin E2 production. A compromised AM compartment resulted in increased susceptibility to acute lung injury induced by lipopolysaccharide and to pulmonary infections with influenza A virus or SARS-CoV-2. Our results highlight the complexity of prenatal RTM programming and reveal their dependency on in trans eicosanoid production by neutrophils for lifelong self-renewal.
Collapse
Affiliation(s)
- Erwan Pernet
- McGill University Health Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada.
| | - Sarah Sun
- Department of Medicine, Section of Genetic Medicine, University of Chicago, Chicago, IL, USA
| | - Nicole Sarden
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases and Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Saideep Gona
- Department of Medicine, Section of Genetic Medicine, University of Chicago, Chicago, IL, USA
| | - Angela Nguyen
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases and Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Nargis Khan
- McGill University Health Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
| | - Martin Mawhinney
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Kim A Tran
- McGill University Health Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
| | - Julia Chronopoulos
- McGill University Health Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
| | - Dnyandeo Amberkar
- McGill University Health Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
| | - Mina Sadeghi
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
| | - Alexandre Grant
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
| | - Shradha Wali
- McGill University Health Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
| | - Renaud Prevel
- McGill University Health Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
| | - Jun Ding
- McGill University Health Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
| | - James G Martin
- McGill University Health Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
| | - Ajitha Thanabalasuriar
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Bryan G Yipp
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases and Department of Critical Care, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Luis B Barreiro
- Department of Medicine, Section of Genetic Medicine, University of Chicago, Chicago, IL, USA
- Department of Genetics, CHU Sainte-Justine Research Center, Montreal, Quebec, Canada
| | - Maziar Divangahi
- McGill University Health Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada.
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada.
- Department of Pathology, McGill University, Montreal, Quebec, Canada.
- McGill International TB Centre, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
18
|
Thomas PG, Shubina M, Balachandran S. ZBP1/DAI-Dependent Cell Death Pathways in Influenza A Virus Immunity and Pathogenesis. Curr Top Microbiol Immunol 2023; 442:41-63. [PMID: 31970498 DOI: 10.1007/82_2019_190] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Influenza A viruses (IAV) are members of the Orthomyxoviridae family of negative-sense RNA viruses. The greatest diversity of IAV strains is found in aquatic birds, but a subset of strains infects other avian as well as mammalian species, including humans. In aquatic birds, infection is largely restricted to the gastrointestinal tract and spread is through feces, while in humans and other mammals, respiratory epithelial cells are the primary sites supporting productive replication and transmission. IAV triggers the death of most cell types in which it replicates, both in culture and in vivo. When well controlled, such cell death is considered an effective host defense mechanism that eliminates infected cells and limits virus spread. Unchecked or inopportune cell death also results in immunopathology. In this chapter, we discuss the impact of cell death in restricting virus spread, supporting the adaptive immune response and driving pathogenesis in the mammalian respiratory tract. Recent studies have begun to shed light on the signaling pathways underlying IAV-activated cell death. These pathways, initiated by the pathogen sensor protein ZBP1 (also called DAI and DLM1), cause infected cells to undergo apoptosis, necroptosis, and pyroptosis. We outline mechanisms of ZBP1-mediated cell death signaling following IAV infection.
Collapse
Affiliation(s)
- Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, MS 351, 262 Danny Thomas Place, 38105, Memphis, TN, USA.
| | - Maria Shubina
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Room 224 Reimann Building, 333 Cottman Ave., 19111, Philadelphia, PA, USA
| | - Siddharth Balachandran
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Room 224 Reimann Building, 333 Cottman Ave., 19111, Philadelphia, PA, USA.
| |
Collapse
|
19
|
Sabikunnahar B, Lahue KG, Asarian L, Fang Q, McGill MM, Haynes L, Teuscher C, Krementsov DN. Sex differences in susceptibility to influenza A virus infection depend on host genotype. PLoS One 2022; 17:e0273050. [PMID: 36112601 PMCID: PMC9481031 DOI: 10.1371/journal.pone.0273050] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 08/01/2022] [Indexed: 11/19/2022] Open
Abstract
Infection with the respiratory pathogen influenza A virus (IAV) causes significant morbidity and mortality each year. While host genotype is thought to contribute to severity of disease, naturally occurring genetic determinants remain mostly unknown. Moreover, more severe disease is seen in women compared with men, but genetic mechanisms underlying this sex difference remain obscure. Here, using IAV infection in a mouse model of naturally selected genetic diversity, namely C57BL6/J (B6) mice carrying chromosomes (Chr) derived from the wild-derived and genetically divergent PWD/PhJ (PWD) mouse strain (B6.ChrPWD consomic mice), we examined the effects of genotype and sex on severity of IAV-induced disease. Compared with B6, parental PWD mice were completely protected from IAV-induced disease, a phenotype that was fully recapitulated in the B6.Chr16PWD strain carrying the PWD-derived allele of Mx1. In contrast, several other consomic strains, including B6.Chr3PWD and B6.Chr5PWD, demonstrated greatly increased susceptibility. Notably, B6.Chr5PWD and B6.ChrX.3PWD strains, the latter carrying the distal one-third of ChrX from PWD, exhibited increased morbidity and mortality specifically in male but not female mice. Follow up analyses focused on B6 and B6.ChrX.3PWD strains demonstrated moderately elevated viral load in B6.ChrX3PWD male, but not female mice. Transcriptional profiling demonstrated genotype- and sex-specific gene expression profiles in the infected lung, with male B6.ChrX.3 mice exhibiting the most significant changes, including upregulation of a proinflammatory gene expression program associated with myeloid cells, and altered sex-biased expression of several X-linked genes that represent positional candidates, including Tlr13 and Slc25a53. Taken together, our results identify novel loci on autosomes and the X chromosome regulating IAV susceptibility and demonstrate that sex differences in IAV susceptibility are genotype-dependent, suggesting that future genetic association studies need to consider sex as a covariate.
Collapse
Affiliation(s)
- Bristy Sabikunnahar
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States of America
| | - Karolyn G. Lahue
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States of America
| | - Loredana Asarian
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States of America
| | - Qian Fang
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States of America
| | - Mahalia M. McGill
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States of America
| | - Laura Haynes
- Department of Immunology, University of Connecticut School of Medicine, UConn Center on Aging, Farmington, CT, United States of America
| | - Cory Teuscher
- Department of Medicine, University of Vermont, Burlington, VT, United States of America
- Department of Pathology, University of Vermont, Burlington, VT, United States of America
| | - Dimitry N. Krementsov
- Department of Biomedical and Health Sciences, University of Vermont, Burlington, VT, United States of America
- * E-mail:
| |
Collapse
|
20
|
Zhang G, Wang J, Zhao Z, Xin T, Fan X, Shen Q, Raheem A, Lee CR, Jiang H, Ding J. Regulated necrosis, a proinflammatory cell death, potentially counteracts pathogenic infections. Cell Death Dis 2022; 13:637. [PMID: 35869043 PMCID: PMC9307826 DOI: 10.1038/s41419-022-05066-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/29/2022] [Accepted: 07/04/2022] [Indexed: 02/07/2023]
Abstract
Since the discovery of cell apoptosis, other gene-regulated cell deaths are gradually appreciated, including pyroptosis, ferroptosis, and necroptosis. Necroptosis is, so far, one of the best-characterized regulated necrosis. In response to diverse stimuli (death receptor or toll-like receptor stimulation, pathogenic infection, or other factors), necroptosis is initiated and precisely regulated by the receptor-interacting protein kinase 3 (RIPK3) with the involvement of its partners (RIPK1, TRIF, DAI, or others), ultimately leading to the activation of its downstream substrate, mixed lineage kinase domain-like (MLKL). Necroptosis plays a significant role in the host's defense against pathogenic infections. Although much has been recognized regarding modulatory mechanisms of necroptosis during pathogenic infection, the exact role of necroptosis at different stages of infectious diseases is still being unveiled, e.g., how and when pathogens utilize or evade necroptosis to facilitate their invasion and how hosts manipulate necroptosis to counteract these detrimental effects brought by pathogenic infections and further eliminate the encroaching pathogens. In this review, we summarize and discuss the recent progress in the role of necroptosis during a series of viral, bacterial, and parasitic infections with zoonotic potentials, aiming to provide references and directions for the prevention and control of infectious diseases of both human and animals.
Collapse
Affiliation(s)
- Guangzhi Zhang
- grid.464332.4Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing, 100193 China
| | - Jinyong Wang
- grid.508381.70000 0004 0647 272XShenzhen Bay Laboratory, Institute of Infectious Diseases, Shenzhen, 518000 China ,grid.258164.c0000 0004 1790 3548Institute of Respiratory Diseases, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, Shenzhen, 518020 Guangdong China
| | - Zhanran Zhao
- grid.47840.3f0000 0001 2181 7878Department of Molecular and Cell Biology and Cancer Research Laboratory, University of California, Berkeley, CA 94720-3200 USA
| | - Ting Xin
- grid.464332.4Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing, 100193 China
| | - Xuezheng Fan
- grid.464332.4Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing, 100193 China
| | - Qingchun Shen
- grid.464332.4Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing, 100193 China
| | - Abdul Raheem
- grid.464332.4Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing, 100193 China ,grid.35155.370000 0004 1790 4137Present Address: Huazhong Agricultural University, Wuhan, China
| | - Chae Rhim Lee
- grid.47840.3f0000 0001 2181 7878Department of Molecular and Cell Biology and Cancer Research Laboratory, University of California, Berkeley, CA 94720-3200 USA ,grid.266093.80000 0001 0668 7243Present Address: University of California, Irvine, CA USA
| | - Hui Jiang
- grid.464332.4Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing, 100193 China
| | - Jiabo Ding
- grid.464332.4Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Beijing, 100193 China
| |
Collapse
|
21
|
Guo YL, Gurung C, Fendereski M, Huang F. Dicer and PKR as Novel Regulators of Embryonic Stem Cell Fate and Antiviral Innate Immunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:2259-2266. [PMID: 35577384 PMCID: PMC9179006 DOI: 10.4049/jimmunol.2200042] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/21/2022] [Indexed: 05/17/2023]
Abstract
Embryonic stem cells (ESCs) represent a unique cell population in the blastocyst stage embryo. They have been intensively studied as a promising cell source for regenerative medicine. Recent studies have revealed that both human and mouse ESCs are deficient in expressing IFNs and have attenuated inflammatory responses. Apparently, the ability to express IFNs and respond to certain inflammatory cytokines is not "innate" to ESCs but rather is developmentally acquired by somatic cells during differentiation. Accumulating evidence supports a hypothesis that the attenuated innate immune response may serve as a protective mechanism allowing ESCs to avoid immunological cytotoxicity. This review describes our current understanding of the molecular basis that shapes the immune properties of ESCs. We highlight the recent findings on Dicer and dsRNA-activated protein kinase R as novel regulators of ESC fate and antiviral immunity and discuss how ESCs use alternative mechanisms to accommodate their stem cell properties.
Collapse
Affiliation(s)
- Yan-Lin Guo
- Cell and Molecular Biology Program, University of Southern Mississippi, Hattiesburg, MS; and
| | - Chandan Gurung
- Cell and Molecular Biology Program, University of Southern Mississippi, Hattiesburg, MS; and
| | - Mona Fendereski
- Cell and Molecular Biology Program, University of Southern Mississippi, Hattiesburg, MS; and
| | - Faqing Huang
- Chemistry and Biochemistry Program, University of Southern Mississippi, Hattiesburg, MS
| |
Collapse
|
22
|
Xin Y, Chen S, Tang K, Wu Y, Guo Y. Identification of Nifurtimox and Chrysin as Anti-Influenza Virus Agents by Clinical Transcriptome Signature Reversion. Int J Mol Sci 2022; 23:ijms23042372. [PMID: 35216485 PMCID: PMC8876279 DOI: 10.3390/ijms23042372] [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: 01/14/2022] [Revised: 02/12/2022] [Accepted: 02/18/2022] [Indexed: 12/28/2022] Open
Abstract
The rapid development in the field of transcriptomics provides remarkable biomedical insights for drug discovery. In this study, a transcriptome signature reversal approach was conducted to identify the agents against influenza A virus (IAV) infection through dissecting gene expression changes in response to disease or compounds’ perturbations. Two compounds, nifurtimox and chrysin, were identified by a modified Kolmogorov–Smirnov test statistic based on the transcriptional signatures from 81 IAV-infected patients and the gene expression profiles of 1309 compounds. Their activities were verified in vitro with half maximal effective concentrations (EC50s) from 9.1 to 19.1 μM against H1N1 or H3N2. It also suggested that the two compounds interfered with multiple sessions in IAV infection by reversing the expression of 28 IAV informative genes. Through network-based analysis of the 28 reversed IAV informative genes, a strong synergistic effect of the two compounds was revealed, which was confirmed in vitro. By using the transcriptome signature reversion (TSR) on clinical datasets, this study provides an efficient scheme for the discovery of drugs targeting multiple host factors regarding clinical signs and symptoms, which may also confer an opportunity for decelerating drug-resistant variant emergence.
Collapse
Affiliation(s)
- Yijing Xin
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (Y.X.); (S.C.); (K.T.); (Y.W.)
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Shubing Chen
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (Y.X.); (S.C.); (K.T.); (Y.W.)
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Ke Tang
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (Y.X.); (S.C.); (K.T.); (Y.W.)
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - You Wu
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (Y.X.); (S.C.); (K.T.); (Y.W.)
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Ying Guo
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (Y.X.); (S.C.); (K.T.); (Y.W.)
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Correspondence: ; Tel.: +86-010-63161716
| |
Collapse
|
23
|
Chronopoulos J, Martin JG, Divangahi M. Transplacental and Breast Milk Transfer of IgG1 Are Both Required for Prolonged Protection of Offspring Against Influenza A Infection. Front Immunol 2022; 13:823207. [PMID: 35185914 PMCID: PMC8850295 DOI: 10.3389/fimmu.2022.823207] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 01/14/2022] [Indexed: 11/29/2022] Open
Abstract
The immune system during pregnancy teeters between maintaining fetal tolerance and providing protection against pathogens. Due to this delicate balance, pregnant women and their offspring often have increased susceptibilities to infection. During the first year of life, infant immunity against infection is mainly mediated via passively transferred maternal antibodies. However, our understanding of the route of transfer of the maternal antibodies for conferring protection to influenza A virus (IAV) infection in offspring is incomplete. Here we have demonstrated that offspring from IAV-infected mice were significantly protected against IAV infection. This remarkable increase in survival is mediated via the elevated maternal serum IgG1. By cross-fostering, we further showed that this enhanced host resistance was only achieved in mice born to and nursed by IAV-infected mothers. Collectively, our data suggest that the prolonged protection of offspring against IAV infection requires maternal IgG1 from both the placenta and breast milk.
Collapse
Affiliation(s)
- Julia Chronopoulos
- Meakins-Christie Laboratories, Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - James G. Martin
- Meakins-Christie Laboratories, Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Maziar Divangahi
- Meakins-Christie Laboratories, Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
- Department of Microbiology and Immunology and Department of Pathology, Montreal, QC, Canada
- McGill International TB Center, Montreal, QC, Canada
| |
Collapse
|
24
|
BCG vaccination provides protection against IAV but not SARS-CoV-2. Cell Rep 2022; 38:110502. [PMID: 35235831 PMCID: PMC8858710 DOI: 10.1016/j.celrep.2022.110502] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 11/30/2021] [Accepted: 02/15/2022] [Indexed: 11/25/2022] Open
Abstract
Since the vast majority of species solely rely on innate immunity for host defense, it stands to reason that a critical evolutionary trait like immunological memory evolved in this primitive branch of our immune system. There is ample evidence that vaccines such as bacillus Calmette-Guérin (BCG) induce protective innate immune memory responses (trained immunity) against heterologous pathogens. Here we show that while BCG vaccination significantly reduces morbidity and mortality against influenza A virus (IAV), it fails to provide protection against severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). In contrast to IAV, SARS-CoV-2 infection leads to unique pulmonary vasculature damage facilitating viral dissemination to other organs, including the bone marrow (BM), a central site for BCG-mediated trained immunity. Finally, monocytes from BCG-vaccinated individuals mount an efficient cytokine response to IAV infection, while this response is minimal following SARS-CoV-2. Collectively, our data suggest that the protective capacity of BCG vaccination is contingent on viral pathogenesis and tissue tropism.
Collapse
|
25
|
Liu X, Xie X, Ren Y, Shao Z, Zhang N, Li L, Ding X, Zhang L. The role of necroptosis in disease and treatment. MedComm (Beijing) 2021; 2:730-755. [PMID: 34977874 PMCID: PMC8706757 DOI: 10.1002/mco2.108] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 12/11/2022] Open
Abstract
Necroptosis, a distinctive type of programmed cell death different from apoptosis or necrosis, triggered by a series of death receptors such as tumor necrosis factor receptor 1 (TNFR1), TNFR2, and Fas. In case that apoptosis process is blocked, necroptosis pathway is initiated with the activation of three key downstream mediators which are receptor-interacting serine/threonine protein kinase 1 (RIPK1), RIPK3, and mixed lineage kinase domain-like protein (MLKL). The whole process eventually leads to destruction of the cell membrane integrity, swelling of organelles, and severe inflammation. Over the past decade, necroptosis has been found widely involved in life process of human beings and animals. In this review, we attempt to explore the therapeutic prospects of necroptosis regulators by describing its molecular mechanism and the role it played in pathological condition and tissue homeostasis, and to summarize the research and clinical applications of corresponding regulators including small molecule inhibitors, chemicals, Chinese herbal extracts, and biological agents in the treatment of various diseases.
Collapse
Affiliation(s)
- Xiaoxiao Liu
- Department of Radiation OncologyAffiliated Hospital of Xuzhou Medical UniversityXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
- Jiangsu Center for the Collaboration and Innovation of Cancer BiotherapyCancer InstituteXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
| | - Xin Xie
- Department of Radiation OncologyAffiliated Hospital of Xuzhou Medical UniversityXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
- Jiangsu Center for the Collaboration and Innovation of Cancer BiotherapyCancer InstituteXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
| | - Yuanyuan Ren
- Department of Radiation OncologyAffiliated Hospital of Xuzhou Medical UniversityXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
- Jiangsu Center for the Collaboration and Innovation of Cancer BiotherapyCancer InstituteXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
| | - Zhiying Shao
- Department of Radiation OncologyAffiliated Hospital of Xuzhou Medical UniversityXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
- Jiangsu Center for the Collaboration and Innovation of Cancer BiotherapyCancer InstituteXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
- Cancer InstituteXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
| | - Nie Zhang
- Department of Radiation OncologyAffiliated Hospital of Xuzhou Medical UniversityXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
- Jiangsu Center for the Collaboration and Innovation of Cancer BiotherapyCancer InstituteXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
| | - Liantao Li
- Department of Radiation OncologyAffiliated Hospital of Xuzhou Medical UniversityXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
- Jiangsu Center for the Collaboration and Innovation of Cancer BiotherapyCancer InstituteXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
| | - Xin Ding
- Department of Radiation OncologyAffiliated Hospital of Xuzhou Medical UniversityXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
- Jiangsu Center for the Collaboration and Innovation of Cancer BiotherapyCancer InstituteXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
| | - Longzhen Zhang
- Department of Radiation OncologyAffiliated Hospital of Xuzhou Medical UniversityXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
- Jiangsu Center for the Collaboration and Innovation of Cancer BiotherapyCancer InstituteXuzhou Medical UniversityXuzhouJiangsu ProvinceP. R. China
| |
Collapse
|
26
|
Wang L, Zhou L, Zhou Y, Liu L, Jiang W, Zhang H, Liu H. Necroptosis in Pulmonary Diseases: A New Therapeutic Target. Front Pharmacol 2021; 12:737129. [PMID: 34594225 PMCID: PMC8476758 DOI: 10.3389/fphar.2021.737129] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 09/01/2021] [Indexed: 12/15/2022] Open
Abstract
In the past decades, apoptosis has been the most well-studied regulated cell death (RCD) that has essential functions in tissue homeostasis throughout life. However, a novel form of RCD called necroptosis, which requires receptor-interacting protein kinase-3 (RIPK3) and mixed-lineage kinase domain-like pseudokinase (MLKL), has recently been receiving increasing scientific attention. The phosphorylation of RIPK3 enables the recruitment and phosphorylation of MLKL, which oligomerizes and translocates to the plasma membranes, ultimately leading to plasma membrane rupture and cell death. Although apoptosis elicits no inflammatory responses, necroptosis triggers inflammation or causes an innate immune response to protect the body through the release of damage-associated molecular patterns (DAMPs). Increasing evidence now suggests that necroptosis is implicated in the pathogenesis of several human diseases such as systemic inflammation, respiratory diseases, cardiovascular diseases, neurodegenerative diseases, neurological diseases, and cancer. This review summarizes the emerging insights of necroptosis and its contribution toward the pathogenesis of lung diseases.
Collapse
Affiliation(s)
- Lingling Wang
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Zhou
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuhao Zhou
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lu Liu
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weiling Jiang
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huojun Zhang
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huiguo Liu
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
27
|
Dai Y, Cao Y, Chen Z, Huang J, Xiao J, Zou J, Feng H. RIPK3 collaborates with RIPK1 to inhibit MAVS-mediated signaling during black carp antiviral innate immunity. FISH & SHELLFISH IMMUNOLOGY 2021; 115:142-149. [PMID: 34147612 DOI: 10.1016/j.fsi.2021.06.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 06/06/2021] [Accepted: 06/14/2021] [Indexed: 06/12/2023]
Abstract
Both the activation and attenuation of MAVS/IFN signaling are critical for host defensing against viral infection and thus lead to an elaborate regulation of MAVS-mediated signaling. However, the regulatory mechanisms concerning MAVS/IFN signaling in teleost fish are not well understood. RIPK3 has been identified as a key regulator of necroptosis, apoptosis, and inflammatory signaling in human and mammals. Here we report the identification of the RIPK3 homologue from black carp Mylopharyngodon piceus (bcRIPK3) and describe its role in regulating MAVS/IFN signaling. qPCR results demonstrated that bcRIPK3 was transcriptionally activated in response to poly (I:C) or LPS stimulation. Immunoblot assay and immunofluorescent staining assay showed that bcRIPK3 was a cytosolic protein with molecular weights of 47 kDa. Like its mammalian counterparts, bcRIPK3 exhibited a conserved function in inducing cell death. The reporter assay and plaque assay showed that overexpression of bcRIPK3 restricted bcMAVS-activated transcription of the interferon promoters of black carp and zebrafish, and suppressed bcMAVS-mediated antiviral activity. Notably, EPC cells co-expressing bcRIPK3, bcRIPK1 and bcMAVS presented much attenuated antiviral activity than the cells co-expressing bcRIPK3 and bcMAVS; and the subsequent co-IP assay identified the interaction between bcRIPK3 and bcRIPK1. Our findings collectively elucidate for the first time in teleost that black carp RIPK3 interacts with RIPK1 to inhibit MAVS-mediated antiviral signaling.
Collapse
Affiliation(s)
- Yuhan Dai
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Yingyi Cao
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Zhaoyuan Chen
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Jiayi Huang
- 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.
| | - Jun Zou
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Hao Feng
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China.
| |
Collapse
|
28
|
Viral dosing of influenza A infection reveals involvement of RIPK3 and FADD, but not MLKL. Cell Death Dis 2021; 12:471. [PMID: 33976111 PMCID: PMC8113499 DOI: 10.1038/s41419-021-03746-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 02/03/2023]
Abstract
RIPK3 was reported to play an important role in the protection against influenza A virus (IAV) in vivo. Here we show that the requirement of RIPK3 for protection against IAV infection in vivo is only apparent within a limited dose range of IAV challenge. We found that this protective outcome is independent from RIPK3 kinase activity and from MLKL. This shows that platform function of RIPK3 rather than its kinase activity is required for protection, suggesting that a RIPK3 function independent of necroptosis is implicated. In line with this finding, we show that FADD-dependent apoptosis has a crucial additional effect in protection against IAV infection. Altogether, we show that RIPK3 contributes to protection against IAV in a narrow challenge dose range by a mechanism that is independent of its kinase activity and its capacity to induce necroptosis.
Collapse
|
29
|
Zhang N, Ma Y, Tian Y, Zhou Y, Tang Y, Hu S. Downregulation of microRNA‑221 facilitates H1N1 influenza A virus replication through suppression of type‑IFN response by targeting the SOCS1/NF‑κB pathway. Mol Med Rep 2021; 24:497. [PMID: 33955508 PMCID: PMC8127060 DOI: 10.3892/mmr.2021.12136] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 03/10/2021] [Indexed: 12/23/2022] Open
Abstract
Accumulating data has indicated that host microRNAs (miRNAs/miRs) play essential roles in innate immune responses to viral infection; however, the roles and the underlying mechanisms of miRNAs in influenza A virus (IAV) replication remain unclear. The present study examined on the effects of miRNAs on hemagglutinin (H)1 neuraminidase (N)1 replication and antiviral innate immunity. Using a microarray assay, the expression profiles of miRNA molecules in IAV-infected A549 cells were analyzed. The results indicated that miR-221 was significantly downregulated in IAV-infected A549 cells. It was also observed that IAV infection decreased the expression levels of miR-221 in A549 cells in a dose- and time-dependent manner. Functionally, upregulation of miR-221 repressed IAV replication, whereas knockdown of miR-221 had an opposite effect. Subsequently, it was demonstrated that miR-221 overexpression could enhance IAV-triggered IFN-α and IFN-β production and IFN-stimulated gene expression levels, while miR-221-knockdown had the opposite effect. Target prediction and dual luciferase assays indicated that suppressor of cytokine signaling 1 (SOCS1) was a direct target of miR-221 in A549 cells. Furthermore, knockdown of SOCS1 efficiently abrogated the influences caused by miR-221 inhibition on IAV replication and the type-I IFN response. It was also found that the miR-221 positively regulated NF-κB activation in IAV-infected A549 cells. Taken together, these data suggested that miR-221-downregulation promotes IAV replication by suppressing type-I IFN response through targeting SOCS1/NF-κB pathway. These findings suggest that miR-221 may serve as a novel potential therapeutic target for IAV treatment.
Collapse
Affiliation(s)
- Nali Zhang
- Department of Respiratory Medicine and Intensive Care Unit, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, P.R. China
| | - Yuan Ma
- Department of Respiratory Medicine and Intensive Care Unit, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, P.R. China
| | - Yuheng Tian
- Department of Respiratory Medicine and Intensive Care Unit, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, P.R. China
| | - Yafei Zhou
- Department of Respiratory Medicine and Intensive Care Unit, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, P.R. China
| | - Yuhua Tang
- Department of Respiratory Medicine and Intensive Care Unit, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, P.R. China
| | - Shaobo Hu
- Department of Respiratory Medicine and Intensive Care Unit, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, P.R. China
| |
Collapse
|
30
|
Hu X, Zhao Y, Yang Y, Gong W, Sun X, Yang L, Zhang Q, Jin M. Akkermansia muciniphila Improves Host Defense Against Influenza Virus Infection. Front Microbiol 2021; 11:586476. [PMID: 33603716 PMCID: PMC7884316 DOI: 10.3389/fmicb.2020.586476] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 12/21/2020] [Indexed: 12/24/2022] Open
Abstract
Influenza virus infection can alter the composition of the gut microbiota, while its pathogenicity can, in turn, be highly influenced by the gut microbiota. However, the details underlying these associations remain to be determined. The H7N9 influenza virus is an emerging zoonotic pathogen which has caused the death of 616 humans and has incurred huge losses in the poultry industry. Here, we investigated the effects of infection with highly pathogenic H7N9 on gut microbiota and determined potential anti-influenza microbes. 16S rRNA sequencing results show that H7N9 infection alters the mouse gut microbiota by promoting the growth of Akkermansia, Ruminococcus 1, and Ruminococcaceae UCG-010, and reducing the abundance of Rikenellaceae RC9 gut group and Lachnoclostridium. Although the abundance of Akkermansia muciniphila is positively related to H7N9 infection, the oral administration of cultures, especially of pasteurized A. muciniphila, can significantly reduce weight loss and mortality caused by H7N9 infection in mice. Furthermore, oral administration of live or pasteurized A. muciniphila significantly reduces pulmonary viral titers and the levels IL-1β and IL-6 but enhances the levels of IFN-β, IFN-γ, and IL-10 in H7N9-infected mice, suggesting that the anti-influenza role of A. muciniphila is due to its anti-inflammatory and immunoregulatory properties. Taken together, we showed that the changes in the gut microbiota are associated with H7N9 infection and demonstrated the anti-influenza role of A. muciniphila, which enriches current knowledge about how specific gut bacterial strains protect against influenza infection and suggests a potential anti-influenza probiotic.
Collapse
Affiliation(s)
- Xiaotong Hu
- Unit of Animal Infectious Diseases, State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ya Zhao
- Unit of Animal Infectious Diseases, State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yong Yang
- Unit of Animal Infectious Diseases, State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Wenxiao Gong
- Unit of Animal Infectious Diseases, State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xiaomei Sun
- Unit of Animal Infectious Diseases, State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Li Yang
- Unit of Animal Infectious Diseases, State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Qiang Zhang
- Unit of Animal Infectious Diseases, State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China
| | - Meilin Jin
- Unit of Animal Infectious Diseases, State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.,Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, China.,Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, Wuhan, China
| |
Collapse
|
31
|
Lin YH, Platt MP, Gilley RP, Brown D, Dube PH, Yu Y, Gonzalez-Juarbe N. Influenza Causes MLKL-Driven Cardiac Proteome Remodeling During Convalescence. Circ Res 2021; 128:570-584. [PMID: 33501852 DOI: 10.1161/circresaha.120.318511] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
RATIONALE Patients with and without cardiovascular diseases have been shown to be at risk of influenza-mediated cardiac complications. Recent clinical reports support the notion of a direct link between laboratory-confirmed influenza virus infections and adverse cardiac events. OBJECTIVE Define the molecular mechanisms underlying influenza virus-induced cardiac pathogenesis after resolution of pulmonary infection and the role of necroptosis in this process. METHODS AND RESULTS Hearts from wild-type and necroptosis-deficient (MLKL [mixed lineage kinase domain-like protein]-KO) mice were dissected 12 days after initial influenza A virus (IAV) infection when viral titers were undetectable in the lungs. Immunofluorescence microscopy and plaque assays showed presence of viable IAV particles in the myocardium without generation of interferon responses. Global proteome and phosphoproteome analyses using high-resolution accurate mass-based LC-MS/MS and label-free quantitation showed that the global proteome as well as the phosphoproteome profiles were significantly altered in IAV-infected mouse hearts in a strain-independent manner. Necroptosis-deficient mice had increased survival and reduced weight loss post-IAV infection, as well as increased antioxidant and mitochondrial function, indicating partial protection to IAV infection. These findings were confirmed in vitro by pretreatment of human and rat myocytes with antioxidants or necroptosis inhibitors, which blunted oxidative stress and mitochondrial damage after IAV infection. CONCLUSIONS This study provides the first evidence that the cardiac proteome and phosphoproteome are significantly altered post-pulmonary influenza infection. Moreover, viral particles can persist in the heart after lung clearance, altering mitochondrial function and promoting cell death without active replication and interferon responses. Finally, our findings show inhibition of necroptosis or prevention of mitochondrial damage as possible therapeutic interventions to reduce cardiac damage during influenza infections. Graphic Abstract: A graphic abstract is available for this article.
Collapse
Affiliation(s)
- Yi-Han Lin
- Infectious Diseases and Genomic Medicine Group, J Craig Venter Institute, Rockville, MD (Y.-H.L., M.P.P., D.B., Y.Y., N.G.-J.)
| | - Maryann P Platt
- Infectious Diseases and Genomic Medicine Group, J Craig Venter Institute, Rockville, MD (Y.-H.L., M.P.P., D.B., Y.Y., N.G.-J.)
| | - Ryan P Gilley
- Department of Microbiology, Immunology and Molecular Genetics, The University of Texas Health Science Center at San Antonio, TX (R.P.G., P.H.D.)
| | - David Brown
- Infectious Diseases and Genomic Medicine Group, J Craig Venter Institute, Rockville, MD (Y.-H.L., M.P.P., D.B., Y.Y., N.G.-J.)
| | - Peter H Dube
- Department of Microbiology, Immunology and Molecular Genetics, The University of Texas Health Science Center at San Antonio, TX (R.P.G., P.H.D.)
| | - Yanbao Yu
- Infectious Diseases and Genomic Medicine Group, J Craig Venter Institute, Rockville, MD (Y.-H.L., M.P.P., D.B., Y.Y., N.G.-J.)
| | - Norberto Gonzalez-Juarbe
- Infectious Diseases and Genomic Medicine Group, J Craig Venter Institute, Rockville, MD (Y.-H.L., M.P.P., D.B., Y.Y., N.G.-J.)
| |
Collapse
|
32
|
Zanetti LC, Weinlich R. Necroptosis, the Other Main Caspase-Independent Cell Death. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1301:123-138. [PMID: 34370290 DOI: 10.1007/978-3-030-62026-4_7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The past decades witnessed the discovery of novel modes of cell death, such as ferroptosis, pyroptosis and necroptosis, all of them presenting common necrotic traits. In this chapter, we revisit the early discoveries that unveiled necroptosis as a distinct cell death mechanism. We describe necroptosis, its main regulators and their role in maintaining cellular homeostasis and in the disease state. We conclude by discussing its phenotypic similarities with ferroptosis and the possible crosstalk between these pathways.
Collapse
Affiliation(s)
- Larissa C Zanetti
- Hospital Israelita Albert Einstein. Av. Albert Einstein, São Paulo, SP, Brazil.
| | - Ricardo Weinlich
- Hospital Israelita Albert Einstein. Av. Albert Einstein, São Paulo, SP, Brazil
| |
Collapse
|
33
|
Detection of immunogenic cell death and its relevance for cancer therapy. Cell Death Dis 2020; 11:1013. [PMID: 33243969 PMCID: PMC7691519 DOI: 10.1038/s41419-020-03221-2] [Citation(s) in RCA: 621] [Impact Index Per Article: 124.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 02/07/2023]
Abstract
Chemotherapy, radiation therapy, as well as targeted anticancer agents can induce clinically relevant tumor-targeting immune responses, which critically rely on the antigenicity of malignant cells and their capacity to generate adjuvant signals. In particular, immunogenic cell death (ICD) is accompanied by the exposure and release of numerous damage-associated molecular patterns (DAMPs), which altogether confer a robust adjuvanticity to dying cancer cells, as they favor the recruitment and activation of antigen-presenting cells. ICD-associated DAMPs include surface-exposed calreticulin (CALR) as well as secreted ATP, annexin A1 (ANXA1), type I interferon, and high-mobility group box 1 (HMGB1). Additional hallmarks of ICD encompass the phosphorylation of eukaryotic translation initiation factor 2 subunit-α (EIF2S1, better known as eIF2α), the activation of autophagy, and a global arrest in transcription and translation. Here, we outline methodological approaches for measuring ICD markers in vitro and ex vivo for the discovery of next-generation antineoplastic agents, the development of personalized anticancer regimens, and the identification of optimal therapeutic combinations for the clinical management of cancer.
Collapse
|
34
|
The diverse roles of RIP kinases in host-pathogen interactions. Semin Cell Dev Biol 2020; 109:125-143. [PMID: 32859501 PMCID: PMC7448748 DOI: 10.1016/j.semcdb.2020.08.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 08/09/2020] [Accepted: 08/09/2020] [Indexed: 12/16/2022]
Abstract
Receptor Interacting Protein Kinases (RIPKs) are cellular signaling molecules that are critical for homeostatic signaling in both communicable and non-communicable disease processes. In particular, RIPK1, RIPK2, RIPK3 and RIPK7 have emerged as key mediators of intracellular signal transduction including inflammation, autophagy and programmed cell death, and are thus essential for the early control of many diverse pathogenic organisms. In this review, we discuss the role of each RIPK in host responses to bacterial and viral pathogens, with a focus on studies that have used pathogen infection models rather than artificial stimulation with purified pathogen associated molecular patterns. We also discuss the intricate mechanisms of host evasion by pathogens that specifically target RIPKs for inactivation, and finally, we will touch on the controversial issue of drug development for kinase inhibitors to treat chronic inflammatory and neurological disorders, and the implications this may have on the outcome of pathogen infections.
Collapse
|
35
|
Zheng M, Karki R, Vogel P, Kanneganti TD. Caspase-6 Is a Key Regulator of Innate Immunity, Inflammasome Activation, and Host Defense. Cell 2020; 181:674-687.e13. [PMID: 32298652 PMCID: PMC7425208 DOI: 10.1016/j.cell.2020.03.040] [Citation(s) in RCA: 335] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/13/2019] [Accepted: 03/17/2020] [Indexed: 02/07/2023]
Abstract
Caspases regulate cell death, immune responses, and homeostasis. Caspase-6 is categorized as an executioner caspase but shows key differences from the other executioners. Overall, little is known about the functions of caspase-6 in biological processes apart from apoptosis. Here, we show that caspase-6 mediates innate immunity and inflammasome activation. Furthermore, we demonstrate that caspase-6 promotes the activation of programmed cell death pathways including pyroptosis, apoptosis, and necroptosis (PANoptosis) and plays an essential role in host defense against influenza A virus (IAV) infection. In addition, caspase-6 promoted the differentiation of alternatively activated macrophages (AAMs). Caspase-6 facilitated the RIP homotypic interaction motif (RHIM)-dependent binding of RIPK3 to ZBP1 via its interaction with RIPK3. Altogether, our findings reveal a vital role for caspase-6 in facilitating ZBP1-mediated inflammasome activation, cell death, and host defense during IAV infection, opening additional avenues for treatment of infectious and autoinflammatory diseases and cancer.
Collapse
Affiliation(s)
- Min Zheng
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Rajendra Karki
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Peter Vogel
- Animal Resources Center and the Veterinary Pathology Core, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | |
Collapse
|
36
|
Laghlali G, Lawlor KE, Tate MD. Die Another Way: Interplay between Influenza A Virus, Inflammation and Cell Death. Viruses 2020; 12:v12040401. [PMID: 32260457 PMCID: PMC7232208 DOI: 10.3390/v12040401] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/01/2020] [Accepted: 04/01/2020] [Indexed: 02/08/2023] Open
Abstract
Influenza A virus (IAV) is a major concern to human health due to the ongoing global threat of a pandemic. Inflammatory and cell death signalling pathways play important roles in host defence against IAV infection. However, severe IAV infections in humans are characterised by excessive inflammation and tissue damage, often leading to fatal disease. While the molecular mechanisms involved in the induction of inflammation during IAV infection have been well studied, the pathways involved in IAV-induced cell death and their impact on immunopathology have not been fully elucidated. There is increasing evidence of significant crosstalk between cell death and inflammatory pathways and a greater understanding of their role in host defence and disease may facilitate the design of new treatments for IAV infection.
Collapse
Affiliation(s)
- Gabriel Laghlali
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (G.L.); (K.E.L.)
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC 3168, Australia
- Master de Biologie, École Normale Supérieure de Lyon, Université Claude Bernard Lyon I, Université de Lyon, 69007 Lyon, France
| | - Kate E. Lawlor
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (G.L.); (K.E.L.)
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC 3168, Australia
| | - Michelle D. Tate
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (G.L.); (K.E.L.)
- Department of Molecular and Translational Sciences, Monash University, Clayton, VIC 3168, Australia
- Correspondence: ; Tel.: +61-85722742
| |
Collapse
|
37
|
Bian P, Ye C, Zheng X, Luo C, Yang J, Li M, Wang Y, Yang J, Zhou Y, Zhang F, Lian J, Zhang Y, Jia Z, Lei Y. RIPK3 Promotes JEV Replication in Neurons via Downregulation of IFI44L. Front Microbiol 2020; 11:368. [PMID: 32265853 PMCID: PMC7105639 DOI: 10.3389/fmicb.2020.00368] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/19/2020] [Indexed: 12/17/2022] Open
Abstract
Japanese encephalitis virus (JEV), the leading cause of viral encephalitis in Asia, is neurovirulent and neuroinvasive. Neurons are the main target of JEV infection and propagation. Receptor interacting serine/threonine-protein kinase 3 (RIPK3) has been reported to contribute to neuroinflammation and neuronal death in many central nervous system diseases. In this study, we found that the progression of JE was alleviated in RIPK3-knockout (RIPK3–/–) mice in both peripheral and intracerebral infection. RIPK3-knockdown (RIPK3-RNAi) neuro2a cells showed higher cell viability during JEV infection. Moreover, the JEV load was significantly decreased in RIPK3–/– mouse-derived primary neurons and RIPK3-RNAi neuro2a cells compared with wild-type neurons, but this was not observed in microglia. Furthermore, RNA sequencing of brain tissues showed that the level of the interferon (IFN)-induced protein 44-like gene (IFI44L) was significantly increased in JEV-infected RIPK3–/– mouse brains, RIPK3–/– neurons, and RIPK3-RNAi-neuro2a cells. Then, it was demonstrated that the propagation of JEV was inhibited in IFI44L-overexpressing neuro2a cells and enhanced in IFI44L and RIPK3 double knockdown neuro2a cells. Taken together, our results showed that the increased expression of RIPK3 following JEV infection played complicated roles. On the one hand, RIPK3 participated in neuroinflammation and neuronal death during JEV infection. On the other hand, RIPK3 inhibited the expression of IFI44L to some extent, leading to the propagation of JEV in neurons, which might be a strategy for JEV to evade the cellular innate immune response.
Collapse
Affiliation(s)
- Peiyu Bian
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Chuantao Ye
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Xuyang Zheng
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Chuanyu Luo
- Pathogenic Biology, Medical College of Yan'an University, Yan'an, China
| | - Jiali Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, China
| | - Mengyuan Li
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Yuan Wang
- Department of Microbiology, School of Preclinical Medicine, Air Force Medical University, Xi'an, China
| | - Jing Yang
- Department of Microbiology, School of Preclinical Medicine, Air Force Medical University, Xi'an, China
| | - Yun Zhou
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Fanglin Zhang
- Department of Microbiology, School of Preclinical Medicine, Air Force Medical University, Xi'an, China
| | - Jianqi Lian
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Ying Zhang
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Zhansheng Jia
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Yingfeng Lei
- Department of Microbiology, School of Preclinical Medicine, Air Force Medical University, Xi'an, China
| |
Collapse
|
38
|
Khoury MK, Gupta K, Franco SR, Liu B. Necroptosis in the Pathophysiology of Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:272-285. [PMID: 31783008 PMCID: PMC6983729 DOI: 10.1016/j.ajpath.2019.10.012] [Citation(s) in RCA: 215] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/17/2019] [Accepted: 10/23/2019] [Indexed: 12/19/2022]
Abstract
Over the past 15 years, elegant studies have demonstrated that in certain conditions, programed cell death resembles necrosis and depends on a unique molecular pathway with no overlap with apoptosis. This form of regulated necrosis is represented by necroptosis, in which the receptor-interacting protein kinase-3 and its substrate mixed-lineage kinase domain-like protein play a crucial role. With the development of knockout mouse models and molecular inhibitors unique to necroptotic proteins, this cell death has been found to occur in virtually all tissues and diseases evaluated. There are different immunologic consequences depending on whether cells die through apoptosis or necroptosis. Therefore, distinguishing between these two forms of cell death may be crucial during pathologic evaluations. In this review, we provide an understanding of necroptotic cell-death and highlight diseases in which necroptosis has been found to play a role. We also discuss the inhibitors of necroptosis and the ways these inhibitors have been used in preclinical models of diseases. These two discussions offer an understanding of the role of necroptosis in diseases and will foster efforts to pharmacologically target this unique yet pervasive form of programed cell death in the clinic.
Collapse
Affiliation(s)
- Mitri K Khoury
- Division of Vascular Surgery, Department of Surgery, University of Wisconsin-Madison, Madison, Wisconsin
| | - Kartik Gupta
- Division of Vascular Surgery, Department of Surgery, University of Wisconsin-Madison, Madison, Wisconsin
| | - Sarah R Franco
- Division of Vascular Surgery, Department of Surgery, University of Wisconsin-Madison, Madison, Wisconsin
| | - Bo Liu
- Division of Vascular Surgery, Department of Surgery, University of Wisconsin-Madison, Madison, Wisconsin.
| |
Collapse
|
39
|
Wang Y, Hao Q, Florence JM, Jung BG, Kurdowska AK, Samten B, Idell S, Tang H. Influenza Virus Infection Induces ZBP1 Expression and Necroptosis in Mouse Lungs. Front Cell Infect Microbiol 2019; 9:286. [PMID: 31440477 PMCID: PMC6694206 DOI: 10.3389/fcimb.2019.00286] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 07/24/2019] [Indexed: 12/16/2022] Open
Abstract
Programmed cell death and especially necroptosis, a programmed and regulated form of necrosis, have been recently implicated in the progression and outcomes of influenza in mouse models. Moreover, Z-DNA/RNA binding protein 1 (ZBP1) has been identified as a key signaling molecule for necroptosis induced by Influenza A virus (IAV). Direct evidence of IAV-induced necroptosis has not been shown in infected lungs in vivo. It is also unclear as to what cell types undergo necroptosis during pulmonary IAV infection and whether ZBP1 expression can be regulated by inflammatory mediators. In this study, we found that IAV infection induced ZBP1 expression in mouse lungs. We identified that mediators implicated in the pathogenesis of IAV infection including interferons (IFNs), TNFα, and agonists for Toll-like receptors 3 and 4 were potent inducers of ZBP1 expression in primary murine alveolar epithelial cells, bone marrow derived macrophages, and dendritic cells. We further found that IAV infection induced a strong necroptosis through phosphorylation of the necroptosis effector mixed lineage kinase domain-like protein in infiltrating immune cells and alveolar epithelial cells by day 7 post-infection. Lastly, we found different cell type-specific responses to IAV-induced cell death upon inhibition of caspases and/or necroptosis pathways. Our findings provide direct evidence that IAV infection induces necroptosis in mouse lungs, which may involve local induction of ZBP1 and different programmed cell death signaling mechanisms in alveolar epithelial and infiltrating inflammatory cells in the lungs.
Collapse
Affiliation(s)
- Yun Wang
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, United States
| | - Qin Hao
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, United States
| | - Jon M Florence
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, United States
| | - Bock-Gie Jung
- Department of Pulmonary Immunology, The University of Texas Health Science Center at Tyler, Tyler, TX, United States
| | - Anna K Kurdowska
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, United States
| | - Buka Samten
- Department of Pulmonary Immunology, The University of Texas Health Science Center at Tyler, Tyler, TX, United States
| | - Steven Idell
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, United States.,Texas Lung Injury Institute, The University of Texas Health Science Center at Tyler, Tyler, TX, United States
| | - Hua Tang
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, United States
| |
Collapse
|
40
|
Salem D, Subang R, Pernet E, Divangahi M, Pineau C, Cayrol R, Levine JS, Rauch J. Necroptotic cell binding of β 2 -glycoprotein I provides a potential autoantigenic stimulus in systemic lupus erythematosus. Immunol Cell Biol 2019; 97:799-814. [PMID: 31187539 DOI: 10.1111/imcb.12279] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 06/07/2019] [Accepted: 06/08/2019] [Indexed: 01/11/2023]
Abstract
Systemic lupus erythematosus (SLE) is characterized by the development of autoantibodies against diverse self-antigens with damage to multiple organs. Immunization with the SLE autoantigen β2 -glycoprotein I (β2 GPI) and lipopolysaccharide (LPS), a known trigger of necroptosis, induces a murine model of SLE. We hypothesized that necroptotic cells, like apoptotic cells, provide a "scaffold" of cellular self-antigens, but, unlike apoptotic cells, necroptotic cells do so in a proinflammatory and immunogenic context. We demonstrate that β2 GPI indeed binds to necroptotic cells and serves as a target for anti-β2 GPI autoantibodies. We further demonstrate that necroptotic, but not apoptotic, cells promote antigenic presentation of β2 GPI to CD4 T cells by dendritic cells. Finally, we show that β2 GPI/LPS-immunized mice deficient in RIPK3 (receptor-interacting serine/threonine-protein kinase 3) or MLKL (mixed lineage kinase domain like), and consequently unable to undergo necroptosis, have reduced SLE autoantibody production and pathology. RIPK3-/- mice had low levels of SLE autoantibodies and no renal pathology, while MLKL-/- mice produced low levels of SLE autoantibodies initially, but later developed levels comparable with wild type (WT) mice and pathology intermediate to that of WT and RIPK3-/- mice. Serum cytokine levels induced by LPS tended to be lower in RIPK3-/- and MLKL-/- mice than in WT mice, suggesting that impaired proinflammatory cytokine production may impact the initiation of autoantibody production in both strains. Our data suggest that self-antigen (i.e. β2 GPI) presented in the context of necroptosis and proinflammatory signals may be sufficient to overcome immune tolerance and induce SLE.
Collapse
Affiliation(s)
- David Salem
- Division of Rheumatology, Department of Medicine, McGill University, Infectious Diseases and Immunity in Global Health Programme, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Rebecca Subang
- Division of Rheumatology, Department of Medicine, McGill University, Infectious Diseases and Immunity in Global Health Programme, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Erwan Pernet
- Department of Medicine, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Maziar Divangahi
- Department of Medicine, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Christian Pineau
- Division of Rheumatology, Department of Medicine, McGill University, Infectious Diseases and Immunity in Global Health Programme, McGill University Health Centre, Montreal, Quebec, Canada
| | - Romain Cayrol
- Department of Pathology and Cellular Biology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Jerrold S Levine
- Section of Nephrology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA.,Section of Nephrology, Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA
| | - Joyce Rauch
- Division of Rheumatology, Department of Medicine, McGill University, Infectious Diseases and Immunity in Global Health Programme, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.,Section of Nephrology, Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, USA
| |
Collapse
|
41
|
H7N9 influenza A virus activation of necroptosis in human monocytes links innate and adaptive immune responses. Cell Death Dis 2019; 10:442. [PMID: 31165725 PMCID: PMC6549191 DOI: 10.1038/s41419-019-1684-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 05/06/2019] [Accepted: 05/23/2019] [Indexed: 12/16/2022]
Abstract
We previously demonstrated that avian influenza A H7N9 virus preferentially infected CD14+ monocyte in human peripheral blood mononuclear cells (PBMCs), which led to apoptosis. To better understand H7N9 pathogenesis in relation to monocyte cell death, we showed here that extensive phosphorylation of mixed lineage kinase domain-like (MLKL) protein occurred concurrently with the activation of caspases-8, -9 and -3 in H7N9-infected monocytes at 6 h post infection (hpi), indicating that apoptosis and necroptosis pathways were simultaneously activated. The apoptotic morphology was readily observed in H7N9-infected monocytes with transmission electron microscopy (TEM), while the pan-caspase inhibitor, IDN6556 (IDN), accelerated cell death through necroptosis as evidenced by the increased level of pMLKL accompanied with cell swelling and plasma membrane rupture. Most importantly, H7N9-induced cell death could only be stopped by the combined treatment of IDN and necrosulfonamide (NSA), a pMLKL membrane translocation inhibitor, but not by individual inhibition of caspase or RIPK3. Our data further showed that activation of apoptosis and necroptosis pathways in monocytes differentially contributed to the immune response of monocytes upon H7N9 infection. Specifically, caspase inhibition significantly enhanced, while RIPK3 inhibition reduced the early expression of type I interferons and cytokine/chemokines in H7N9-infected monocytes. Moreover, culture supernatants from IDN-treated H7N9-infected monocyte promoted the expression of co-stimulatory molecule CD80, CD83 and CD86 on freshly isolated monocytes and monocyte-derived dendritic cells (MDCs) and enhanced the capacity of MDCs to induce CD3+ T-cell proliferation in vitro. In contrast, these immune stimulatory effects were abrogated by using culture supernatants from H7N9-infected monocyte with RIPK3 inhibition. In conclusion, our findings indicated that H7N9 infection activated both apoptosis and necroptosis in monocytes. An intact RIPK3 activity is required for upregulation of innate immune responses, while caspase activation suppresses the immune response.
Collapse
|
42
|
Sai K, Parsons C, House JS, Kathariou S, Ninomiya-Tsuji J. Necroptosis mediators RIPK3 and MLKL suppress intracellular Listeria replication independently of host cell killing. J Cell Biol 2019; 218:1994-2005. [PMID: 30975711 PMCID: PMC6548127 DOI: 10.1083/jcb.201810014] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 02/27/2019] [Accepted: 03/21/2019] [Indexed: 01/10/2023] Open
Abstract
RIPK3, a key mediator of necroptosis, has been implicated in the host defense against viral infection primary in immune cells. However, gene expression analysis revealed that RIPK3 is abundantly expressed not only in immune organs but also in the gastrointestinal tract, particularly in the small intestine. We found that orally inoculated Listeria monocytogenes, a bacterial foodborne pathogen, efficiently spread and caused systemic infection in Ripk3-deficient mice while almost no dissemination was observed in wild-type mice. Listeria infection activated the RIPK3-MLKL pathway in cultured cells, which resulted in suppression of intracellular replication of Listeria Surprisingly, Listeria infection-induced phosphorylation of MLKL did not result in host cell killing. We found that MLKL directly binds to Listeria and inhibits their replication in the cytosol. Our findings have revealed a novel functional role of the RIPK3-MLKL pathway in nonimmune cell-derived host defense against Listeria invasion, which is mediated through cell death-independent mechanisms.
Collapse
Affiliation(s)
- Kazuhito Sai
- Department of Biological Sciences, North Carolina State University, Raleigh, NC
| | - Cameron Parsons
- Department of Food, Bioprocessing, and Nutrition Sciences, North Carolina State University, Raleigh, NC
| | - John S House
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC
| | - Sophia Kathariou
- Department of Food, Bioprocessing, and Nutrition Sciences, North Carolina State University, Raleigh, NC
| | - Jun Ninomiya-Tsuji
- Department of Biological Sciences, North Carolina State University, Raleigh, NC
| |
Collapse
|
43
|
Leukotriene B4–type I interferon axis regulates macrophage-mediated disease tolerance to influenza infection. Nat Microbiol 2019; 4:1389-1400. [DOI: 10.1038/s41564-019-0444-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 04/03/2019] [Indexed: 11/09/2022]
|
44
|
Le Saux CJ, Chapman HA. Idiopathic Pulmonary Fibrosis: Cell Death and Inflammation Revisited. Am J Respir Cell Mol Biol 2019; 59:137-138. [PMID: 29698615 DOI: 10.1165/rcmb.2018-0083ed] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Claude Jourdan Le Saux
- 1 Department of Medicine University of California, San Francisco San Francisco, California
| | - Harold A Chapman
- 1 Department of Medicine University of California, San Francisco San Francisco, California
| |
Collapse
|
45
|
Gentle IE. Supramolecular Complexes in Cell Death and Inflammation and Their Regulation by Autophagy. Front Cell Dev Biol 2019; 7:73. [PMID: 31131275 PMCID: PMC6509160 DOI: 10.3389/fcell.2019.00073] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 04/10/2019] [Indexed: 12/23/2022] Open
Abstract
Signaling activation is a tightly regulated process involving myriad posttranslational modifications such as phosphorylation/dephosphorylation, ubiquitylation/deubiquitylation, proteolytical cleavage events as well as translocation of proteins to new compartments within the cell. In addition to each of these events potentially regulating individual proteins, the assembly of very large supramolecular complexes has emerged as a common theme in signal transduction and is now known to regulate many signaling events. This is particularly evident in pathways regulating both inflammation and cell death/survival. Regulation of the assembly and silencing of these complexes plays important roles in immune signaling and inflammation and the fate of cells to either die or survive. Here we will give a summary of some of the better studied supramolecular complexes involved in inflammation and cell death, particularly with a focus on diseases caused by their autoactivation and the role autophagy either plays or may be playing in their regulation.
Collapse
Affiliation(s)
- Ian E Gentle
- Faculty of Medicine, Institute of Medical Microbiology and Hygiene, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
| |
Collapse
|
46
|
Peltzer N, Walczak H. Cell Death and Inflammation – A Vital but Dangerous Liaison. Trends Immunol 2019; 40:387-402. [DOI: 10.1016/j.it.2019.03.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 03/12/2019] [Accepted: 03/12/2019] [Indexed: 01/07/2023]
|
47
|
Amaral EP, Costa DL, Namasivayam S, Riteau N, Kamenyeva O, Mittereder L, Mayer-Barber KD, Andrade BB, Sher A. A major role for ferroptosis in Mycobacterium tuberculosis-induced cell death and tissue necrosis. J Exp Med 2019; 216:556-570. [PMID: 30787033 PMCID: PMC6400546 DOI: 10.1084/jem.20181776] [Citation(s) in RCA: 271] [Impact Index Per Article: 45.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 12/15/2018] [Accepted: 02/01/2019] [Indexed: 12/17/2022] Open
Abstract
Necrotic tissue damage is a major pathological feature of tuberculosis. Here, Amaral et al. show that ferroptosis, a newly described regulated cell death pathway, plays an important role in Mycobacterium tuberculosis–induced cellular necrosis both in vitro and in vivo. Necrotic cell death during Mycobacterium tuberculosis (Mtb) infection is considered host detrimental since it facilitates mycobacterial spread. Ferroptosis is a type of regulated necrosis induced by accumulation of free iron and toxic lipid peroxides. We observed that Mtb-induced macrophage necrosis is associated with reduced levels of glutathione and glutathione peroxidase-4 (Gpx4), along with increased free iron, mitochondrial superoxide, and lipid peroxidation, all of which are important hallmarks of ferroptosis. Moreover, necrotic cell death in Mtb-infected macrophage cultures was suppressed by ferrostatin-1 (Fer-1), a well-characterized ferroptosis inhibitor, as well as by iron chelation. Additional experiments in vivo revealed that pulmonary necrosis in acutely infected mice is associated with reduced Gpx4 expression as well as increased lipid peroxidation and is likewise suppressed by Fer-1 treatment. Importantly, Fer-1–treated infected animals also exhibited marked reductions in bacterial load. Together, these findings implicate ferroptosis as a major mechanism of necrosis in Mtb infection and as a target for host-directed therapy of tuberculosis.
Collapse
Affiliation(s)
- Eduardo P Amaral
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Diego L Costa
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Sivaranjani Namasivayam
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Nicolas Riteau
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD.,University of Orleans and CNRS, UMR7355, Orleans, France
| | - Olena Kamenyeva
- Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Lara Mittereder
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Katrin D Mayer-Barber
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health Bethesda, MD
| | - Bruno B Andrade
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Bahia, Brazil.,Multinational Organization Network Sponsoring Translational and Epidemiological Research Initiative, José Silveira Foundation, Salvador, Brazil.,Wellcome Centre for Infectious Disease Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN.,Universidade Salvador, Laureate University, Salvador, Bahia, Brazil.,Escola Bahiana de Medicina e Saúde Pública, Salvador, Bahia, Brazil
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|
48
|
Kaufmann E, Sanz J, Dunn JL, Khan N, Mendonça LE, Pacis A, Tzelepis F, Pernet E, Dumaine A, Grenier JC, Mailhot-Léonard F, Ahmed E, Belle J, Besla R, Mazer B, King IL, Nijnik A, Robbins CS, Barreiro LB, Divangahi M. BCG Educates Hematopoietic Stem Cells to Generate Protective Innate Immunity against Tuberculosis. Cell 2018; 172:176-190.e19. [PMID: 29328912 DOI: 10.1016/j.cell.2017.12.031] [Citation(s) in RCA: 786] [Impact Index Per Article: 112.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 11/06/2017] [Accepted: 12/19/2017] [Indexed: 12/31/2022]
Abstract
The dogma that adaptive immunity is the only arm of the immune response with memory capacity has been recently challenged by several studies demonstrating evidence for memory-like innate immune training. However, the underlying mechanisms and location for generating such innate memory responses in vivo remain unknown. Here, we show that access of Bacillus Calmette-Guérin (BCG) to the bone marrow (BM) changes the transcriptional landscape of hematopoietic stem cells (HSCs) and multipotent progenitors (MPPs), leading to local cell expansion and enhanced myelopoiesis at the expense of lymphopoiesis. Importantly, BCG-educated HSCs generate epigenetically modified macrophages that provide significantly better protection against virulent M. tuberculosis infection than naïve macrophages. By using parabiotic and chimeric mice, as well as adoptive transfer approaches, we demonstrate that training of the monocyte/macrophage lineage via BCG-induced HSC reprogramming is sustainable in vivo. Our results indicate that targeting the HSC compartment provides a novel approach for vaccine development.
Collapse
Affiliation(s)
- Eva Kaufmann
- Meakins-Christie Laboratories, Department of Medicine, Department of Microbiology and Immunology, Department of Pathology, McGill International TB Centre, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Joaquin Sanz
- Department of Biochemistry, Faculty of Medicine, Université de Montréal, QC H3T 1J4, Canada; Department of Genetics, CHU Sainte-Justine Research Center, Montreal, QC H3T 1C5, Canada
| | - Jonathan L Dunn
- Meakins-Christie Laboratories, Department of Medicine, Department of Microbiology and Immunology, Department of Pathology, McGill International TB Centre, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Nargis Khan
- Meakins-Christie Laboratories, Department of Medicine, Department of Microbiology and Immunology, Department of Pathology, McGill International TB Centre, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Laura E Mendonça
- Meakins-Christie Laboratories, Department of Medicine, Department of Microbiology and Immunology, Department of Pathology, McGill International TB Centre, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Alain Pacis
- Department of Biochemistry, Faculty of Medicine, Université de Montréal, QC H3T 1J4, Canada; Department of Genetics, CHU Sainte-Justine Research Center, Montreal, QC H3T 1C5, Canada
| | - Fanny Tzelepis
- Meakins-Christie Laboratories, Department of Medicine, Department of Microbiology and Immunology, Department of Pathology, McGill International TB Centre, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Erwan Pernet
- Meakins-Christie Laboratories, Department of Medicine, Department of Microbiology and Immunology, Department of Pathology, McGill International TB Centre, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Anne Dumaine
- Department of Genetics, CHU Sainte-Justine Research Center, Montreal, QC H3T 1C5, Canada
| | | | | | - Eisha Ahmed
- Meakins-Christie Laboratories, Department of Medicine, Department of Microbiology and Immunology, Department of Pathology, McGill International TB Centre, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Jad Belle
- Department of Physiology, Complex Traits Group, McGill University, Montreal, QC H3G 0B1, Canada
| | - Rickvinder Besla
- Department of Immunology, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Bruce Mazer
- Meakins-Christie Laboratories, Department of Medicine, Department of Microbiology and Immunology, Department of Pathology, McGill International TB Centre, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Irah L King
- Meakins-Christie Laboratories, Department of Medicine, Department of Microbiology and Immunology, Department of Pathology, McGill International TB Centre, McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Anastasia Nijnik
- Department of Physiology, Complex Traits Group, McGill University, Montreal, QC H3G 0B1, Canada
| | - Clinton S Robbins
- Department of Immunology, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Luis B Barreiro
- Department of Genetics, CHU Sainte-Justine Research Center, Montreal, QC H3T 1C5, Canada; Department of Pediatrics, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1C5, Canada.
| | - Maziar Divangahi
- Meakins-Christie Laboratories, Department of Medicine, Department of Microbiology and Immunology, Department of Pathology, McGill International TB Centre, McGill University Health Centre, Montreal, QC H4A 3J1, Canada.
| |
Collapse
|
49
|
Downey J, Pernet E, Coulombe F, Divangahi M. Dissecting host cell death programs in the pathogenesis of influenza. Microbes Infect 2018; 20:560-569. [PMID: 29679740 PMCID: PMC7110448 DOI: 10.1016/j.micinf.2018.03.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 03/27/2018] [Accepted: 03/28/2018] [Indexed: 02/06/2023]
Abstract
Influenza A virus (IAV) is a pulmonary pathogen, responsible for significant yearly morbidity and mortality. Due to the absence of highly effective antiviral therapies and vaccine, as well as the constant threat of an emerging pandemic strain, there is considerable need to better understand the host-pathogen interactions and the factors that dictate a protective versus detrimental immune response to IAV. Even though evidence of IAV-induced cell death in human pulmonary epithelial and immune cells has been observed for almost a century, very little is known about the consequences of cell death on viral pathogenesis. Recent study indicates that both the type of cell death program and its kinetics have major implications on host defense and survival. In this review, we discuss advances in our understanding of cell death programs during influenza virus infection, in hopes of fostering new areas of investigation for targeted clinical intervention.
Collapse
Affiliation(s)
- Jeffrey Downey
- Department of Medicine, Department of Microbiology & Immunology, Department of Pathology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, Quebec H4A 3J1, Canada
| | - Erwan Pernet
- Department of Medicine, Department of Microbiology & Immunology, Department of Pathology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, Quebec H4A 3J1, Canada
| | - François Coulombe
- Department of Medicine, Department of Microbiology & Immunology, Department of Pathology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, Quebec H4A 3J1, Canada
| | - Maziar Divangahi
- Department of Medicine, Department of Microbiology & Immunology, Department of Pathology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, Quebec H4A 3J1, Canada.
| |
Collapse
|
50
|
Yue Y, Nabar NR, Shi CS, Kamenyeva O, Xiao X, Hwang IY, Wang M, Kehrl JH. SARS-Coronavirus Open Reading Frame-3a drives multimodal necrotic cell death. Cell Death Dis 2018. [PMID: 30185776 DOI: 10.1038/s41419‐018‐0917‐y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The molecular mechanisms underlying the severe lung pathology that occurs during SARS-CoV infections remain incompletely understood. The largest of the SARS-CoV accessory protein open reading frames (SARS 3a) oligomerizes, dynamically inserting into late endosomal, lysosomal, and trans-Golgi-network membranes. While previously implicated in a non-inflammatory apoptotic cell death pathway, here we extend the range of SARS 3a pathophysiologic targets by examining its effects on necrotic cell death pathways. We show that SARS 3a interacts with Receptor Interacting Protein 3 (Rip3), which augments the oligomerization of SARS 3a helping drive necrotic cell death. In addition, by inserting into lysosomal membranes SARS 3a triggers lysosomal damage and dysfunction. Consequently, Transcription Factor EB (TFEB) translocates to the nucleus increasing the transcription of autophagy- and lysosome-related genes. Finally, SARS 3a activates caspase-1 either directly or via an enhanced potassium efflux, which triggers NLRP3 inflammasome assembly. In summary, Rip3-mediated oligomerization of SARS 3a causes necrotic cell death, lysosomal damage, and caspase-1 activation-all likely contributing to the clinical manifestations of SARS-CoV infection.
Collapse
Affiliation(s)
- Yuan Yue
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.,B Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Neel R Nabar
- B Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA. .,Department of Molecular, Tumor, and Cell Biology, Karolinska Institutet, Stockholm, Sweden, 17165.
| | - Chong-Shan Shi
- B Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Olena Kamenyeva
- Biological Imaging Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Xun Xiao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.,B Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Il-Young Hwang
- B Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Min Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - John H Kehrl
- B Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|