1
|
Thoma A, Alomosh R, Bond HL, Akter-Miah T, Al-Shanti N, Degens H, Pekovic-Vaughan V, Lightfoot AP. A combination of major histocompatibility complex (MHC) I overexpression and type I interferon induce mitochondrial dysfunction in human skeletal myoblasts. J Cell Physiol 2024:e31458. [PMID: 39382090 DOI: 10.1002/jcp.31458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/30/2024] [Accepted: 09/26/2024] [Indexed: 10/10/2024]
Abstract
The overexpression of major histocompatibility complex (MHC) I on the surface of muscle fibers is a characteristic hallmark of the idiopathic inflammatory myopathies (IIMs), collectively termed myositis. Alongside MHC-I overexpression, subtypes of myositis, display a distinct type I interferon (IFN) signature. This study examined the combinational effects of elevated MHC-I and type I IFNs (IFNα/β) on mitochondrial function, as mitochondrial dysfunction is often seen in IIMs. Human skeletal muscle myoblasts were transfected with an MHC-I isoform using the mammalian HLA-A2/Kb vector. Mitochondrial respiration, mitochondrial membrane potential, and reactive oxygen/nitrogen species generation were assessed with or without IFNα and IFNβ. We show that MHC-I overexpression in human skeletal muscle myoblasts led to decreased basal glycolysis and mitochondrial respiration, cellular spare respiratory capacity, adenosine triphosphate-linked respiration, and an increased proton leak, which were all exaggerated by type I IFNs. Mitochondrial membrane depolarization was induced by MHC-I overexpression both in absence and presence of type I IFNs. Human myoblasts overexpressing MHC-I showed elevated nitric oxide generation that was abolished when combined with IFN. MHC-I on its own did not result in an increased reactive oxygen species (ROS) production, but IFN on their own, or combined with MHC-I overexpression did induce elevated ROS generation. Surprisingly, we observed no gross changes in mitochondrial reticular structure or markers of mitochondrial dynamics. We present new evidence that MHC-I overexpression and type I IFNs aggravate the effects each has on mitochondrial function in human skeletal muscle cells, providing novel insights into their mechanisms of action and suggesting important implications in the further study of myositis pathogenesis.
Collapse
Affiliation(s)
- Anastasia Thoma
- Department of Life Sciences, Faculty of Science & Engineering, Manchester Metropolitan University, Manchester, UK
| | - Razan Alomosh
- Department of Life Sciences, Faculty of Science & Engineering, Manchester Metropolitan University, Manchester, UK
| | - Holly L Bond
- Department of Life Sciences, Faculty of Science & Engineering, Manchester Metropolitan University, Manchester, UK
| | - Tania Akter-Miah
- Department of Life Sciences, Faculty of Science & Engineering, Manchester Metropolitan University, Manchester, UK
| | - Nasser Al-Shanti
- Department of Life Sciences, Faculty of Science & Engineering, Manchester Metropolitan University, Manchester, UK
| | - Hans Degens
- Department of Life Sciences, Faculty of Science & Engineering, Manchester Metropolitan University, Manchester, UK
- Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania
| | - Vanja Pekovic-Vaughan
- Department of Musculoskeletal and Ageing Science, Faculty of Health and Life Science, University of Liverpool, Liverpool, UK
| | - Adam P Lightfoot
- Department of Life Sciences, Faculty of Science & Engineering, Manchester Metropolitan University, Manchester, UK
| |
Collapse
|
2
|
Adcox HE, Hunt JR, Allen PE, Siff TE, Rodino KG, Ottens AK, Carlyon JA. Orientia tsutsugamushi Ank5 promotes NLRC5 cytoplasmic retention and degradation to inhibit MHC class I expression. Nat Commun 2024; 15:8069. [PMID: 39277599 PMCID: PMC11401901 DOI: 10.1038/s41467-024-52119-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 08/27/2024] [Indexed: 09/17/2024] Open
Abstract
How intracellular bacteria subvert the major histocompatibility complex (MHC) class I pathway is poorly understood. Here, we show that the obligate intracellular bacterium Orientia tsutsugamushi uses its effector protein, Ank5, to inhibit nuclear translocation of the MHC class I gene transactivator, NLRC5, and orchestrate its proteasomal degradation. Ank5 uses a tyrosine in its fourth ankyrin repeat to bind the NLRC5 N-terminus while its F-box directs host SCF complex ubiquitination of NLRC5 in the leucine-rich repeat region that dictates susceptibility to Orientia- and Ank5-mediated degradation. The ability of O. tsutsugamushi strains to degrade NLRC5 correlates with ank5 genomic carriage. Ectopically expressed Ank5 that can bind but not degrade NLRC5 protects the transactivator during Orientia infection. Thus, Ank5 is an immunoevasin that uses its bipartite architecture to rid host cells of NLRC5 and reduce surface MHC class I molecules. This study offers insight into how intracellular pathogens can impair MHC class I expression.
Collapse
Affiliation(s)
- Haley E Adcox
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, USA
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, School of Medicine, Charlottesville, VA, USA
| | - Jason R Hunt
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, USA
| | - Paige E Allen
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, USA
| | - Thomas E Siff
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, USA
| | - Kyle G Rodino
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew K Ottens
- Department of Anatomy and Neurobiology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, USA
| | - Jason A Carlyon
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, VA, USA.
| |
Collapse
|
3
|
Kafle S, Montoya B, Tang L, Tam YK, Muramatsu H, Pardi N, Sigal LJ. The roles of CD4 + T cell help, sex, and dose in the induction of protective CD8 + T cells against a lethal poxvirus by mRNA-LNP vaccines. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102279. [PMID: 39188304 PMCID: PMC11345529 DOI: 10.1016/j.omtn.2024.102279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 07/16/2024] [Indexed: 08/28/2024]
Abstract
The role of CD4+ T cells in the induction of protective CD8+ T cells by mRNA lipid nanoparticle (LNP) vaccines is unknown. We used B6 or Tlr9 -/- mice depleted or not of CD4+ T cells and LNP vaccines loaded with mRNAs encoding the ectromelia virus (ECTV) MHC class I H-2 Kb-restricted immunodominant CD8+ T cell epitope TSYKFESV (TSYKFESV mRNA-LNPs) or the ECTV EVM158 protein, which contains TSYKFESV (EVM-158 mRNA-LNPs). Following prime and boost with 10 μg of either vaccine, Kb-TSYKFESV-specific CD8+ T cells fully protected male and female mice from ECTV at 29 (both mRNA-LNPs) or 90 days (EVM158 mRNA-LNPs) post boost (dpb) independently of CD4+ T cells. However, at 29 dpb with 1 μg mRNA-LNPs, males had lower frequencies of Kb-TSYKFESV-specific CD8+ T cells and were much less well protected than females from ECTV, also independently of CD4+ T cells. At 90 dpb with 1 μg EVM158 mRNA-LNPs, the frequencies of Kb-TSYKFESV-specific CD8+ T cells in males and females were similar, and both were similarly partially protected from ECTV, independently of CD4+ T cells. Therefore, at optimal or suboptimal doses of mRNA-LNP vaccines, CD4+ T cell help is unnecessary to induce protective anti-poxvirus CD8+ T cells specific to a dominant epitope. At suboptimal doses, protection of males requires more time to develop.
Collapse
Affiliation(s)
- Samita Kafle
- Department of Microbiology and Immunology, Bluemle Life Science Building, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Brian Montoya
- Department of Microbiology and Immunology, Bluemle Life Science Building, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Lingjuan Tang
- Department of Microbiology and Immunology, Bluemle Life Science Building, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Ying K. Tam
- Acuitas Therapeutics, Vancouver, BC V6T 1Z3, Canada
| | - Hiromi Muramatsu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Luis J. Sigal
- Department of Microbiology and Immunology, Bluemle Life Science Building, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
4
|
Melo-Silva CR, Sigal LJ. Innate and adaptive immune responses that control lymph-borne viruses in the draining lymph node. Cell Mol Immunol 2024; 21:999-1007. [PMID: 38918577 PMCID: PMC11364670 DOI: 10.1038/s41423-024-01188-0] [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: 03/30/2024] [Accepted: 05/23/2024] [Indexed: 06/27/2024] Open
Abstract
The interstitial fluids in tissues are constantly drained into the lymph nodes (LNs) as lymph through afferent lymphatic vessels and from LNs into the blood through efferent lymphatics. LNs are strategically positioned and have the appropriate cellular composition to serve as sites of adaptive immune initiation against invading pathogens. However, for lymph-borne viruses, which disseminate from the entry site to other tissues through the lymphatic system, immune cells in the draining LN (dLN) also play critical roles in curbing systemic viral dissemination during primary and secondary infections. Lymph-borne viruses in tissues can be transported to dLNs as free virions in the lymph or within infected cells. Regardless of the entry mechanism, infected myeloid antigen-presenting cells, including various subtypes of dendritic cells, inflammatory monocytes, and macrophages, play a critical role in initiating the innate immune response within the dLN. This innate immune response involves cellular crosstalk between infected and bystander innate immune cells that ultimately produce type I interferons (IFN-Is) and other cytokines and recruit inflammatory monocytes and natural killer (NK) cells. IFN-I and NK cell cytotoxicity can restrict systemic viral spread during primary infections and prevent serious disease. Additionally, the memory CD8+ T-cells that reside or rapidly migrate to the dLN can contribute to disease prevention during secondary viral infections. This review explores the intricate innate immune responses orchestrated within dLNs that contain primary viral infections and the role of memory CD8+ T-cells following secondary infection or CD8+ T-cell vaccination.
Collapse
Affiliation(s)
- Carolina R Melo-Silva
- Department of Microbiology and Immunology, Thomas Jefferson University, Bluemle Life Sciences Building Room 709, 233 South 10th Street, Philadelphia, PA, 19107, USA.
| | - Luis J Sigal
- Department of Microbiology and Immunology, Thomas Jefferson University, Bluemle Life Sciences Building Room 709, 233 South 10th Street, Philadelphia, PA, 19107, USA.
| |
Collapse
|
5
|
Bisimwa PN, Ongus JR, Tonui R, Bisimwa EB, Steinaa L. Resistance to African swine fever virus among African domestic pigs appears to be associated with a distinct polymorphic signature in the RelA gene and upregulation of RelA transcription. Virol J 2024; 21:93. [PMID: 38658979 PMCID: PMC11041040 DOI: 10.1186/s12985-024-02351-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 03/24/2024] [Indexed: 04/26/2024] Open
Abstract
African swine fever virus (ASFV) is a highly contagious and fatal hemorrhagic disease of domestic pigs, which poses a major threat to the swine industry worldwide. Studies have shown that indigenous African pigs tolerate ASFV infection better than European pigs. The porcine v-rel avian reticuloendotheliosis viral oncogene homolog A (RelA) encoding a p65 kD protein, a major subunit of the NF-kB transcription factor, plays important roles in controlling both innate and adaptive immunity during infection with ASFV. In the present study, RelA genes from ASFV-surviving and symptomatic pigs were sequenced and found to contain polymorphisms revealing two discrete RelA amino acid sequences. One was found in the surviving pigs, and the other in symptomatic pigs. In total, 16 nonsynonymous SNPs (nsSNPs) resulting in codon changes were identified using bioinformatics software (SIFT and Polyphen v2) and web-based tools (MutPre and PredictSNP). Seven nsSNPs (P374-S, T448-S, P462-R, V464-P, Q478-H, L495-E, and P499-Q) were predicted to alter RelA protein function and stability, while 5 of these (P374-S, T448-S, P462-R, L495-E, and Q499-P) were predicted as disease-related SNPs.Additionally, the inflammatory cytokine levels of IFN-α, IL-10, and TNF-α at both the protein and the mRNA transcript levels were measured using ELISA and Real-Time PCR, respectively. The resulting data was used in correlation analysis to assess the association between cytokine levels and the RelA gene expression. Higher levels of IFN-α and detectable levels of IL-10 protein and RelA mRNA were observed in surviving pigs compared to healthy (non-infected). A positive correlation of IFN-α cytokine levels with RelA mRNA expression was also obtained. In conclusion, 7 polymorphic events in the coding region of the RelA gene may contribute to the tolerance of ASFV in pigs.
Collapse
Affiliation(s)
- Patrick N Bisimwa
- Molecular Biology Laboratory, Department of Animal Sciences and Production, Université Evangélique en Afrique, Bukavu, Democratic Republic of Congo.
- Institut Supérieur de Dévelopement Rural de Kaziba, Kaziba, Democratic Republic of Congo.
| | - Juliette R Ongus
- Department of Medical Laboratory Sciences, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
- Biotechnology Laboratory, Departement of Molecular Biology and Biotechnology, Pan African University Institute of Basic Sciences, Technology and Innovation, Nairobi, Kenya
| | - Ronald Tonui
- Department of Medical Laboratory Sciences, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
| | - Espoir B Bisimwa
- Molecular Biology Laboratory, Department of Animal Sciences and Production, Université Evangélique en Afrique, Bukavu, Democratic Republic of Congo
| | - Lucilla Steinaa
- Animal and Human Health Program, International Livestock Research Institute, Nairobi, Kenya
| |
Collapse
|
6
|
Gao Z, He X, Chen G, Fang Y, Meng Z, Tian H, Zhang H, Jing Z. The Viral Protein Poly(A) Polymerase Catalytic Subunit Interacts with Guanylate-Binding Proteins 2 to Antagonize the Antiviral Ability of Targeting Ectromelia Virus. Int J Mol Sci 2023; 24:15750. [PMID: 37958732 PMCID: PMC10648259 DOI: 10.3390/ijms242115750] [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: 09/18/2023] [Revised: 10/16/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
The recent spread of the monkeypox virus among humans has heightened concerns regarding orthopoxvirus infections. Consequently, conducting a comprehensive study on the immunobiology of the monkeypox virus is imperative for the development of effective therapeutics. Ectromelia virus (ECTV) closely resembles the genetic and disease characteristics of monkeypox virus, making it a valuable research tool for studying orthopoxvirus-host interactions. Guanylate-binding proteins (GBPs), highly expressed interferon-stimulated genes (ISGs), have antagonistic effects against various intracellular pathogenic microorganisms. Our previous research has shown that GBP2 has a mild but statistically significant inhibitory effect on ECTV infection. The presence of a significant number of molecules in the poxvirus genome that encode the host immune response raises questions about whether it also includes proteins that counteract the antiviral activity of GBP2. Using IP/MS and co-IP technology, we discovered that the poly(A) polymerase catalytic subunit (PAPL) protein of ECTV is a viral regulatory molecule that interacts with GBP2. Further studies have shown that PAPL antagonizes the antiviral activity of GBP2 by reducing its protein levels. Knocking out the PAPL gene of ECTV with the CRISPR/Cas9 system significantly diminishes the replication ability of the virus, indicating the indispensable role of PAPL in the replication process of ECTV. In conclusion, our study presents preliminary evidence supporting the significance of PAPL as a virulence factor that can interact with GBP2.
Collapse
Affiliation(s)
- Zhenzhen Gao
- State Key Laboratory for Animal Disease Control and Prevention, Ministry of Agriculture Key Laboratory of Veterinary Public Health, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Z.G.); (X.H.); (G.C.); (Y.F.); (H.T.); (H.Z.)
| | - Xiaobing He
- State Key Laboratory for Animal Disease Control and Prevention, Ministry of Agriculture Key Laboratory of Veterinary Public Health, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Z.G.); (X.H.); (G.C.); (Y.F.); (H.T.); (H.Z.)
| | - Guohua Chen
- State Key Laboratory for Animal Disease Control and Prevention, Ministry of Agriculture Key Laboratory of Veterinary Public Health, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Z.G.); (X.H.); (G.C.); (Y.F.); (H.T.); (H.Z.)
| | - Yongxiang Fang
- State Key Laboratory for Animal Disease Control and Prevention, Ministry of Agriculture Key Laboratory of Veterinary Public Health, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Z.G.); (X.H.); (G.C.); (Y.F.); (H.T.); (H.Z.)
| | - Zejing Meng
- School of Public Health, Lanzhou University, Lanzhou 730000, China;
| | - Huihui Tian
- State Key Laboratory for Animal Disease Control and Prevention, Ministry of Agriculture Key Laboratory of Veterinary Public Health, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Z.G.); (X.H.); (G.C.); (Y.F.); (H.T.); (H.Z.)
| | - Hui Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Ministry of Agriculture Key Laboratory of Veterinary Public Health, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Z.G.); (X.H.); (G.C.); (Y.F.); (H.T.); (H.Z.)
| | - Zhizhong Jing
- State Key Laboratory for Animal Disease Control and Prevention, Ministry of Agriculture Key Laboratory of Veterinary Public Health, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, China; (Z.G.); (X.H.); (G.C.); (Y.F.); (H.T.); (H.Z.)
- School of Public Health, Lanzhou University, Lanzhou 730000, China;
| |
Collapse
|
7
|
Poledniczek M, Neumayer C, Kopp CW, Schlager O, Gremmel T, Jozkowicz A, Gschwandtner ME, Koppensteiner R, Wadowski PP. Micro- and Macrovascular Effects of Inflammation in Peripheral Artery Disease-Pathophysiology and Translational Therapeutic Approaches. Biomedicines 2023; 11:2284. [PMID: 37626780 PMCID: PMC10452462 DOI: 10.3390/biomedicines11082284] [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: 06/25/2023] [Revised: 08/10/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Inflammation has a critical role in the development and progression of atherosclerosis. On the molecular level, inflammatory pathways negatively impact endothelial barrier properties and thus, tissue homeostasis. Conformational changes and destruction of the glycocalyx further promote pro-inflammatory pathways also contributing to pro-coagulability and a prothrombotic state. In addition, changes in the extracellular matrix composition lead to (peri-)vascular remodelling and alterations of the vessel wall, e.g., aneurysm formation. Moreover, progressive fibrosis leads to reduced tissue perfusion due to loss of functional capillaries. The present review aims at discussing the molecular and clinical effects of inflammatory processes on the micro- and macrovasculature with a focus on peripheral artery disease.
Collapse
Affiliation(s)
- Michael Poledniczek
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (M.P.); (C.W.K.); (O.S.); (M.E.G.); (R.K.)
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
| | - Christoph Neumayer
- Division of Vascular Surgery, Department of General Surgery, Medical University of Vienna, 1090 Vienna, Austria;
| | - Christoph W. Kopp
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (M.P.); (C.W.K.); (O.S.); (M.E.G.); (R.K.)
| | - Oliver Schlager
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (M.P.); (C.W.K.); (O.S.); (M.E.G.); (R.K.)
| | - Thomas Gremmel
- Department of Internal Medicine I, Cardiology and Intensive Care Medicine, Landesklinikum Mistelbach-Gänserndorf, 2130 Mistelbach, Austria;
- Institute of Cardiovascular Pharmacotherapy and Interventional Cardiology, Karl Landsteiner Society, 3100 St. Pölten, Austria
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biophysics, Biochemistry and Biotechnology, Jagiellonian University, 31-007 Krakow, Poland;
| | - Michael E. Gschwandtner
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (M.P.); (C.W.K.); (O.S.); (M.E.G.); (R.K.)
| | - Renate Koppensteiner
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (M.P.); (C.W.K.); (O.S.); (M.E.G.); (R.K.)
| | - Patricia P. Wadowski
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (M.P.); (C.W.K.); (O.S.); (M.E.G.); (R.K.)
| |
Collapse
|
8
|
Panzer B, Kopp CW, Neumayer C, Koppensteiner R, Jozkowicz A, Poledniczek M, Gremmel T, Jilma B, Wadowski PP. Toll-like Receptors as Pro-Thrombotic Drivers in Viral Infections: A Narrative Review. Cells 2023; 12:1865. [PMID: 37508529 PMCID: PMC10377790 DOI: 10.3390/cells12141865] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/12/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Toll-like receptors (TLRs) have a critical role in the pathogenesis and disease course of viral infections. The induced pro-inflammatory responses result in the disturbance of the endovascular surface layer and impair vascular homeostasis. The injury of the vessel wall further promotes pro-thrombotic and pro-coagulatory processes, eventually leading to micro-vessel plugging and tissue necrosis. Moreover, TLRs have a direct role in the sensing of viruses and platelet activation. TLR-mediated upregulation of von Willebrand factor release and neutrophil, as well as macrophage extra-cellular trap formation, further contribute to (micro-) thrombotic processes during inflammation. The following review focuses on TLR signaling pathways of TLRs expressed in humans provoking pro-thrombotic responses, which determine patient outcome during viral infections, especially in those with cardiovascular diseases.
Collapse
Affiliation(s)
- Benjamin Panzer
- Department of Cardiology, Wilhelminenspital, 1090 Vienna, Austria
| | - Christoph W Kopp
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
| | - Christoph Neumayer
- Division of Vascular Surgery, Department of Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Renate Koppensteiner
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
| | - Alicja Jozkowicz
- Faculty of Biophysics, Biochemistry and Biotechnology, Department of Medical Biotechnology, Jagiellonian University, 30-387 Krakow, Poland
| | - Michael Poledniczek
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
| | - Thomas Gremmel
- Institute of Cardiovascular Pharmacotherapy and Interventional Cardiology, Karl Landsteiner Society, 3100 St. Pölten, Austria
- Department of Internal Medicine I, Cardiology and Intensive Care Medicine, Landesklinikum Mistelbach-Gänserndorf, 2130 Mistelbach, Austria
| | - Bernd Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Patricia P Wadowski
- Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
9
|
Resistance To Poxvirus Lethality Does Not Require the Necroptosis Proteins RIPK3 or MLKL. J Virol 2023; 97:e0194522. [PMID: 36651749 PMCID: PMC9973014 DOI: 10.1128/jvi.01945-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Receptor-interacting protein kinase 3 (RIPK3) and mixed lineage kinase domain-like pseudokinase (MLKL) are proteins that are critical for necroptosis, a mechanism of programmed cell death that is both activated when apoptosis is inhibited and thought to be antiviral. Here, we investigated the role of RIPK3 and MLKL in controlling the Orthopoxvirus ectromelia virus (ECTV), a natural pathogen of the mouse. We found that C57BL/6 (B6) mice deficient in RIPK3 (Ripk3-/-) or MLKL (Mlkl-/-) were as susceptible as wild-type (WT) B6 mice to ECTV lethality after low-dose intraperitoneal infection and were as resistant as WT B6 mice after ECTV infection through the natural footpad route. Additionally, after footpad infection, Mlkl-/- mice, but not Ripk3-/- mice, endured lower viral titers than WT mice in the draining lymph node (dLN) at three days postinfection and in the spleen or in the liver at seven days postinfection. Despite the improved viral control, Mlkl-/- mice did not differ from WT mice in the expression of interferons or interferon-stimulated genes or in the recruitment of natural killer (NK) cells and inflammatory monocytes (iMOs) to the dLN. Additionally, the CD8 T-cell responses in Mlkl-/- and WT mice were similar, even though in the dLNs of Mlkl-/- mice, professional antigen-presenting cells were more heavily infected. Finally, the histopathology in the livers of Mlkl-/- and WT mice at 7 dpi did not differ. Thus, the mechanism of the increased virus control by Mlkl-/- mice remains to be defined. IMPORTANCE The molecules RIPK3 and MLKL are required for necroptotic cell death, which is widely thought of as an antiviral mechanism. Here we show that C57BL/6 (B6) mice deficient in RIPK3 or MLKL are as susceptible as WT B6 mice to ECTV lethality after a low-dose intraperitoneal infection and are as resistant as WT B6 mice after ECTV infection through the natural footpad route. Mice deficient in MLKL are more efficient than WT mice at controlling virus loads in various organs. This improved viral control is not due to enhanced interferon, natural killer cell, or CD8 T-cell responses. Overall, the data indicate that deficiencies in the molecules that are critical to necroptosis do not necessarily result in worse outcomes following viral infection and may improve virus control.
Collapse
|
10
|
Rahman MM, McFadden G. Role of cytokines in poxvirus host tropism and adaptation. Curr Opin Virol 2022; 57:101286. [PMID: 36427482 PMCID: PMC9704024 DOI: 10.1016/j.coviro.2022.101286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/19/2022] [Accepted: 10/26/2022] [Indexed: 11/23/2022]
Abstract
Poxviruses are a diverse family of double-stranded DNA viruses that cause mild-to-severe disease in selective hosts, including humans. Although most poxviruses are restricted to their hosts, some members can leap host species and cause zoonotic diseases and, therefore, are genuine threats to human and animal health. The recent global spread of monkeypox in humans suggests that zoonotic poxviruses can adapt to a new host, spread rapidly in the new host, and evolve to better evade host innate barriers. Unlike many other viruses, poxviruses express an extensive repertoire of self-defense proteins that play a vital role in the evasion of host innate and adaptive immune responses in their newest host species. The function of these viral immune modulators and host-specific cytokine responses can result in different host tropism and poxvirus disease progression. Here, we review the role of different cytokines that control poxvirus host tropism and adaptation.
Collapse
|
11
|
Melo-Silva CR, Roman MI, Knudson CJ, Tang L, Xu RH, Tassetto M, Dolan P, Andino R, Sigal LJ. Interferon partly dictates a divergent transcriptional response in poxvirus-infected and bystander inflammatory monocytes. Cell Rep 2022; 41:111676. [PMID: 36417857 PMCID: PMC9798443 DOI: 10.1016/j.celrep.2022.111676] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 09/07/2022] [Accepted: 10/25/2022] [Indexed: 11/23/2022] Open
Abstract
Inflammatory monocytes (iMOs) and B cells are the main targets of the poxvirus ectromelia virus (ECTV) in the lymph nodes of mice and play distinct roles in surviving the infection. Infected and bystander iMOs control ECTV's systemic spread, preventing early death, while B cells make antibodies that eliminate ECTV. Our work demonstrates that within an infected animal that survives ECTV infection, intrinsic and bystander infection of iMOs and B cells differentially control the transcription of genes important for immune cell function and, perhaps, cell identity. Bystander cells upregulate metabolism, antigen presentation, and interferon-stimulated genes. Infected cells downregulate many cell-type-specific genes and upregulate transcripts typical of non-immune cells. Bystander (Bys) and infected (Inf) iMOs non-redundantly contribute to the cytokine milieu and the interferon response. Furthermore, we uncover how type I interferon (IFN-I) or IFN-γ signaling differentially regulates immune pathways in Inf and Bys iMOs and that, at steady state, IFN-I primes iMOs for rapid IFN-I production and antigen presentation.
Collapse
Affiliation(s)
- Carolina R. Melo-Silva
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Marisa I. Roman
- Department of Physics, St. Joseph University, Philadelphia PA 19131, USA
| | - Cory J. Knudson
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107, USA,GlaxoSmithKline, 1250 S. Collegeville Road, Collegeville, PA 19426, USA
| | - Lingjuan Tang
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Ren-Huan Xu
- Advanced RNA Vaccine Technologies, Inc., 12358 Parklawn Dr, North Bethesda, MD 20852, USA
| | - Michel Tassetto
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Patrick Dolan
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94158, USA,Laboratory of Viral Diseases, NIAID, NIH, Bethesda, MD 20892-3210, USA
| | - Raul Andino
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94158, USA
| | - Luis J. Sigal
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107, USA,Lead contact,Correspondence:
| |
Collapse
|
12
|
Perego M, Fu S, Cao Y, Kossenkov A, Yao M, Bonner E, Alicea-Torres K, Liu W, Jiang Z, Chen Z, Fuchs SY, Zhou J, Gabrilovich DI. Mechanisms regulating transitory suppressive activity of neutrophils in newborns: PMNs-MDSCs in newborns. J Leukoc Biol 2022; 112:955-968. [PMID: 35726818 PMCID: PMC9794389 DOI: 10.1002/jlb.4hi0921-514rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 05/27/2022] [Accepted: 05/28/2022] [Indexed: 12/30/2022] Open
Abstract
Transitory appearance of immune suppressive polymorphonuclear neutrophils (PMNs) defined as myeloid-derived suppressor cells (PMNs-MDSCs) in newborns is important for their protection from inflammation associated with newly established gut microbiota. Here, we report that inhibition of the type I IFN (IFN1) pathway played a major role in regulation of PMNs-MDSCs-suppressive activity during first weeks of life. Expression of the IFN1 receptor IFNAR1 was markedly lower in PMNs-MDSCs. However, in newborn mice, down-regulation of IFNAR1 was not sufficient to render PMNs immune suppressive. That also required the presence of a positive signal from lactoferrin via its receptor low-density lipoprotein receptor-related protein 2. The latter effect was mediated via NF-κB activation, which was tempered by IFN1 in a manner that involved suppressor of cytokine signaling 3. Thus, we discovered a mechanism of tight regulation of immune suppressive PMNs-MDSCs in newborns, which may be used in the development of therapies of neonatal pathologies.
Collapse
Affiliation(s)
| | - Shuyu Fu
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Yingjiao Cao
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Sciences, Tianjin Medical University, Tianjin, China
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | | | - Meng Yao
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Sciences, Tianjin Medical University, Tianjin, China
| | - Erin Bonner
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Kevin Alicea-Torres
- The Wistar Institute, Philadelphia, Pennsylvania, USA
- Biology Department, University of Puerto Rico-Humacao, Humacao, Puerto Rico, USA
| | - Wangkai Liu
- Department of Pediatrics, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhilong Jiang
- Department of Respiratory and Critical Care Medicine, Zhongshan Hospital; Shanghai Institute of Respiratory Disease, Fudan University, Shanghai, China
| | - Zhihong Chen
- Department of Respiratory and Critical Care Medicine, Zhongshan Hospital; Shanghai Institute of Respiratory Disease, Fudan University, Shanghai, China
| | - Serge Y Fuchs
- Department of Biomedical Sciences, School of Veterinary Medicine University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jie Zhou
- Key Laboratory of Immune Microenvironment and Disease of the Ministry of Education, Department of Immunology, School of Basic Sciences, Tianjin Medical University, Tianjin, China
| | | |
Collapse
|
13
|
Lang R, Li H, Luo X, Liu C, Zhang Y, Guo S, Xu J, Bao C, Dong W, Yu Y. Expression and mechanisms of interferon-stimulated genes in viral infection of the central nervous system (CNS) and neurological diseases. Front Immunol 2022; 13:1008072. [PMID: 36325336 PMCID: PMC9618809 DOI: 10.3389/fimmu.2022.1008072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 09/28/2022] [Indexed: 09/16/2023] Open
Abstract
Interferons (IFNs) bind to cell surface receptors and activate the expression of interferon-stimulated genes (ISGs) through intracellular signaling cascades. ISGs and their expression products have various biological functions, such as antiviral and immunomodulatory effects, and are essential effector molecules for IFN function. ISGs limit the invasion and replication of the virus in a cell-specific and region-specific manner in the central nervous system (CNS). In addition to participating in natural immunity against viral infections, studies have shown that ISGs are essential in the pathogenesis of CNS disorders such as neuroinflammation and neurodegenerative diseases. The aim of this review is to present a macroscopic overview of the characteristics of ISGs that restrict viral neural invasion and the expression of the ISGs underlying viral infection of CNS cells. Furthermore, we elucidate the characteristics of ISGs expression in neurological inflammation, neuropsychiatric disorders such as depression as well as neurodegenerative disorders, including Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). Finally, we summarize several ISGs (ISG15, IFIT2, IFITM3) that have been studied more in recent years for their antiviral infection in the CNS and their research progress in neurological diseases.
Collapse
Affiliation(s)
- Rui Lang
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Huiting Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Xiaoqin Luo
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Cencen Liu
- Department of Pathology, People’s Hospital of Zhongjiang County, DeYang, China
| | - Yiwen Zhang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - ShunYu Guo
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jingyi Xu
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Changshun Bao
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Clinical Research Center for Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Neurological diseases and brain function laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Wei Dong
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Yang Yu
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| |
Collapse
|
14
|
Córdoba-David G, García-Giménez J, Cardoso Castelo-Branco R, Carrasco S, Cannata P, Ortiz A, Ramos AM. Crosstalk between TBK1/IKKε and the type I interferon pathway contributes to tubulointerstitial inflammation and kidney tubular injury. Front Pharmacol 2022; 13:987979. [PMID: 36386242 PMCID: PMC9647636 DOI: 10.3389/fphar.2022.987979] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/22/2022] [Indexed: 09/01/2023] Open
Abstract
The type I interferon (TI-IFN) pathway regulates innate immunity, inflammation, and apoptosis during infection. However, the contribution of the TI-IFN pathway or upstream signaling pathways to tubular injury in kidney disease is poorly understood. Upon observing evidence of activation of upstream regulators of the TI-IFN pathway in a transcriptomics analysis of murine kidney tubulointerstitial injury, we have now addressed the impact of the TI-IFN and upstream signaling pathways on kidney tubulointerstitial injury. In cultured tubular cells and kidney tissue, IFNα/β binding to IFNAR activated the TI-IFN pathway and recruited antiviral interferon-stimulated genes (ISG) and NF-κB-associated proinflammatory responses. TWEAK and lipopolysaccharide (LPS) signaled through TBK1/IKKε and IRF3 to activate both ISGs and NF-κB. In addition, TWEAK recruited TLR4 to stimulate TBK1/IKKε-dependent ISG and inflammatory responses. Dual pharmacological inhibition of TBK1/IKKε with amlexanox decreased TWEAK- or LPS-induced ISG and cytokine responses, as well as cell death induced by a complex inflammatory milieu that included TWEAK. TBK1 or IRF3 siRNA prevented the TWEAK-induced ISG and inflammatory gene expression while IKKε siRNA did not. In vivo, kidney IFNAR and IFNβ were increased in murine LPS and folic acid nephrotoxicity while IFNAR was increased in human kidney biopsies with tubulointerstitial damage. Inhibition of TBK1/IKKε with amlexanox or IFNAR neutralization decreased TI-IFN pathway activation and protected from kidney injury induced by folic acid or LPS. In conclusion, TI-IFNs, TWEAK, and LPS engage interrelated proinflammatory and antiviral responses in tubular cells. Moreover, inhibition of TBK1/IKKε with amlexanox, and IFNAR targeting, may protect from tubulointerstitial kidney injury.
Collapse
Affiliation(s)
- Gina Córdoba-David
- Department of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Jorge García-Giménez
- Department of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain
| | | | - Susana Carrasco
- Department of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain
- RICORS 2040, Madrid, Spain
| | - Pablo Cannata
- Department of Pathology, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Alberto Ortiz
- Department of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain
- RICORS 2040, Madrid, Spain
- Department of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Adrián M. Ramos
- Department of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain
- RICORS 2040, Madrid, Spain
| |
Collapse
|
15
|
Kusiak A, Brady G. Bifurcation of signalling in human innate immune pathways to NF-kB and IRF family activation. Biochem Pharmacol 2022; 205:115246. [PMID: 36088989 DOI: 10.1016/j.bcp.2022.115246] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 11/28/2022]
Abstract
The human innate immune response can be activated through a wide range of stimuli. This multi-faceted system can be triggered by a range of immunostimulants including pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). These stimuli drive intracellular signalling pathways that branch off downstream to activate several distinct transcription factors. The two most impactful of which in innate immune outcomes are the NF-κB and the IRF family members. Both transcription factor families play defining roles in driving inflammation as well as the antiviral response. Pathways leading to their simultaneous activation share common upstream components but eventually distinct regulators which directly facilitate their activation. This review will discuss the current state of knowledge about what is known about how these pathways bifurcate to activate NF-κB and IRF family members.
Collapse
Affiliation(s)
- Aleksandra Kusiak
- Trinity Translational Medicine Institute, St James' Campus, Trinity College Dublin, D08 W9RT Dublin, Ireland.
| | - Gareth Brady
- Trinity Translational Medicine Institute, St James' Campus, Trinity College Dublin, D08 W9RT Dublin, Ireland.
| |
Collapse
|
16
|
Son M, Frank T, Holst-Hansen T, Wang AG, Junkin M, Kashaf SS, Trusina A, Tay S. Spatiotemporal NF-κB dynamics encodes the position, amplitude, and duration of local immune inputs. SCIENCE ADVANCES 2022; 8:eabn6240. [PMID: 36044569 PMCID: PMC9432835 DOI: 10.1126/sciadv.abn6240] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 07/19/2022] [Indexed: 05/31/2023]
Abstract
Infected cells communicate through secreted signaling molecules like cytokines, which carry information about pathogens. How differences in cytokine secretion affect inflammatory signaling over space and how responding cells decode information from propagating cytokines are not understood. By computationally and experimentally studying NF-κB dynamics in cocultures of signal-sending cells (macrophages) and signal-receiving cells (fibroblasts), we find that cytokine signals are transmitted by wave-like propagation of NF-κB activity and create well-defined activation zones in responding cells. NF-κB dynamics in responding cells can simultaneously encode information about cytokine dose, duration, and distance to the cytokine source. Spatially resolved transcriptional analysis reveals that responding cells transmit local cytokine information to distance-specific proinflammatory gene expression patterns, creating "gene expression zones." Despite single-cell variability, the size and duration of the signaling zone are tightly controlled by the macrophage secretion profile. Our results highlight how macrophages tune cytokine secretion to control signal transmission distance and how inflammatory signaling interprets these signals in space and time.
Collapse
Affiliation(s)
- Minjun Son
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
- Institute for Genomics and Systems Biology, University of Chicago, Chicago, IL 60637, USA
| | - Tino Frank
- Department of Biosystems Science and Engineering, ETH Zürich, Basel 4058, Switzerland
| | | | - Andrew G. Wang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Michael Junkin
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
- Department of Biosystems Science and Engineering, ETH Zürich, Basel 4058, Switzerland
| | - Sara S. Kashaf
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Ala Trusina
- Niels Bohr Institute, University of Copenhagen, Copenhagen 2100, Denmark
| | - Savaş Tay
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
- Institute for Genomics and Systems Biology, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
17
|
Microglial NF-κB drives tau spreading and toxicity in a mouse model of tauopathy. Nat Commun 2022; 13:1969. [PMID: 35413950 PMCID: PMC9005658 DOI: 10.1038/s41467-022-29552-6] [Citation(s) in RCA: 122] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 03/21/2022] [Indexed: 01/31/2023] Open
Abstract
Activation of microglia is a prominent pathological feature in tauopathies, including Alzheimer's disease. How microglia activation contributes to tau toxicity remains largely unknown. Here we show that nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling, activated by tau, drives microglial-mediated tau propagation and toxicity. Constitutive activation of microglial NF-κB exacerbated, while inactivation diminished, tau seeding and spreading in young PS19 mice. Inhibition of NF-κB activation enhanced the retention while reduced the release of internalized pathogenic tau fibrils from primary microglia and rescued microglial autophagy deficits. Inhibition of microglial NF-κB in aged PS19 mice rescued tau-mediated learning and memory deficits, restored overall transcriptomic changes while increasing neuronal tau inclusions. Single cell RNA-seq revealed that tau-associated disease states in microglia were diminished by NF-κB inactivation and further transformed by constitutive NF-κB activation. Our study establishes a role for microglial NF-κB signaling in mediating tau spreading and toxicity in tauopathy.
Collapse
|
18
|
AlDaif BA, Mercer AA, Fleming SB. The parapoxvirus Orf virus ORF116 gene encodes an antagonist of the interferon response. J Gen Virol 2021; 102. [PMID: 34890310 DOI: 10.1099/jgv.0.001695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Orf virus (ORFV) is the type species of the Parapoxvirus genus of the Poxviridae family. Genetic and functional studies have revealed ORFV has multiple immunomodulatory genes that manipulate innate immune responses, during the early stage of infection. ORF116 is a novel gene of ORFV with hitherto unknown function. Characterization of an ORF116 deletion mutant showed that it replicated in primary lamb testis cells with reduced levels compared to the wild-type and produced a smaller plaque phenotype. ORF116 was shown to be expressed prior to DNA replication. The potential function of ORF116 was investigated by gene-expression microarray analysis in HeLa cells infected with wild-type ORFV or the ORF116 deletion mutant. The analysis of differential cellular gene expression revealed a number of interferon-stimulated genes (ISGs) differentially expressed at either 4 or 6 h post infection. IFI44 showed the greatest differential expression (4.17-fold) between wild-type and knockout virus. Other ISGs that were upregulated in the knockout included RIG-I, IFIT2, MDA5, OAS1, OASL, DDX60, ISG20 and IFIT1 and in addition the inflammatory cytokine IL-8. These findings were validated by infecting HeLa cells with an ORF116 revertant recombinant virus and analysis of transcript expression by quantitative real time-PCR (qRT-PCR). These observations suggested a role for the ORFV gene ORF116 in modulating the IFN response and inflammatory cytokines. This study represents the first functional analysis of ORF116.
Collapse
Affiliation(s)
- Basheer A AlDaif
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Andrew A Mercer
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Stephen B Fleming
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
19
|
Anaeigoudari A, Mollaei HR, Arababadi MK, Nosratabadi R. Severe Acute Respiratory Syndrome Coronavirus 2: The Role of the Main Components of the Innate Immune System. Inflammation 2021; 44:2151-2169. [PMID: 34524614 PMCID: PMC8442517 DOI: 10.1007/s10753-021-01519-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 06/08/2021] [Accepted: 07/08/2021] [Indexed: 02/08/2023]
Abstract
At the end of December 2019, the COVID-19 pandemic began in Wuhan of China. COVID-19 affects different people with a wide spectrum of clinical manifestations, ranging from asymptomatic with recovery without hospitalization up to a severe acute respiratory syndrome (SARS). The innate and adaptive immunity appears responsible for the defense against the virus and recovery from the disease. The innate immune system, as the first line of defense, is essential for the detection of virus and subsequent activation of acquired immunity. The innate immune response is carried out by sentinel cells such as monocytes/macrophages and dendritic cells and by receptors known as pattern recognition receptors (PRR). These receptors can recognize various components of the virus, which lead to intracellular signaling and subsequently the synthesis of various cytokines. These cytokines then recruit other immune cells, activate adaptive immune responses, and inhibit viral spreading. The most common receptors include Toll-like receptors, C-type lectin receptors, and RIG-I like receptors. This review describes the current knowledge about the interplay between innate immune responses and SARS-CoV-2 with a focus on the innate immune cells and the role of their receptors in viral RNA recognition, as well as their mechanisms for recognizing SARS-CoV-2.
Collapse
Affiliation(s)
- Akbar Anaeigoudari
- Department of Physiology, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Hamid Reza Mollaei
- Department of Medical Microbiology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Kazemi Arababadi
- Department of Laboratory Sciences, Faculty of Paramedicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Reza Nosratabadi
- Department of Medical Immunology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
- Department of Medical Immunology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
20
|
Ostaszewski M, Niarakis A, Mazein A, Kuperstein I, Phair R, Orta‐Resendiz A, Singh V, Aghamiri SS, Acencio ML, Glaab E, Ruepp A, Fobo G, Montrone C, Brauner B, Frishman G, Monraz Gómez LC, Somers J, Hoch M, Kumar Gupta S, Scheel J, Borlinghaus H, Czauderna T, Schreiber F, Montagud A, Ponce de Leon M, Funahashi A, Hiki Y, Hiroi N, Yamada TG, Dräger A, Renz A, Naveez M, Bocskei Z, Messina F, Börnigen D, Fergusson L, Conti M, Rameil M, Nakonecnij V, Vanhoefer J, Schmiester L, Wang M, Ackerman EE, Shoemaker JE, Zucker J, Oxford K, Teuton J, Kocakaya E, Summak GY, Hanspers K, Kutmon M, Coort S, Eijssen L, Ehrhart F, Rex DAB, Slenter D, Martens M, Pham N, Haw R, Jassal B, Matthews L, Orlic‐Milacic M, Senff Ribeiro A, Rothfels K, Shamovsky V, Stephan R, Sevilla C, Varusai T, Ravel J, Fraser R, Ortseifen V, Marchesi S, Gawron P, Smula E, Heirendt L, Satagopam V, Wu G, Riutta A, Golebiewski M, Owen S, Goble C, Hu X, Overall RW, Maier D, Bauch A, Gyori BM, Bachman JA, Vega C, Grouès V, Vazquez M, Porras P, Licata L, Iannuccelli M, Sacco F, Nesterova A, Yuryev A, de Waard A, Turei D, Luna A, Babur O, Soliman S, Valdeolivas A, Esteban‐Medina M, Peña‐Chilet M, Rian K, Helikar T, Puniya BL, Modos D, Treveil A, Olbei M, De Meulder B, Ballereau S, Dugourd A, Naldi A, Noël V, Calzone L, Sander C, Demir E, Korcsmaros T, Freeman TC, Augé F, Beckmann JS, Hasenauer J, Wolkenhauer O, Wilighagen EL, Pico AR, Evelo CT, Gillespie ME, Stein LD, Hermjakob H, D'Eustachio P, Saez‐Rodriguez J, Dopazo J, Valencia A, Kitano H, Barillot E, Auffray C, Balling R, Schneider R. COVID19 Disease Map, a computational knowledge repository of virus-host interaction mechanisms. Mol Syst Biol 2021; 17:e10387. [PMID: 34664389 PMCID: PMC8524328 DOI: 10.15252/msb.202110387] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 12/13/2022] Open
Abstract
We need to effectively combine the knowledge from surging literature with complex datasets to propose mechanistic models of SARS-CoV-2 infection, improving data interpretation and predicting key targets of intervention. Here, we describe a large-scale community effort to build an open access, interoperable and computable repository of COVID-19 molecular mechanisms. The COVID-19 Disease Map (C19DMap) is a graphical, interactive representation of disease-relevant molecular mechanisms linking many knowledge sources. Notably, it is a computational resource for graph-based analyses and disease modelling. To this end, we established a framework of tools, platforms and guidelines necessary for a multifaceted community of biocurators, domain experts, bioinformaticians and computational biologists. The diagrams of the C19DMap, curated from the literature, are integrated with relevant interaction and text mining databases. We demonstrate the application of network analysis and modelling approaches by concrete examples to highlight new testable hypotheses. This framework helps to find signatures of SARS-CoV-2 predisposition, treatment response or prioritisation of drug candidates. Such an approach may help deal with new waves of COVID-19 or similar pandemics in the long-term perspective.
Collapse
Affiliation(s)
- Marek Ostaszewski
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | - Anna Niarakis
- Université Paris‐SaclayLaboratoire Européen de Recherche pour la Polyarthrite rhumatoïde ‐ GenhotelUniv EvryEvryFrance
- Lifeware GroupInria Saclay‐Ile de FrancePalaiseauFrance
| | - Alexander Mazein
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | - Inna Kuperstein
- Institut CuriePSL Research UniversityParisFrance
- INSERMParisFrance
- MINES ParisTechPSL Research UniversityParisFrance
| | - Robert Phair
- Integrative Bioinformatics, Inc.Mountain ViewCAUSA
| | - Aurelio Orta‐Resendiz
- Institut PasteurUniversité de Paris, Unité HIVInflammation et PersistanceParisFrance
- Bio Sorbonne Paris CitéUniversité de ParisParisFrance
| | - Vidisha Singh
- Université Paris‐SaclayLaboratoire Européen de Recherche pour la Polyarthrite rhumatoïde ‐ GenhotelUniv EvryEvryFrance
| | - Sara Sadat Aghamiri
- Inserm‐ Institut national de la santé et de la recherche médicaleParisFrance
| | - Marcio Luis Acencio
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | - Enrico Glaab
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | - Andreas Ruepp
- Institute of Experimental Genetics (IEG)Helmholtz Zentrum München‐German Research Center for Environmental Health (GmbH)NeuherbergGermany
| | - Gisela Fobo
- Institute of Experimental Genetics (IEG)Helmholtz Zentrum München‐German Research Center for Environmental Health (GmbH)NeuherbergGermany
| | - Corinna Montrone
- Institute of Experimental Genetics (IEG)Helmholtz Zentrum München‐German Research Center for Environmental Health (GmbH)NeuherbergGermany
| | - Barbara Brauner
- Institute of Experimental Genetics (IEG)Helmholtz Zentrum München‐German Research Center for Environmental Health (GmbH)NeuherbergGermany
| | - Goar Frishman
- Institute of Experimental Genetics (IEG)Helmholtz Zentrum München‐German Research Center for Environmental Health (GmbH)NeuherbergGermany
| | - Luis Cristóbal Monraz Gómez
- Institut CuriePSL Research UniversityParisFrance
- INSERMParisFrance
- MINES ParisTechPSL Research UniversityParisFrance
| | - Julia Somers
- Department of Molecular and Medical GeneticsOregon Health & Sciences UniversityPortlandORUSA
| | - Matti Hoch
- Department of Systems Biology and BioinformaticsUniversity of RostockRostockGermany
| | | | - Julia Scheel
- Department of Systems Biology and BioinformaticsUniversity of RostockRostockGermany
| | - Hanna Borlinghaus
- Department of Computer and Information ScienceUniversity of KonstanzKonstanzGermany
| | - Tobias Czauderna
- Faculty of Information TechnologyDepartment of Human‐Centred ComputingMonash UniversityClaytonVic.Australia
| | - Falk Schreiber
- Department of Computer and Information ScienceUniversity of KonstanzKonstanzGermany
- Faculty of Information TechnologyDepartment of Human‐Centred ComputingMonash UniversityClaytonVic.Australia
| | | | | | - Akira Funahashi
- Department of Biosciences and InformaticsKeio UniversityYokohamaJapan
| | - Yusuke Hiki
- Department of Biosciences and InformaticsKeio UniversityYokohamaJapan
| | - Noriko Hiroi
- Graduate School of Media and GovernanceResearch Institute at SFCKeio UniversityKanagawaJapan
| | - Takahiro G Yamada
- Department of Biosciences and InformaticsKeio UniversityYokohamaJapan
| | - Andreas Dräger
- Computational Systems Biology of Infections and Antimicrobial‐Resistant PathogensInstitute for Bioinformatics and Medical Informatics (IBMI)University of TübingenTübingenGermany
- Department of Computer ScienceUniversity of TübingenTübingenGermany
- German Center for Infection Research (DZIF), partner siteTübingenGermany
| | - Alina Renz
- Computational Systems Biology of Infections and Antimicrobial‐Resistant PathogensInstitute for Bioinformatics and Medical Informatics (IBMI)University of TübingenTübingenGermany
- Department of Computer ScienceUniversity of TübingenTübingenGermany
| | - Muhammad Naveez
- Department of Systems Biology and BioinformaticsUniversity of RostockRostockGermany
- Institute of Applied Computer SystemsRiga Technical UniversityRigaLatvia
| | - Zsolt Bocskei
- Sanofi R&DTranslational SciencesChilly‐MazarinFrance
| | - Francesco Messina
- Dipartimento di Epidemiologia Ricerca Pre‐Clinica e Diagnostica AvanzataNational Institute for Infectious Diseases 'Lazzaro Spallanzani' I.R.C.C.S.RomeItaly
- COVID‐19 INMI Network Medicine for IDs Study GroupNational Institute for Infectious Diseases 'Lazzaro Spallanzani' I.R.C.C.SRomeItaly
| | - Daniela Börnigen
- Bioinformatics Core FacilityUniversitätsklinikum Hamburg‐EppendorfHamburgGermany
| | - Liam Fergusson
- Royal (Dick) School of Veterinary MedicineThe University of EdinburghEdinburghUK
| | - Marta Conti
- Faculty of Mathematics and Natural SciencesUniversity of BonnBonnGermany
| | - Marius Rameil
- Faculty of Mathematics and Natural SciencesUniversity of BonnBonnGermany
| | - Vanessa Nakonecnij
- Faculty of Mathematics and Natural SciencesUniversity of BonnBonnGermany
| | - Jakob Vanhoefer
- Faculty of Mathematics and Natural SciencesUniversity of BonnBonnGermany
| | - Leonard Schmiester
- Faculty of Mathematics and Natural SciencesUniversity of BonnBonnGermany
- Center for MathematicsChair of Mathematical Modeling of Biological SystemsTechnische Universität MünchenGarchingGermany
| | - Muying Wang
- Department of Chemical and Petroleum EngineeringUniversity of PittsburghPittsburghPAUSA
| | - Emily E Ackerman
- Department of Chemical and Petroleum EngineeringUniversity of PittsburghPittsburghPAUSA
| | - Jason E Shoemaker
- Department of Chemical and Petroleum EngineeringUniversity of PittsburghPittsburghPAUSA
- Department of Computational and Systems BiologyUniversity of PittsburghPittsburghPAUSA
| | | | | | | | | | | | - Kristina Hanspers
- Institute of Data Science and BiotechnologyGladstone InstitutesSan FranciscoCAUSA
| | - Martina Kutmon
- Department of Bioinformatics ‐ BiGCaTNUTRIMMaastricht UniversityMaastrichtThe Netherlands
- Maastricht Centre for Systems Biology (MaCSBio)Maastricht UniversityMaastrichtThe Netherlands
| | - Susan Coort
- Department of Bioinformatics ‐ BiGCaTNUTRIMMaastricht UniversityMaastrichtThe Netherlands
| | - Lars Eijssen
- Department of Bioinformatics ‐ BiGCaTNUTRIMMaastricht UniversityMaastrichtThe Netherlands
- Maastricht University Medical CentreMaastrichtThe Netherlands
| | - Friederike Ehrhart
- Department of Bioinformatics ‐ BiGCaTNUTRIMMaastricht UniversityMaastrichtThe Netherlands
- Maastricht University Medical CentreMaastrichtThe Netherlands
| | | | - Denise Slenter
- Department of Bioinformatics ‐ BiGCaTNUTRIMMaastricht UniversityMaastrichtThe Netherlands
| | - Marvin Martens
- Department of Bioinformatics ‐ BiGCaTNUTRIMMaastricht UniversityMaastrichtThe Netherlands
| | - Nhung Pham
- Department of Bioinformatics ‐ BiGCaTNUTRIMMaastricht UniversityMaastrichtThe Netherlands
| | - Robin Haw
- MaRS CentreOntario Institute for Cancer ResearchTorontoONCanada
| | - Bijay Jassal
- MaRS CentreOntario Institute for Cancer ResearchTorontoONCanada
| | | | | | - Andrea Senff Ribeiro
- MaRS CentreOntario Institute for Cancer ResearchTorontoONCanada
- Universidade Federal do ParanáCuritibaBrasil
| | - Karen Rothfels
- MaRS CentreOntario Institute for Cancer ResearchTorontoONCanada
| | | | - Ralf Stephan
- MaRS CentreOntario Institute for Cancer ResearchTorontoONCanada
| | - Cristoffer Sevilla
- European Bioinformatics Institute (EMBL‐EBI)European Molecular Biology LaboratoryHinxton, CambridgeshireUK
| | - Thawfeek Varusai
- European Bioinformatics Institute (EMBL‐EBI)European Molecular Biology LaboratoryHinxton, CambridgeshireUK
| | - Jean‐Marie Ravel
- INSERM UMR_S 1256Nutrition, Genetics, and Environmental Risk Exposure (NGERE)Faculty of Medicine of NancyUniversity of LorraineNancyFrance
- Laboratoire de génétique médicaleCHRU NancyNancyFrance
| | - Rupsha Fraser
- Queen's Medical Research InstituteThe University of EdinburghEdinburghUK
| | - Vera Ortseifen
- Senior Research Group in Genome Research of Industrial MicroorganismsCenter for BiotechnologyBielefeld UniversityBielefeldGermany
| | - Silvia Marchesi
- Department of Surgical ScienceUppsala UniversityUppsalaSweden
| | - Piotr Gawron
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
- Institute of Computing SciencePoznan University of TechnologyPoznanPoland
| | - Ewa Smula
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | - Laurent Heirendt
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | - Venkata Satagopam
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | - Guanming Wu
- Department of Medical Informatics and Clinical EpidemiologyOregon Health & Science UniversityPortlandORUSA
| | - Anders Riutta
- Institute of Data Science and BiotechnologyGladstone InstitutesSan FranciscoCAUSA
| | | | - Stuart Owen
- Department of Computer ScienceThe University of ManchesterManchesterUK
| | - Carole Goble
- Department of Computer ScienceThe University of ManchesterManchesterUK
| | - Xiaoming Hu
- Heidelberg Institute for Theoretical Studies (HITS)HeidelbergGermany
| | - Rupert W Overall
- German Center for Neurodegenerative Diseases (DZNE) DresdenDresdenGermany
- Center for Regenerative Therapies Dresden (CRTD)Technische Universität DresdenDresdenGermany
- Institute for BiologyHumboldt University of BerlinBerlinGermany
| | | | | | - Benjamin M Gyori
- Harvard Medical SchoolLaboratory of Systems PharmacologyBostonMAUSA
| | - John A Bachman
- Harvard Medical SchoolLaboratory of Systems PharmacologyBostonMAUSA
| | - Carlos Vega
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | - Valentin Grouès
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | | | - Pablo Porras
- European Bioinformatics Institute (EMBL‐EBI)European Molecular Biology LaboratoryHinxton, CambridgeshireUK
| | - Luana Licata
- Department of BiologyUniversity of Rome Tor VergataRomeItaly
| | | | - Francesca Sacco
- Department of BiologyUniversity of Rome Tor VergataRomeItaly
| | | | | | | | - Denes Turei
- Institute for Computational BiomedicineHeidelberg UniversityHeidelbergGermany
| | - Augustin Luna
- cBio Center, Divisions of Biostatistics and Computational BiologyDepartment of Data SciencesDana‐Farber Cancer InstituteBostonMAUSA
- Department of Cell BiologyHarvard Medical SchoolBostonMAUSA
| | - Ozgun Babur
- Computer Science DepartmentUniversity of Massachusetts BostonBostonMAUSA
| | | | - Alberto Valdeolivas
- Institute for Computational BiomedicineHeidelberg UniversityHeidelbergGermany
| | - Marina Esteban‐Medina
- Clinical Bioinformatics AreaFundación Progreso y Salud (FPS)Hospital Virgen del RocioSevillaSpain
- Computational Systems Medicine GroupInstitute of Biomedicine of Seville (IBIS)Hospital Virgen del RocioSevillaSpain
| | - Maria Peña‐Chilet
- Clinical Bioinformatics AreaFundación Progreso y Salud (FPS)Hospital Virgen del RocioSevillaSpain
- Computational Systems Medicine GroupInstitute of Biomedicine of Seville (IBIS)Hospital Virgen del RocioSevillaSpain
- Bioinformatics in Rare Diseases (BiER)Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)FPS, Hospital Virgen del RocíoSevillaSpain
| | - Kinza Rian
- Clinical Bioinformatics AreaFundación Progreso y Salud (FPS)Hospital Virgen del RocioSevillaSpain
- Computational Systems Medicine GroupInstitute of Biomedicine of Seville (IBIS)Hospital Virgen del RocioSevillaSpain
| | - Tomáš Helikar
- Department of BiochemistryUniversity of Nebraska‐LincolnLincolnNEUSA
| | | | - Dezso Modos
- Quadram Institute BioscienceNorwichUK
- Earlham InstituteNorwichUK
| | - Agatha Treveil
- Quadram Institute BioscienceNorwichUK
- Earlham InstituteNorwichUK
| | - Marton Olbei
- Quadram Institute BioscienceNorwichUK
- Earlham InstituteNorwichUK
| | | | - Stephane Ballereau
- Cancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
| | - Aurélien Dugourd
- Institute for Computational BiomedicineHeidelberg UniversityHeidelbergGermany
- Institute of Experimental Medicine and Systems BiologyFaculty of Medicine, RWTHAachen UniversityAachenGermany
| | | | - Vincent Noël
- Institut CuriePSL Research UniversityParisFrance
- INSERMParisFrance
- MINES ParisTechPSL Research UniversityParisFrance
| | - Laurence Calzone
- Institut CuriePSL Research UniversityParisFrance
- INSERMParisFrance
- MINES ParisTechPSL Research UniversityParisFrance
| | - Chris Sander
- cBio Center, Divisions of Biostatistics and Computational BiologyDepartment of Data SciencesDana‐Farber Cancer InstituteBostonMAUSA
- Department of Cell BiologyHarvard Medical SchoolBostonMAUSA
| | - Emek Demir
- Department of Molecular and Medical GeneticsOregon Health & Sciences UniversityPortlandORUSA
| | | | - Tom C Freeman
- The Roslin InstituteUniversity of EdinburghEdinburghUK
| | - Franck Augé
- Sanofi R&DTranslational SciencesChilly‐MazarinFrance
| | | | - Jan Hasenauer
- Helmholtz Zentrum München – German Research Center for Environmental HealthInstitute of Computational BiologyNeuherbergGermany
- Interdisciplinary Research Unit Mathematics and Life SciencesUniversity of BonnBonnGermany
| | - Olaf Wolkenhauer
- Department of Systems Biology and BioinformaticsUniversity of RostockRostockGermany
| | - Egon L Wilighagen
- Department of Bioinformatics ‐ BiGCaTNUTRIMMaastricht UniversityMaastrichtThe Netherlands
| | - Alexander R Pico
- Institute of Data Science and BiotechnologyGladstone InstitutesSan FranciscoCAUSA
| | - Chris T Evelo
- Department of Bioinformatics ‐ BiGCaTNUTRIMMaastricht UniversityMaastrichtThe Netherlands
- Maastricht Centre for Systems Biology (MaCSBio)Maastricht UniversityMaastrichtThe Netherlands
| | - Marc E Gillespie
- MaRS CentreOntario Institute for Cancer ResearchTorontoONCanada
- St. John’s University College of Pharmacy and Health SciencesQueensNYUSA
| | - Lincoln D Stein
- MaRS CentreOntario Institute for Cancer ResearchTorontoONCanada
- Department of Molecular GeneticsUniversity of TorontoTorontoONCanada
| | - Henning Hermjakob
- European Bioinformatics Institute (EMBL‐EBI)European Molecular Biology LaboratoryHinxton, CambridgeshireUK
| | | | | | - Joaquin Dopazo
- Clinical Bioinformatics AreaFundación Progreso y Salud (FPS)Hospital Virgen del RocioSevillaSpain
- Computational Systems Medicine GroupInstitute of Biomedicine of Seville (IBIS)Hospital Virgen del RocioSevillaSpain
- Bioinformatics in Rare Diseases (BiER)Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)FPS, Hospital Virgen del RocíoSevillaSpain
- FPS/ELIXIR‐esHospital Virgen del RocíoSevillaSpain
| | - Alfonso Valencia
- Barcelona Supercomputing Center (BSC)BarcelonaSpain
- Institució Catalana de Recerca i Estudis Avançats (ICREA)BarcelonaSpain
| | - Hiroaki Kitano
- Systems Biology InstituteTokyoJapan
- Okinawa Institute of Science and Technology Graduate SchoolOkinawaJapan
| | - Emmanuel Barillot
- Institut CuriePSL Research UniversityParisFrance
- INSERMParisFrance
- MINES ParisTechPSL Research UniversityParisFrance
| | - Charles Auffray
- Cancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
| | - Rudi Balling
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | - Reinhard Schneider
- Luxembourg Centre for Systems BiomedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | | |
Collapse
|
21
|
Verweyen EL, Schulert GS. Interfering with interferons: targeting the JAK-STAT pathway in complications of systemic juvenile idiopathic arthritis (SJIA). Rheumatology (Oxford) 2021; 61:926-935. [PMID: 34459891 PMCID: PMC9123899 DOI: 10.1093/rheumatology/keab673] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/27/2021] [Accepted: 08/23/2021] [Indexed: 02/06/2023] Open
Abstract
Systemic JIA (SJIA) is distinguished from other forms of JIA by the prevalence of the severe, life-threatening complications macrophage activation syndrome (SJIA-MAS) and lung disease (SJIA-LD). Alternative therapeutics are urgently needed, as disease pathogenesis diverges from what is observed in SJIA, and currently available biologics are insufficient. SJIA-MAS, defined by a cytokine storm and dysregulated proliferation of T-lymphocytes, and SJIA-LD which presents with lymphocytic interstitial inflammation and pulmonary alveolar proteinosis, are both thought to be driven by IFNs, in particular the type II IFN-γ. Involvement of IFNs and a possible crosstalk of type I IFNs with existing biologics indicate a distinct role for the JAK-STAT signalling pathway in the pathogenesis of SJIA-MAS and SJIA-LD. Here, we review this role of JAK-STATs and IFNs in SJIA complications and discuss how new insights of ongoing research are shaping future therapeutic advances in the form of JAK inhibitors and antibodies targeting IFNs.
Collapse
Affiliation(s)
- Emely L Verweyen
- Division of Rheumatology, Cincinnati Children's Hospital Medical Center
| | - Grant S Schulert
- Division of Rheumatology, Cincinnati Children's Hospital Medical Center,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA,Correspondence to: Grant Schulert, Division of Rheumatology, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati College of Medicine, MLC 4010, 3333 Burnet Avenue, Cincinnati, OH 45208, USA.
E-mail:
| |
Collapse
|
22
|
Mavian C, López-Bueno A, Martín R, Nitsche A, Alcamí A. Comparative Pathogenesis, Genomics and Phylogeography of Mousepox. Viruses 2021; 13:v13061146. [PMID: 34203773 PMCID: PMC8232671 DOI: 10.3390/v13061146] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/02/2021] [Accepted: 06/10/2021] [Indexed: 01/18/2023] Open
Abstract
Ectromelia virus (ECTV), the causative agent of mousepox, has threatened laboratory mouse colonies worldwide for almost a century. Mousepox has been valuable for the understanding of poxvirus pathogenesis and immune evasion. Here, we have monitored in parallel the pathogenesis of nine ECTVs in BALB/cJ mice and report the full-length genome sequence of eight novel ECTV isolates or strains, including the first ECTV isolated from a field mouse, ECTV-MouKre. This approach allowed us to identify several genes, absent in strains attenuated through serial passages in culture, that may play a role in virulence and a set of putative genes that may be involved in enhancing viral growth in vitro. We identified a putative strong inhibitor of the host inflammatory response in ECTV-MouKre, an isolate that did not cause local foot swelling and developed a moderate virulence. Most of the ECTVs, except ECTV-Hampstead, encode a truncated version of the P4c protein that impairs the recruitment of virions into the A-type inclusion bodies, and our data suggest that P4c may play a role in viral dissemination and transmission. This is the first comprehensive report that sheds light into the phylogenetic and geographic relationship of the worldwide outbreak dynamics for the ECTV species.
Collapse
Affiliation(s)
- Carla Mavian
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Campus de Cantoblanco, Universidad Autónoma de Madrid, Nicolás Cabrera 1, 28049 Madrid, Spain; (C.M.); (A.L.-B.); (R.M.)
| | - Alberto López-Bueno
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Campus de Cantoblanco, Universidad Autónoma de Madrid, Nicolás Cabrera 1, 28049 Madrid, Spain; (C.M.); (A.L.-B.); (R.M.)
| | - Rocío Martín
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Campus de Cantoblanco, Universidad Autónoma de Madrid, Nicolás Cabrera 1, 28049 Madrid, Spain; (C.M.); (A.L.-B.); (R.M.)
| | - Andreas Nitsche
- Centre for Biological Threats and Special Pathogens, Highly Pathogenic Viruses (ZBS1), Robert Koch Institute, 13353 Berlin, Germany;
| | - Antonio Alcamí
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Campus de Cantoblanco, Universidad Autónoma de Madrid, Nicolás Cabrera 1, 28049 Madrid, Spain; (C.M.); (A.L.-B.); (R.M.)
- Correspondence:
| |
Collapse
|
23
|
Melo-Silva CR, Alves-Peixoto P, Heath N, Tang L, Montoya B, Knudson CJ, Stotesbury C, Ferez M, Wong E, Sigal LJ. Resistance to lethal ectromelia virus infection requires Type I interferon receptor in natural killer cells and monocytes but not in adaptive immune or parenchymal cells. PLoS Pathog 2021; 17:e1009593. [PMID: 34015056 PMCID: PMC8172060 DOI: 10.1371/journal.ppat.1009593] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 06/02/2021] [Accepted: 04/28/2021] [Indexed: 11/18/2022] Open
Abstract
Type I interferons (IFN-I) are antiviral cytokines that signal through the ubiquitous IFN-I receptor (IFNAR). Following footpad infection with ectromelia virus (ECTV), a mouse-specific pathogen, C57BL/6 (B6) mice survive without disease, while B6 mice broadly deficient in IFNAR succumb rapidly. We now show that for survival to ECTV, only hematopoietic cells require IFNAR expression. Survival to ECTV specifically requires IFNAR in both natural killer (NK) cells and monocytes. However, intrinsic IFNAR signaling is not essential for adaptive immune cell responses or to directly protect non-hematopoietic cells such as hepatocytes, which are principal ECTV targets. Mechanistically, IFNAR-deficient NK cells have reduced cytolytic function, while lack of IFNAR in monocytes dampens IFN-I production and hastens virus dissemination. Thus, during a pathogenic viral infection, IFN-I coordinates innate immunity by stimulating monocytes in a positive feedback loop and by inducing NK cell cytolytic function.
Collapse
Affiliation(s)
- Carolina R. Melo-Silva
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Pedro Alves-Peixoto
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Natasha Heath
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Lingjuan Tang
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Brian Montoya
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Cory J. Knudson
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Colby Stotesbury
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Maria Ferez
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Eric Wong
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Luis J. Sigal
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
24
|
Knudson CJ, Alves-Peixoto P, Muramatsu H, Stotesbury C, Tang L, Lin PJC, Tam YK, Weissman D, Pardi N, Sigal LJ. Lipid-nanoparticle-encapsulated mRNA vaccines induce protective memory CD8 T cells against a lethal viral infection. Mol Ther 2021; 29:2769-2781. [PMID: 33992803 DOI: 10.1016/j.ymthe.2021.05.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/21/2021] [Accepted: 05/11/2021] [Indexed: 12/24/2022] Open
Abstract
It is well established that memory CD8 T cells protect susceptible strains of mice from mousepox, a lethal viral disease caused by ectromelia virus (ECTV), the murine counterpart to human variola virus. While mRNA vaccines induce protective antibody (Ab) responses, it is unknown whether they also induce protective memory CD8 T cells. We now show that immunization with different doses of unmodified or N(1)-methylpseudouridine-modified mRNA (modified mRNA) in lipid nanoparticles (LNP) encoding the ECTV gene EVM158 induced similarly strong CD8 T cell responses to the epitope TSYKFESV, albeit unmodified mRNA-LNP had adverse effects at the inoculation site. A single immunization with 10 μg modified mRNA-LNP protected most susceptible mice from mousepox, and booster vaccination increased the memory CD8 T cell pool, providing full protection. Moreover, modified mRNA-LNP encoding TSYKFESV appended to green fluorescent protein (GFP) protected against wild-type ECTV infection while lymphocytic choriomeningitis virus glycoprotein (GP) modified mRNA-LNP protected against ECTV expressing GP epitopes. Thus, modified mRNA-LNP can be used to create protective CD8 T cell-based vaccines against viral infections.
Collapse
Affiliation(s)
- Cory J Knudson
- Department of Microbiology and Immunology, Bluemle Life Science Building, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Pedro Alves-Peixoto
- Department of Microbiology and Immunology, Bluemle Life Science Building, Thomas Jefferson University, Philadelphia, PA 19107, USA; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga 4710-057, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães 4806-909, Portugal
| | - Hiromi Muramatsu
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Colby Stotesbury
- Department of Microbiology and Immunology, Bluemle Life Science Building, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Lingjuan Tang
- Department of Microbiology and Immunology, Bluemle Life Science Building, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | | | - Ying K Tam
- Acuitas Therapeutics, Vancouver, BC V6T 1Z3, Canada
| | - Drew Weissman
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Norbert Pardi
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Luis J Sigal
- Department of Microbiology and Immunology, Bluemle Life Science Building, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
25
|
Wangsanut T, Brann KR, Adcox HE, Carlyon JA. Orientia tsutsugamushi modulates cellular levels of NF-κB inhibitor p105. PLoS Negl Trop Dis 2021; 15:e0009339. [PMID: 33857149 PMCID: PMC8078813 DOI: 10.1371/journal.pntd.0009339] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 04/27/2021] [Accepted: 03/29/2021] [Indexed: 11/26/2022] Open
Abstract
Background Scrub typhus is a neglected tropical disease that threatens more than one billion people. If antibiotic therapy is delayed, often due to mis- or late diagnosis, the case fatality rate can increase considerably. Scrub typhus is caused by the obligate intracellular bacterium, Orientia tsutsugamushi, which invades phagocytes and endothelial cells in vivo and diverse tissue culture cell types in vitro. The ability of O. tsutsugamushi to replicate in the cytoplasm indicates that it has evolved to counter eukaryotic host cell immune defense mechanisms. The transcription factor, NF-κB, is a tightly regulated initiator of proinflammatory and antimicrobial responses. Typically, the inhibitory proteins p105 and IκBα sequester the NF-κB p50:p65 heterodimer in the cytoplasm. Canonical activation of NF-κB via TNFα involves IKKβ-mediated serine phosphorylation of IκBα and p105, which leads to their degradation and enables NF-κB nuclear translocation. A portion of p105 is also processed into p50. O. tsutsugamushi impairs NF-κB translocation into the nucleus, but how it does so is incompletely defined. Principal findings Western blot, densitometry, and quantitative RT-PCR analyses of O. tsutsugamushi infected host cells were used to determine if the pathogen’s ability to inhibit NF-κB is linked to modulation of p105. Results demonstrate that p105 levels are elevated several-fold in O. tsutsugamushi infected HeLa and RF/6A cells with only a nominal increase in p50. The O. tsutsugamushi-stimulated increase in p105 is bacterial dose- and protein synthesis-dependent, but does not occur at the level of host cell transcription. While TNFα-induced phosphorylation of p105 serine 932 proceeds unhindered in infected cells, p105 levels remain elevated and NF-κB p65 is retained in the cytoplasm. Conclusions O. tsutsugamushi specifically stabilizes p105 to inhibit the canonical NF-κB pathway, which advances understanding of how it counters host immunity to establish infection. Scrub typhus is a neglected disease that can be fatal and occurs predominantly in the Asia-Pacific, one of the most densely populated regions of the world. Notably, cases continue to emerge outside this area. The etiologic agent is Orientia tsutsugamushi, a bacterial pathogen that infects certain leukocytes and cells that line blood vessels in animals and humans. The success of O. tsutsugamushi to colonize these cells is at least partially attributable to its ability to counter host immunity. In this study, we demonstrate that O. tsutsugamushi stabilizes p105, a mammalian inhibitor of the transcription factor, NF-κB, which is otherwise key for activating proinflammatory and antimicrobial gene expression. O. tsutsugamushi is the first example of a bacterium that inhibits NF-κB by promoting elevated levels of p105 and impairing its degradation. Our findings provide fundamental information that helps explain how this important pathogen has evolved to stealthily establish infection in host cells.
Collapse
Affiliation(s)
- Tanaporn Wangsanut
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, Unites States of America
| | - Katelynn R. Brann
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, Unites States of America
| | - Haley E. Adcox
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, Unites States of America
| | - Jason A. Carlyon
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, School of Medicine, Richmond, Virginia, Unites States of America
- * E-mail:
| |
Collapse
|
26
|
Temporal dynamics of a CSF1R signaling gene regulatory network involved in epilepsy. PLoS Comput Biol 2021; 17:e1008854. [PMID: 33819288 PMCID: PMC8057615 DOI: 10.1371/journal.pcbi.1008854] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 04/20/2021] [Accepted: 03/04/2021] [Indexed: 01/16/2023] Open
Abstract
Colony Stimulating Factor 1 Receptor (CSF1R) is a potential target for anti-epileptic drugs. However, inhibition of CSF1R is not well tolerated by patients, thereby prompting the need for alternative targets. To develop a framework for identification of such alternatives, we here develop a mathematical model of a pro-inflammatory gene regulatory network (GRN) involved in epilepsy and centered around CSF1R. This GRN comprises validated transcriptional and post-transcriptional regulations involving STAT1, STAT3, NFκB, IL6R, CSF3R, IRF8, PU1, C/EBPα, TNFR1, CSF1 and CSF1R. The model was calibrated on mRNA levels of all GRN components in lipopolysaccharide (LPS)-treated mouse microglial BV-2 cells, and allowed to predict that STAT1 and STAT3 have the strongest impact on the expression of the other GRN components. Microglial BV-2 cells were selected because, the modules from which the GRN was deduced are enriched for microglial marker genes. The function of STAT1 and STAT3 in the GRN was experimentally validated in BV-2 cells. Further, in silico analysis of the GRN dynamics predicted that a pro-inflammatory stimulus can induce irreversible bistability whereby the expression level of GRN components occurs as two distinct states. The irreversibility of the switch may enforce the need for chronic inhibition of the CSF1R GRN in order to achieve therapeutic benefit. The cell-to-cell heterogeneity driven by the bistability may cause variable therapeutic response. In conclusion, our modeling approach uncovered a GRN controlling CSF1R that is predominantly regulated by STAT1 and STAT3. Irreversible inflammation-induced bistability and cell-to-cell heterogeneity of the GRN provide a theoretical foundation to the need for chronic GRN control and the limited potential for disease modification via inhibition of CSF1R. Epilepsy is associated with the induction of complex molecular inflammatory processes. A better understanding of these molecular mechanisms is crucial to optimize therapeutic options. Here, we identified a gene regulatory network (GRN) involved in epilepsy that is controlled by inflammation and which regulates the expression and function of Colony Stimulating Factor 1 receptor (CSF1R), a therapeutic target for anti-epileptic drugs. Using mathematical modeling and experiments with cultured cells, we found that two of eleven components of the network, namely STAT1 and STAT3, exert a tight control on all other components. In addition, we found that inflammation can induce an irreversible switch in the expression of all components of the network, and can cause high cell-to-cell variability. Our findings provide a framework explaining why chronic, not acute, anti-inflammatory treatment is necessary to modulate the network and why drugs targeting CSF1R have limited therapeutic potential.
Collapse
|
27
|
Choubey SK, Nachiappan M, Richard M, Chitra JP, Jeyakanthan J. Structural and functional insights of STAT2-NS5 interaction for the identification of NS5 antagonist – An approach for restoring interferon signaling. Comput Biol Chem 2020; 88:107332. [DOI: 10.1016/j.compbiolchem.2020.107332] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 07/13/2020] [Accepted: 07/16/2020] [Indexed: 11/16/2022]
|
28
|
Wong E, Montoya B, Stotesbury C, Ferez M, Xu RH, Sigal LJ. Langerhans Cells Orchestrate the Protective Antiviral Innate Immune Response in the Lymph Node. Cell Rep 2020; 29:3047-3059.e3. [PMID: 31801072 PMCID: PMC6927544 DOI: 10.1016/j.celrep.2019.10.118] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 09/17/2019] [Accepted: 10/29/2019] [Indexed: 12/22/2022] Open
Abstract
During disseminating viral infections, a swift innate immune response (IIR) in the draining lymph node (dLN) that restricts systemic viral spread is critical for optimal resistance to disease. However, it is unclear how this IIR is orchestrated. We show that after footpad infection of mice with ectromelia virus, dendritic cells (DCs) highly expressing major histocompatibility complex class II (MHC class IIhi DCs), including CD207+ epidermal Langerhans cells (LCs), CD103+CD207+ double-positive dermal DCs (DP-DCs), and CD103−CD207− double-negative dermal DCs (DN-DCs) migrate to the dLN from the skin carrying virus. MHC class IIhi DCs, predominantly LCs and DP-DCs, are the first cells upregulating IIR cytokines in the dLN. Preventing MHC class IIhi DC migration or depletion of LCs, but not DP-DC deficiency, suppresses the IIR in the dLN and results in high viral lethality. Therefore, LCs are the architects of an early IIR in the dLN that is critical for optimal resistance to a disseminating viral infection. Wong et al. show that by producing chemokines that recruit monocytes and by upregulating NKG2D ligands that activate ILCs, Langerhans cells are responsible for the innate immune cascade in the lymph node that is critical for survival of infection with a disseminating virus.
Collapse
Affiliation(s)
- Eric Wong
- Department of Microbiology and Immunology, Thomas Jefferson University, BLSB 709 233 South 10(th) Street, Philadelphia, PA 19107, USA
| | - Brian Montoya
- Department of Microbiology and Immunology, Thomas Jefferson University, BLSB 709 233 South 10(th) Street, Philadelphia, PA 19107, USA
| | - Colby Stotesbury
- Department of Microbiology and Immunology, Thomas Jefferson University, BLSB 709 233 South 10(th) Street, Philadelphia, PA 19107, USA
| | - Maria Ferez
- Department of Microbiology and Immunology, Thomas Jefferson University, BLSB 709 233 South 10(th) Street, Philadelphia, PA 19107, USA
| | - Ren-Huan Xu
- Immune Cell Development and Host Defense Program, Research Institute of Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | - Luis J Sigal
- Department of Microbiology and Immunology, Thomas Jefferson University, BLSB 709 233 South 10(th) Street, Philadelphia, PA 19107, USA.
| |
Collapse
|
29
|
Hernáez B, Alonso G, Georgana I, El-Jesr M, Martín R, Shair KHY, Fischer C, Sauer S, Maluquer de Motes C, Alcamí A. Viral cGAMP nuclease reveals the essential role of DNA sensing in protection against acute lethal virus infection. SCIENCE ADVANCES 2020; 6:6/38/eabb4565. [PMID: 32948585 PMCID: PMC7500930 DOI: 10.1126/sciadv.abb4565] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 08/03/2020] [Indexed: 06/11/2023]
Abstract
Cells contain numerous immune sensors to detect virus infection. The cyclic GMP-AMP (cGAMP) synthase (cGAS) recognizes cytosolic DNA and activates innate immune responses via stimulator of interferon genes (STING), but the impact of DNA sensing pathways on host protective responses has not been fully defined. We demonstrate that cGAS/STING activation is required to resist lethal poxvirus infection. We identified viral Schlafen (vSlfn) as the main STING inhibitor, and ectromelia virus was severely attenuated in the absence of vSlfn. Both vSlfn-mediated virulence and STING inhibitory activity were mapped to the recently discovered poxin cGAMP nuclease domain. Animals were protected from subcutaneous, respiratory, and intravenous infection in the absence of vSlfn, and interferon was the main antiviral protective mechanism controlled by the DNA sensing pathway. Our findings support the idea that manipulation of DNA sensing is an efficient therapeutic strategy in diseases triggered by viral infection or tissue damage-mediated release of self-DNA.
Collapse
Affiliation(s)
- Bruno Hernáez
- Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid), Madrid, Spain
| | - Graciela Alonso
- Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid), Madrid, Spain
| | - Iliana Georgana
- Department of Microbial Sciences, University of Surrey, Guildford, UK
| | - Misbah El-Jesr
- Department of Microbial Sciences, University of Surrey, Guildford, UK
| | - Rocío Martín
- Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid), Madrid, Spain
| | - Kathy H Y Shair
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Cornelius Fischer
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
- Max Delbrück Center for Molecular Medicine, Berlin Institute of Health, Berlin, Germany
| | - Sascha Sauer
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
- Max Delbrück Center for Molecular Medicine, Berlin Institute of Health, Berlin, Germany
| | | | - Antonio Alcamí
- Centro de Biología Molecular Severo Ochoa (Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid), Madrid, Spain.
- Department of Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
30
|
Lu Y, Zhang L. DNA-Sensing Antiviral Innate Immunity in Poxvirus Infection. Front Immunol 2020; 11:1637. [PMID: 32983084 PMCID: PMC7483915 DOI: 10.3389/fimmu.2020.01637] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 06/18/2020] [Indexed: 12/16/2022] Open
Abstract
As pattern recognition receptors, cytosolic DNA sensors quickly induce an effective innate immune response. Poxvirus, a large DNA virus, is capable of evading the host antiviral innate immune response. In this review, we summarize the latest studies on how poxvirus is sensed by the host innate immune system and how poxvirus-encoded proteins antagonize DNA sensors. A comprehensive understanding of the interplay between poxvirus and DNA-sensing antiviral immune responses of the host will contribute to the development of new antiviral therapies and vaccines in the future.
Collapse
Affiliation(s)
- Yue Lu
- Department of Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University, Jinan, China.,Institute of Basic Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.,Key Laboratory for Biotech-Drugs of National Health Commission, Jinan, China.,Key Laboratory for Rare and Uncommon Diseases of Shandong Province, Jinan, China
| | - Leiliang Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University, Jinan, China.,Institute of Basic Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.,Key Laboratory for Biotech-Drugs of National Health Commission, Jinan, China.,Key Laboratory for Rare and Uncommon Diseases of Shandong Province, Jinan, China.,Science and Technology Innovation Center, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
31
|
Putzke S, Feldhues E, Heep I, Ilg T, Lamprecht A. Cationic lipid/pDNA complex formation as potential generic method to generate specific IRF pathway stimulators. Eur J Pharm Biopharm 2020; 155:112-121. [PMID: 32798666 DOI: 10.1016/j.ejpb.2020.08.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 05/21/2020] [Accepted: 08/10/2020] [Indexed: 10/23/2022]
Abstract
Cationic liposome - CpG DNA complexes (lipoplexes) are known as stimulators of innate immunity via Toll-like receptor 9 (TLR9)-triggered activation of the nuclear factor kappa B (NF-κB) pathway. More recent reports suggest that DNA lipoplexes also engage DNA sensors in the cytosol leading to the stimulation of the interferon response factor (IRF) pathway. In this study a range of lipoplexes were formulated by using an invariable helper lipid, three different cationic lipids (DOTAP, DOTMA and DDA) and three different CpG-containing plasmids of different sizes. These lipoplexes exhibited similar hydrodynamic diameters, zeta-potentials and plasmid loading rates, despite the different lipid blends and CpG-containing plasmids. Binding and uptake of liposomal lipids by J774.A1 macrophages and JAWSII dendritic cells increased significantly (up to 4-fold) upon lipoplex formation. Cellular plasmid DNA uptake via lipoplexes compared to naked DNA was increased up to 18-fold. Analysis of signal transduction pathway activation in J774-DUAL™ reporter cells by liposomes or naked CpG plasmid DNA compared to their derived lipoplexes showed only minor activation of the NF-κB pathway, while the IRF pathway displayed massive activation factors of up to 46-fold. DOTAP- and DOTMA lipoplexes also led to massive interferon-alpha and -beta secretion of J774A.1 macrophages and JAWSII dendritic cells, which is a hallmark of IRF pathway activation. Cellular distribution studies on DOTAP lipoplexes suggest delivery of plasmid DNA via vesicular compartments into the cytosol. Taken together, the CpG plasmid DNA lipoplexes generated in this study appear to selectively stimulate DNA receptors activating the IRF pathway, while bypassing TLR9 and NF-κB activation.
Collapse
Affiliation(s)
- Simone Putzke
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, 53121 Bonn, Germany; Bayer Animal Health GmbH, 40789 Monheim am Rhein, Germany
| | | | - Iris Heep
- Bayer Animal Health GmbH, 40789 Monheim am Rhein, Germany
| | - Thomas Ilg
- Bayer Animal Health GmbH, 40789 Monheim am Rhein, Germany
| | - Alf Lamprecht
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, 53121 Bonn, Germany.
| |
Collapse
|
32
|
Stotesbury C, Wong EB, Tang L, Montoya B, Knudson CJ, Melo‐Silva CR, Sigal LJ. Defective early innate immune response to ectromelia virus in the draining lymph nodes of aged mice due to impaired dendritic cell accumulation. Aging Cell 2020; 19:e13170. [PMID: 32657004 PMCID: PMC7433008 DOI: 10.1111/acel.13170] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 04/11/2020] [Accepted: 05/13/2020] [Indexed: 12/14/2022] Open
Abstract
It is known that aging decreases natural resistance to viral diseases due to dysfunctional innate and adaptive immune responses, but the nature of these dysfunctions, particularly in regard to innate immunity, is not well understood. We have previously shown that C57BL/6J (B6) mice lose their natural resistance to footpad infection with ectromelia virus (ECTV) due to impaired maturation and recruitment of natural killer (NK) cells to the draining popliteal lymph node (dLN). More recently, we have also shown that in young B6 mice infected with ECTV, the recruitment of NK cells is dependent on a complex cascade whereby migratory dendritic cells (mDCs) traffic from the skin to the dLN, where they produce CCL2 and CCL7 to recruit inflammatory monocytes (iMOs). In the dLN, mDCs also upregulate NKG2D ligands to induce interferon gamma (IFN-γ) expression by group 1 innate lymphoid cells (G1-ILCs), mostly NK in cells but also some ILC1. In response to the IFN-γ, the incoming uninfected iMOs secret CXCL9 to recruit the critical NK cells. Here, we show that in aged B6 mice, the trafficking of mDCs to the dLN in response to ECTV is decreased, resulting in impaired IFN-γ expression by G1-ILCs, reduced accumulation of iMOs, and attenuated CXCL9 production by iMOs, which likely contributes to decrease in NK cell recruitment. Together, these data indicate that defects in the mDC response to viral infection during aging result in a reduced innate immune response in the dLN and contribute to increased susceptibility to viral disease in the aged.
Collapse
Affiliation(s)
- Colby Stotesbury
- Department of Microbiology and Immunology Thomas Jefferson University Philadelphia PA USA
| | - Eric B. Wong
- Department of Microbiology and Immunology Thomas Jefferson University Philadelphia PA USA
| | - Lingjuan Tang
- Department of Microbiology and Immunology Thomas Jefferson University Philadelphia PA USA
| | - Brian Montoya
- Department of Microbiology and Immunology Thomas Jefferson University Philadelphia PA USA
| | - Cory J. Knudson
- Department of Microbiology and Immunology Thomas Jefferson University Philadelphia PA USA
| | - Carolina R. Melo‐Silva
- Department of Microbiology and Immunology Thomas Jefferson University Philadelphia PA USA
| | - Luis J. Sigal
- Department of Microbiology and Immunology Thomas Jefferson University Philadelphia PA USA
| |
Collapse
|
33
|
Wang Z, Qin J, Zhao J, Li J, Li D, Popp M, Popp F, Alakus H, Kong B, Dong Q, Nelson PJ, Zhao Y, Bruns CJ. Inflammatory IFIT3 renders chemotherapy resistance by regulating post-translational modification of VDAC2 in pancreatic cancer. Theranostics 2020; 10:7178-7192. [PMID: 32641986 PMCID: PMC7330856 DOI: 10.7150/thno.43093] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 04/20/2020] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers worldwide and effective therapy remains a challenge. IFIT3 is an interferon-stimulated gene with antiviral and pro-inflammatory functions. Our previous work has shown that high expression of IFIT3 is correlated with poor survival in PDAC patients who receive chemotherapy suggesting a link between IFIT3 and chemotherapy resistance in PDAC. However, the exact role and molecular mechanism of IFIT3 in chemotherapy resistance in PDAC has been unclear. Methods: A group of transcriptome datasets were downloaded and analyzed for the characterization of IFIT3 in PDAC. Highly metastatic PDAC cell line L3.6pl and patient-derived primary cell TBO368 were used and IFIT3 knockdown and the corresponding knockin cells were established for in vitro studies. Chemotherapy-induced apoptosis, ROS production, confocal immunofluorescence, subcellular fractionation, chromatin-immunoprecipitation, co-immunoprecipitation and mass spectrometry analysis were determined to further explore the biological role of IFIT3 in chemotherapy resistance of PDAC. Results: Based on PDAC transcriptome data, we show that IFIT3 expression is associated with the squamous molecular subtype of PDAC and an increase in inflammatory response and apoptosis pathways. We further identify a crucial role for IFIT3 in the regulation of mitochondria-associated apoptosis during chemotherapy. Knockdown of IFIT3 attenuates the chemotherapy resistance of PDAC cells to gemcitabine, paclitaxel, and FOLFIRINOX regimen treatments, independent of individual chemotherapy regimens. While IFIT3 overexpression was found to promote drug resistance. Co-immunoprecipitation identified a direct interaction between IFIT3 and the mitochondrial channel protein VDAC2, an important regulator of mitochondria-associated apoptosis. It was subsequently found that IFIT3 regulates the post-translational modification-O-GlcNAcylation of VDAC2 by stabilizing the interaction of VDAC2 with O-GlcNAc transferase. Increased O-GlcNAcylation of VDAC2 protected PDAC cells from chemotherapy induced apoptosis. Conclusions: These results effectively demonstrate a central mechanism by which IFIT3 expression can affect chemotherapy resistance in PDAC. Targeting IFIT3/VDAC2 may represent a novel strategy to sensitize aggressive forms of pancreatic cancer to conventional chemotherapy regimens.
Collapse
Affiliation(s)
- Zhefang Wang
- Department of General, Visceral, Tumor and Transplantation Surgery, University Hospital Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Jie Qin
- Department of General, Visceral, Tumor and Transplantation Surgery, University Hospital Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Jiangang Zhao
- Department of General, Visceral, Tumor and Transplantation Surgery, University Hospital Cologne, Kerpener Straße 62, 50937 Cologne, Germany
- Department of General, Visceral und Vascular Surgery, Ludwig-Maximilian-University (LMU), 81377 Munich, Germany
| | - Jiahui Li
- Department of General, Visceral, Tumor and Transplantation Surgery, University Hospital Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Dai Li
- Department of General, Visceral, Tumor and Transplantation Surgery, University Hospital Cologne, Kerpener Straße 62, 50937 Cologne, Germany
- Department of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, PR China
| | - Marie Popp
- Department of General, Visceral, Tumor and Transplantation Surgery, University Hospital Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Felix Popp
- Department of General, Visceral, Tumor and Transplantation Surgery, University Hospital Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Hakan Alakus
- Department of General, Visceral, Tumor and Transplantation Surgery, University Hospital Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Bo Kong
- Department of Surgery, Klinikum Rechts der Isar, Technische Universität München, Munich, Germany
| | - Qiongzhu Dong
- Department of General Surgery, Huashan Hospital & Cancer Metastasis Institute & Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Peter J. Nelson
- Medizinische Klinik und Poliklinik IV, University of Munich, Munich, Germany
| | - Yue Zhao
- Department of General, Visceral, Tumor and Transplantation Surgery, University Hospital Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Christiane J. Bruns
- Department of General, Visceral, Tumor and Transplantation Surgery, University Hospital Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| |
Collapse
|
34
|
Namineni S, O'Connor T, Faure-Dupuy S, Johansen P, Riedl T, Liu K, Xu H, Singh I, Shinde P, Li F, Pandyra A, Sharma P, Ringelhan M, Muschaweckh A, Borst K, Blank P, Lampl S, Neuhaus K, Durantel D, Farhat R, Weber A, Lenggenhager D, Kündig TM, Staeheli P, Protzer U, Wohlleber D, Holzmann B, Binder M, Breuhahn K, Assmus LM, Nattermann J, Abdullah Z, Rolland M, Dejardin E, Lang PA, Lang KS, Karin M, Lucifora J, Kalinke U, Knolle PA, Heikenwalder M. A dual role for hepatocyte-intrinsic canonical NF-κB signaling in virus control. J Hepatol 2020; 72:960-975. [PMID: 31954207 DOI: 10.1016/j.jhep.2019.12.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 12/02/2019] [Accepted: 12/11/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Hepatic innate immune control of viral infections has largely been attributed to Kupffer cells, the liver-resident macrophages. However, hepatocytes, the parenchymal cells of the liver, also possess potent immunological functions in addition to their known metabolic functions. Owing to their abundance in the liver and known immunological functions, we aimed to investigate the direct antiviral mechanisms employed by hepatocytes. METHODS Using lymphocytic choriomeningitis virus (LCMV) as a model of liver infection, we first assessed the role of myeloid cells by depletion prior to infection. We investigated the role of hepatocyte-intrinsic innate immune signaling by infecting mice lacking canonical NF-κB signaling (IkkβΔHep) specifically in hepatocytes. In addition, mice lacking hepatocyte-specific interferon-α/β signaling-(IfnarΔHep), or interferon-α/β signaling in myeloid cells-(IfnarΔMyel) were infected. RESULTS Here, we demonstrate that LCMV activates NF-κB signaling in hepatocytes. LCMV-triggered NF-κB activation in hepatocytes did not depend on Kupffer cells or TNFR1 signaling but rather on Toll-like receptor signaling. LCMV-infected IkkβΔHep livers displayed strongly elevated viral titers due to LCMV accumulation within hepatocytes, reduced interferon-stimulated gene (ISG) expression, delayed intrahepatic immune cell influx and delayed intrahepatic LCMV-specific CD8+ T cell responses. Notably, viral clearance and ISG expression were also reduced in LCMV-infected primary hepatocytes lacking IKKβ, demonstrating a hepatocyte-intrinsic effect. Similar to livers of IkkβΔHep mice, enhanced hepatocytic LCMV accumulation was observed in livers of IfnarΔHep mice, whereas IfnarΔMyel mice were able to control LCMV infection. Hepatocytic NF-κB signaling was also required for efficient ISG induction in HDV-infected dHepaRG cells and interferon-α/β-mediated inhibition of HBV replication in vitro. CONCLUSIONS Together, these data show that hepatocyte-intrinsic NF-κB is a vital amplifier of interferon-α/β signaling, which is pivotal for strong early ISG responses, immune cell infiltration and hepatic viral clearance. LAY SUMMARY Innate immune cells have been ascribed a primary role in controlling viral clearance upon hepatic infections. We identified a novel dual role for NF-κB signaling in infected hepatocytes which was crucial for maximizing interferon responses and initiating adaptive immunity, thereby efficiently controlling hepatic virus replication.
Collapse
Affiliation(s)
- Sukumar Namineni
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany; Institute of Virology, Technical University of Munich and Helmholtz Zentrum München, Schneckenburgerstrasse 8, 81675 Munich, Germany; Institute of Molecular Immunology and Experimental Oncology, Technical University of Munich, Ismaningerstraße 22, 81675 Munich, Germany
| | - Tracy O'Connor
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany; Institute of Molecular Immunology and Experimental Oncology, Technical University of Munich, Ismaningerstraße 22, 81675 Munich, Germany
| | - Suzanne Faure-Dupuy
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Pål Johansen
- Department of Dermatology, University Hospital Zurich and University of Zurich, Gloriastrasse 31, 8091 Zurich, Switzerland
| | - Tobias Riedl
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kaijing Liu
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Haifeng Xu
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, Essen 45147, Germany
| | - Indrabahadur Singh
- Emmy Noether Research Group Epigenetic Machineries and Cancer, Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Prashant Shinde
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, Universitätstr.1, 40225 Düsseldorf, Germany
| | - Fanghui Li
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, Essen 45147, Germany
| | - Aleksandra Pandyra
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, Essen 45147, Germany
| | - Piyush Sharma
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, Essen 45147, Germany; Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA, 38105
| | - Marc Ringelhan
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany; Institute of Virology, Technical University of Munich and Helmholtz Zentrum München, Schneckenburgerstrasse 8, 81675 Munich, Germany; Department of Internal Medicine II, University Hospital rechts der Isar, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Andreas Muschaweckh
- Klinikum rechts der Isar, Department of Neurology, Technical University of Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Katharina Borst
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hanover Medical School and the Helmholtz Centre for Infection Research, Brunswick, Germany
| | - Patrick Blank
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hanover Medical School and the Helmholtz Centre for Infection Research, Brunswick, Germany
| | - Sandra Lampl
- Institute of Molecular Immunology and Experimental Oncology, Technical University of Munich, Ismaningerstraße 22, 81675 Munich, Germany
| | - Katharina Neuhaus
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - David Durantel
- INSERM, U1052, Cancer Research Center of Lyon (CRCL), Université de Lyon (UCBL1), CNRS UMR 5286, Centre Léon Bérard, Lyon, France
| | - Rayan Farhat
- INSERM, U1052, Cancer Research Center of Lyon (CRCL), Université de Lyon (UCBL1), CNRS UMR 5286, Centre Léon Bérard, Lyon, France
| | - Achim Weber
- Department of Pathology and Molecular Pathology, University Hospital of Zurich, 8091 Zurich, Switzerland
| | - Daniela Lenggenhager
- Department of Pathology and Molecular Pathology, University Hospital of Zurich, 8091 Zurich, Switzerland
| | - Thomas M Kündig
- Department of Dermatology, University Hospital Zurich and University of Zurich, Gloriastrasse 31, 8091 Zurich, Switzerland
| | - Peter Staeheli
- Institute of Virology, University of Freiburg, Freiburg, Germany
| | - Ulrike Protzer
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany; Institute of Virology, Technical University of Munich and Helmholtz Zentrum München, Schneckenburgerstrasse 8, 81675 Munich, Germany
| | - Dirk Wohlleber
- Institute of Molecular Immunology and Experimental Oncology, Technical University of Munich, Ismaningerstraße 22, 81675 Munich, Germany
| | - Bernhard Holzmann
- Department of Surgery, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Marco Binder
- Research Group "Dynamics of Early Viral Infection and the Innate Antiviral Response", Division Virus-Associated Carcinogenesis (F170), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Kai Breuhahn
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Jacob Nattermann
- Department of Internal Medicine, University of Bonn, Bonn, Germany
| | | | - Maude Rolland
- Laboratory of Molecular Immunology and Signal Transduction, GIGA-Institute, University of Liège, 4000 Liège, Belgium
| | - Emmanuel Dejardin
- Laboratory of Molecular Immunology and Signal Transduction, GIGA-Institute, University of Liège, 4000 Liège, Belgium
| | - Philipp A Lang
- Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, Universitätstr.1, 40225 Düsseldorf, Germany
| | - Karl S Lang
- Institute of Immunology, Medical Faculty, University of Duisburg-Essen, Hufelandstr. 55, Essen 45147, Germany
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego (UCSD), 9500 Gilman Drive, La Jolla, California 92093, USA
| | - Julie Lucifora
- INSERM, U1052, Cancer Research Center of Lyon (CRCL), Université de Lyon (UCBL1), CNRS UMR 5286, Centre Léon Bérard, Lyon, France
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Hanover Medical School and the Helmholtz Centre for Infection Research, Brunswick, Germany
| | - Percy A Knolle
- Institute of Molecular Immunology and Experimental Oncology, Technical University of Munich, Ismaningerstraße 22, 81675 Munich, Germany
| | - Mathias Heikenwalder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany; Institute of Virology, Technical University of Munich and Helmholtz Zentrum München, Schneckenburgerstrasse 8, 81675 Munich, Germany; Institute of Molecular Immunology and Experimental Oncology, Technical University of Munich, Ismaningerstraße 22, 81675 Munich, Germany.
| |
Collapse
|
35
|
Targeting of the cGAS-STING system by DNA viruses. Biochem Pharmacol 2020; 174:113831. [DOI: 10.1016/j.bcp.2020.113831] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 01/24/2020] [Indexed: 12/15/2022]
|
36
|
Stotesbury C, Alves-Peixoto P, Montoya B, Ferez M, Nair S, Snyder CM, Zhang S, Knudson CJ, Sigal LJ. α2β1 Integrin Is Required for Optimal NK Cell Proliferation during Viral Infection but Not for Acquisition of Effector Functions or NK Cell-Mediated Virus Control. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:1582-1591. [PMID: 32015010 PMCID: PMC7065959 DOI: 10.4049/jimmunol.1900927] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 01/05/2020] [Indexed: 01/13/2023]
Abstract
NK cells play an important role in antiviral resistance. The integrin α2, which dimerizes with integrin β1, distinguishes NK cells from innate lymphoid cells 1 and other leukocytes. Despite its use as an NK cell marker, little is known about the role of α2β1 in NK cell biology. In this study, we show that in mice α2β1 deficiency does not alter the balance of NK cell/ innate lymphoid cell 1 generation and slightly decreases the number of NK cells in the bone marrow and spleen without affecting NK cell maturation. NK cells deficient in α2β1 had no impairment at entering or distributing within the draining lymph node of ectromelia virus (ECTV)-infected mice or at becoming effectors but proliferated poorly in response to ECTV and did not increase in numbers following infection with mouse CMV (MCMV). Still, α2β1-deficient NK cells efficiently protected from lethal mousepox and controlled MCMV titers in the spleen. Thus, α2β1 is required for optimal NK cell proliferation but is dispensable for protection against ECTV and MCMV, two well-established models of viral infection in which NK cells are known to be important.
Collapse
Affiliation(s)
- Colby Stotesbury
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Pedro Alves-Peixoto
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Brian Montoya
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Maria Ferez
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Savita Nair
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Christopher M Snyder
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Shunchuan Zhang
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Cory J Knudson
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Luis J Sigal
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107
| |
Collapse
|
37
|
Mourik BC, de Steenwinkel JEM, de Knegt GJ, Huizinga R, Verbon A, Ottenhoff THM, van Soolingen D, Leenen PJM. Mycobacterium tuberculosis clinical isolates of the Beijing and East-African Indian lineage induce fundamentally different host responses in mice compared to H37Rv. Sci Rep 2019; 9:19922. [PMID: 31882653 PMCID: PMC6934500 DOI: 10.1038/s41598-019-56300-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 11/14/2019] [Indexed: 11/15/2022] Open
Abstract
Substantial differences exist in virulence among Mycobacterium tuberculosis strains in preclinical TB models. In this study we show how virulence affects host responses in mice during the first four weeks of infection with a mycobacterial strain belonging to the Beijing, East-African-Indian or Euro-American lineage. BALB/c mice were infected with clinical isolates of the Beijing-1585 strain or the East-African Indian (EAI)-1627 strain and host responses were compared to mice infected with the non-clinical H37Rv strain of the Euro-American lineage. We found that H37Rv induced a ‘classical’ T-cell influx with high IFN-γ levels, while Beijing-1585 and EAI-1627 induced an influx of B-cells into the lungs together with elevated pulmonary IL-4 protein levels. Myeloid cells in the lungs appeared functionally impaired upon infection with Beijing-1585 and EAI-1627 with reduced iNOS and IL-12 expression levels compared to H37Rv infection. This impairment might be related to significantly reduced expression in the bone marrow of IFN-γ, TNF-α and IFN-β in mice infected with Beijing-1585 and EAI-1627, which could be detected from the third day post infection onwards. Our findings suggest that increased virulence of two clinical isolates compared to H37Rv is associated with a fundamentally different systemic immune response, which already can be detected early during infection.
Collapse
Affiliation(s)
- Bas C Mourik
- Department Medical Microbiology & Infectious Diseases, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jurriaan E M de Steenwinkel
- Department Medical Microbiology & Infectious Diseases, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Gerjo J de Knegt
- Department Medical Microbiology & Infectious Diseases, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ruth Huizinga
- Department of Immunology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Annelies Verbon
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Dick van Soolingen
- National Tuberculosis Reference Laboratory, National Institute of Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Pieter J M Leenen
- Department of Immunology, Erasmus University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
38
|
Wong EB, Montoya B, Ferez M, Stotesbury C, Sigal LJ. Resistance to ectromelia virus infection requires cGAS in bone marrow-derived cells which can be bypassed with cGAMP therapy. PLoS Pathog 2019; 15:e1008239. [PMID: 31877196 PMCID: PMC6974301 DOI: 10.1371/journal.ppat.1008239] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 01/21/2020] [Accepted: 11/25/2019] [Indexed: 01/07/2023] Open
Abstract
Cells sensing infection produce Type I interferons (IFN-I) to stimulate Interferon Stimulated Genes (ISGs) that confer resistance to viruses. During lympho-hematogenous spread of the mouse pathogen ectromelia virus (ECTV), the adaptor STING and the transcription factor IRF7 are required for IFN-I and ISG induction and resistance to ECTV. However, it is unknown which cells sense ECTV and which pathogen recognition receptor (PRR) upstream of STING is required for IFN-I and ISG induction. We found that cyclic-GMP-AMP (cGAMP) synthase (cGAS), a DNA-sensing PRR, is required in bone marrow-derived (BMD) but not in other cells for IFN-I and ISG induction and for resistance to lethal mousepox. Also, local administration of cGAMP, the product of cGAS that activates STING, rescues cGAS but not IRF7 or IFN-I receptor deficient mice from mousepox. Thus, sensing of infection by BMD cells via cGAS and IRF7 is critical for resistance to a lethal viral disease in a natural host. During primary acute systemic viral infections, cells sensing virus through Pathogen Recognition Receptors (PRR) can produce Type I interferons (IFN-I) to induce an anti-viral state that curbs viral spread and protect from viral disease. The dissection of the specific cells, receptors and downstream pathways required for IFN-I production during viral infection in vivo is necessary to improve anti-viral therapies. In this study, we demonstrated that the cytosolic PRR cGAS in hematopoietic cells but not in parenchymal cells is required for protection against ectromelia virus, the archetype for viruses that spread through the lympho-hematogenous route. We also show that cGAS deficiency can be bypassed by local administration of cyclic-GMP-AMP (cGAMP) by inducing IFN-I only in the skin and in the presence of virus. Our study provides novel insights into the cGAS signaling pathway and highlights the potential of cGAMP as an efficient anti-viral treatment.
Collapse
Affiliation(s)
- Eric B. Wong
- Thomas Jefferson University, Department of Microbiology and Immunology, Philadelphia, Pennsylvania, United States of America
| | - Brian Montoya
- Thomas Jefferson University, Department of Microbiology and Immunology, Philadelphia, Pennsylvania, United States of America
| | - Maria Ferez
- Thomas Jefferson University, Department of Microbiology and Immunology, Philadelphia, Pennsylvania, United States of America
| | - Colby Stotesbury
- Thomas Jefferson University, Department of Microbiology and Immunology, Philadelphia, Pennsylvania, United States of America
| | - Luis J. Sigal
- Thomas Jefferson University, Department of Microbiology and Immunology, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
39
|
Owen KL, Brockwell NK, Parker BS. JAK-STAT Signaling: A Double-Edged Sword of Immune Regulation and Cancer Progression. Cancers (Basel) 2019; 11:E2002. [PMID: 31842362 PMCID: PMC6966445 DOI: 10.3390/cancers11122002] [Citation(s) in RCA: 365] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/06/2019] [Accepted: 12/09/2019] [Indexed: 02/07/2023] Open
Abstract
Janus kinase-signal transducer and activator of transcription (JAK-STAT) signaling mediates almost all immune regulatory processes, including those that are involved in tumor cell recognition and tumor-driven immune escape. Antitumor immune responses are largely driven by STAT1 and STAT2 induction of type I and II interferons (IFNs) and the downstream programs IFNs potentiate. Conversely, STAT3 has been widely linked to cancer cell survival, immunosuppression, and sustained inflammation in the tumor microenvironment. The discovery of JAK-STAT cross-regulatory mechanisms, post-translational control, and non-canonical signal transduction has added a new level of complexity to JAK-STAT governance over tumor initiation and progression. Endeavors to better understand the vast effects of JAK-STAT signaling on antitumor immunity have unearthed a wide range of targets, including oncogenes, miRNAs, and other co-regulatory factors, which direct specific phenotypical outcomes subsequent to JAK-STAT stimulation. Yet, the rapidly expanding field of therapeutic developments aimed to resolve JAK-STAT aberrations commonly reported in a multitude of cancers has been marred by off-target effects. Here, we discuss JAK-STAT biology in the context of immunity and cancer, the consequences of pathway perturbations and current therapeutic interventions, to provide insight and consideration for future targeting innovations.
Collapse
Affiliation(s)
- Katie L. Owen
- Cancer Immunology and Therapeutics Programs, Peter MacCallum Cancer Centre, VIC, Melbourne 3000, Australia;
- Sir Peter MacCallum Department of Oncology, University of Melbourne, VIC, Parkville 3052, Australia
| | - Natasha K. Brockwell
- Cancer Immunology and Therapeutics Programs, Peter MacCallum Cancer Centre, VIC, Melbourne 3000, Australia;
- Sir Peter MacCallum Department of Oncology, University of Melbourne, VIC, Parkville 3052, Australia
| | - Belinda S. Parker
- Cancer Immunology and Therapeutics Programs, Peter MacCallum Cancer Centre, VIC, Melbourne 3000, Australia;
- Sir Peter MacCallum Department of Oncology, University of Melbourne, VIC, Parkville 3052, Australia
| |
Collapse
|
40
|
Wong E, Xu RH, Rubio D, Lev A, Stotesbury C, Fang M, Sigal LJ. Migratory Dendritic Cells, Group 1 Innate Lymphoid Cells, and Inflammatory Monocytes Collaborate to Recruit NK Cells to the Virus-Infected Lymph Node. Cell Rep 2019; 24:142-154. [PMID: 29972776 DOI: 10.1016/j.celrep.2018.06.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 04/30/2018] [Accepted: 06/01/2018] [Indexed: 11/17/2022] Open
Abstract
Circulating natural killer (NK) cells help protect the host from lympho-hematogenous acute viral diseases by rapidly entering draining lymph nodes (dLNs) to curb virus dissemination. Here, we identify a highly choreographed mechanism underlying this process. Using footpad infection with ectromelia virus, a pathogenic DNA virus of mice, we show that TLR9/MyD88 sensing induces NKG2D ligands in virus-infected, skin-derived migratory dendritic cells (mDCs) to induce production of IFN-γ by classical NK cells and other types of group 1 innate lymphoid cells (ILCs) already in dLNs, via NKG2D. Uninfected inflammatory monocytes, also recruited to dLNs by mDCs in a TLR9/MyD88-dependent manner, respond to IFN-γ by secreting CXCL9 for optimal CXCR3-dependent recruitment of circulating NK cells. This work unveils a TLR9/MyD88-dependent mechanism whereby in dLNs, three cell types-mDCs, group 1 ILCs (mostly NK cells), and inflammatory monocytes-coordinate the recruitment of protective circulating NK cells to dLNs.
Collapse
Affiliation(s)
- Eric Wong
- Department of Microbiology and Immunology, Thomas Jefferson University, BLSB 709, 233 South 10(th) Street, Philadelphia, PA 19107, USA
| | - Ren-Huan Xu
- Immune Cell Development and Host Defense Program, Research Institute of Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | - Daniel Rubio
- Immune Cell Development and Host Defense Program, Research Institute of Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | - Avital Lev
- Immune Cell Development and Host Defense Program, Research Institute of Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | - Colby Stotesbury
- Department of Microbiology and Immunology, Thomas Jefferson University, BLSB 709, 233 South 10(th) Street, Philadelphia, PA 19107, USA
| | - Min Fang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, 100101, China
| | - Luis J Sigal
- Department of Microbiology and Immunology, Thomas Jefferson University, BLSB 709, 233 South 10(th) Street, Philadelphia, PA 19107, USA.
| |
Collapse
|
41
|
The role of MicroRNAs on endoplasmic reticulum stress in myocardial ischemia and cardiac hypertrophy. Pharmacol Res 2019; 150:104516. [DOI: 10.1016/j.phrs.2019.104516] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 09/12/2019] [Accepted: 10/29/2019] [Indexed: 12/22/2022]
|
42
|
Joyce MA, Berry-Wynne KM, dos Santos T, Addison WR, McFarlane N, Hobman T, Tyrrell DL. HCV and flaviviruses hijack cellular mechanisms for nuclear STAT2 degradation: Up-regulation of PDLIM2 suppresses the innate immune response. PLoS Pathog 2019; 15:e1007949. [PMID: 31374104 PMCID: PMC6677295 DOI: 10.1371/journal.ppat.1007949] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 06/29/2019] [Indexed: 12/22/2022] Open
Abstract
Host encounters with viruses lead to an innate immune response that must be rapid and broadly targeted but also tightly regulated to avoid the detrimental effects of unregulated interferon expression. Viral stimulation of host negative regulatory mechanisms is an alternate method of suppressing the host innate immune response. We examined three key mediators of the innate immune response: NF-KB, STAT1 and STAT2 during HCV infection in order to investigate the paradoxical induction of an innate immune response by HCV despite a multitude of mechanisms combating the host response. During infection, we find that all three are repressed only in HCV infected cells but not in uninfected bystander cells, both in vivo in chimeric mouse livers and in cultured Huh7.5 cells after IFNα treatment. We show here that HCV and Flaviviruses suppress the innate immune response by upregulation of PDLIM2, independent of the host interferon response. We show PDLIM2 is an E3 ubiquitin ligase that also acts to stimulate nuclear degradation of STAT2. Interferon dependent relocalization of STAT1/2 to the nucleus leads to PDLIM2 ubiquitination of STAT2 but not STAT1 and the proteasome-dependent degradation of STAT2, predominantly within the nucleus. CRISPR/Cas9 knockout of PDLIM2 results in increased levels of STAT2 following IFNα treatment, retention of STAT2 within the nucleus of HCV infected cells after IFNα stimulation, increased interferon response, and increased resistance to infection by several flaviviruses, indicating that PDLIM2 is a global regulator of the interferon response. The response of cells to an invading pathogen must be swift and well controlled because of the detrimental effects of chronic inflammation. However, viruses often hijack host control mechanisms. HCV and flaviviruses are known to suppress the innate immune response in cells by a variety of mechanisms. This study clarifies and expands a specific cellular mechanism for global control of the antiviral response after the induction of interferon expression. It shows how several viruses hijack this control mechanism to suppress the innate interferon response.
Collapse
Affiliation(s)
- Michael A. Joyce
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
- * E-mail: (MAJ); (DLT)
| | - Karyn M. Berry-Wynne
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - Theodore dos Santos
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - William R. Addison
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - Nicola McFarlane
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - Tom Hobman
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
- Department of Cell Biology, University of Alberta, Edmonton, Alberta, Canada
| | - D. Lorne Tyrrell
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
- * E-mail: (MAJ); (DLT)
| |
Collapse
|
43
|
Abstract
In the absence of an intact interferon (IFN) response, mammals may be susceptible to lethal viral infection. IFNs are secreted cytokines that activate a signal transduction cascade leading to the induction of hundreds of interferon-stimulated genes (ISGs). Remarkably, approximately 10% of the genes in the human genome have the potential to be regulated by IFNs. What do all of these genes do? It is a complex question without a simple answer. From decades of research, we know that many of the protein products encoded by these ISGs work alone or in concert to achieve one or more cellular outcomes, including antiviral defense, antiproliferative activities, and stimulation of adaptive immunity. The focus of this review is the antiviral activities of the IFN/ISG system. This includes general paradigms of ISG function, supported by specific examples in the literature, as well as methodologies to identify and characterize ISG function.
Collapse
Affiliation(s)
- John W Schoggins
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA;
| |
Collapse
|
44
|
Guo J, Li S, Li Y, Yan C, Wan Q, Wang Z. HSP90 inhibitor 17-AAG prevents apoptosis of cardiomyocytes via miR-93-dependent mitigation of endoplasmic reticulum stress. J Cell Biochem 2019; 120:7888-7896. [PMID: 30556167 DOI: 10.1002/jcb.28064] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 10/22/2018] [Indexed: 01/24/2023]
Abstract
Heart failure accounts for substantial morbidity and mortality worldwide. Accumulating evidence suggests that aberrant cardiac cell death caused by endoplasmic reticulum stress (ERS) is often associated with structural or functional cardiac abnormalities that lead to insufficient cardiac output. The detailed molecular mechanism underlying the pathological death of cardiomyocytes still remains poorly understood. We found that 17-AAG (tanespimycin), an HSP90 (heat shock protein 90) inhibitor often used to kill cancer cells, could potently inhibit tunicamycin-induced ERS and the downstream nuclear factor kappa B activity in neonatal rat cardiomyocytes, leading to diminished apoptotic signaling and thus enhanced cell survival. Interestingly, the antiapoptotic effect of 17-AAG on cardiomyocytes required normal expression of miR-93, an oncogenic microRNA known to promote cell survival and growth. Our study implicated a new pharmacological role of 17-AAG in supporting the miR-93-associated oncogenic signaling to prevent the pathological death of cardiomyocytes. The results opened opportunities for exploring new strategies in the development of therapeutic agents.
Collapse
Affiliation(s)
- Jingjing Guo
- Department of Cardiology, Huaihe Hospital, Henan University, Kaifeng City, Henan, China
| | - Shengnan Li
- Department of Cardiology, Huaihe Hospital, Henan University, Kaifeng City, Henan, China
| | - Yanming Li
- Department of Cardiology, Huaihe Hospital, Henan University, Kaifeng City, Henan, China
| | - Chenyun Yan
- Department of Cardiology, Huaihe Hospital, Henan University, Kaifeng City, Henan, China
| | - Qilin Wan
- Department of Cardiology, Huaihe Hospital, Henan University, Kaifeng City, Henan, China
| | - Zhizhong Wang
- Department of Cardiology, Huaihe Hospital, Henan University, Kaifeng City, Henan, China
| |
Collapse
|
45
|
Saidu NEB, Kavian N, Leroy K, Jacob C, Nicco C, Batteux F, Alexandre J. Dimethyl fumarate, a two-edged drug: Current status and future directions. Med Res Rev 2019; 39:1923-1952. [PMID: 30756407 DOI: 10.1002/med.21567] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/22/2019] [Accepted: 01/24/2019] [Indexed: 12/11/2022]
Abstract
Dimethyl fumarate (DMF) is a fumaric acid ester registered for the treatment of relapsing-remitting multiple sclerosis (RRMS). It induces protein succination leading to inactivation of cysteine-rich proteins. It was first shown to possess cytoprotective and antioxidant effects in noncancer models, which appeared related to the induction of the nuclear factor erythroid 2 (NF-E2)-related factor 2 (NRF2) pathway. DMF also displays antitumor activity in several cellular and mice models. Recently, we showed that the anticancer mechanism of DMF is dose-dependent and is paradoxically related to the decrease in the nuclear translocation of NRF2. Some other studies performed indicate also the potential role of DMF in cancers, which are dependent on the NRF2 antioxidant and cellular detoxification program, such as KRAS-mutated lung adenocarcinoma. It, however, seems that DMF has multiple biological effects as it has been shown to also inhibit the transcription factor nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), thus blocking downstream targets that may be involved in the development and progression of inflammatory cascades leading to various disease processes, including tumors, lymphomas, diabetic retinopathy, arthritis, and psoriasis. Herein, we present the current status and future directions of the use of DMF in various diseases models with particular emphases on its targeting of specific intracellular signal transduction cascades in cancer; to shed some light on its possible mode of action.
Collapse
Affiliation(s)
- Nathaniel Edward Bennett Saidu
- Department of Development, Reproduction and Cancer, Paris Descartes University, Sorbonne Paris Cité, INSERM U1016, Cochin Institute, CARPEM, Paris, France.,Division of Molecular Medicine, Institut Ruđer Bošković, Zagreb, Croatia
| | - Niloufar Kavian
- Department of Development, Reproduction and Cancer, Paris Descartes University, Sorbonne Paris Cité, INSERM U1016, Cochin Institute, CARPEM, Paris, France.,Department of Immunology, Cochin Hospital, AP-HP, Paris, France.,Division of Public Health Laboratory Sciences, HKU Pasteur Research Pole, University of Hong Kong, Hong Kong, SAR China
| | - Karen Leroy
- Department of Development, Reproduction and Cancer, Paris Descartes University, Sorbonne Paris Cité, INSERM U1016, Cochin Institute, CARPEM, Paris, France.,Department of Molecular Genetics, Cochin Hospital, AP-HP, Paris, France
| | - Claus Jacob
- Division of Bioorganic Chemistry, University of Saarland, Saarbruecken, Germany
| | - Carole Nicco
- Department of Development, Reproduction and Cancer, Paris Descartes University, Sorbonne Paris Cité, INSERM U1016, Cochin Institute, CARPEM, Paris, France
| | - Frédéric Batteux
- Department of Development, Reproduction and Cancer, Paris Descartes University, Sorbonne Paris Cité, INSERM U1016, Cochin Institute, CARPEM, Paris, France.,Department of Immunology, Cochin Hospital, AP-HP, Paris, France
| | - Jérôme Alexandre
- Department of Development, Reproduction and Cancer, Paris Descartes University, Sorbonne Paris Cité, INSERM U1016, Cochin Institute, CARPEM, Paris, France.,Department of Medical Oncology, Cochin Hospital, AP-HP, Paris, France
| |
Collapse
|
46
|
Zhao C, Zhang Y, Popel AS. Mechanistic Computational Models of MicroRNA-Mediated Signaling Networks in Human Diseases. Int J Mol Sci 2019; 20:E421. [PMID: 30669429 PMCID: PMC6358731 DOI: 10.3390/ijms20020421] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 01/14/2019] [Accepted: 01/15/2019] [Indexed: 12/17/2022] Open
Abstract
MicroRNAs (miRs) are endogenous non-coding RNA molecules that play important roles in human health and disease by regulating gene expression and cellular processes. In recent years, with the increasing scientific knowledge and new discovery of miRs and their gene targets, as well as the plentiful experimental evidence that shows dysregulation of miRs in a wide variety of human diseases, the computational modeling approach has emerged as an effective tool to help researchers identify novel functional associations between differential miR expression and diseases, dissect the phenotypic expression patterns of miRs in gene regulatory networks, and elucidate the critical roles of miRs in the modulation of disease pathways from mechanistic and quantitative perspectives. Here we will review the recent systems biology studies that employed different kinetic modeling techniques to provide mechanistic insights relating to the regulatory function and therapeutic potential of miRs in human diseases. Some of the key computational aspects to be discussed in detail in this review include (i) models of miR-mediated network motifs in the regulation of gene expression, (ii) models of miR biogenesis and miR⁻target interactions, and (iii) the incorporation of such models into complex disease pathways in order to generate mechanistic, molecular- and systems-level understanding of pathophysiology. Other related bioinformatics tools such as computational platforms that predict miR-disease associations will also be discussed, and we will provide perspectives on the challenges and opportunities in the future development and translational application of data-driven systems biology models that involve miRs and their regulatory pathways in human diseases.
Collapse
Affiliation(s)
- Chen Zhao
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Yu Zhang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
47
|
A virus-encoded type I interferon decoy receptor enables evasion of host immunity through cell-surface binding. Nat Commun 2018; 9:5440. [PMID: 30575728 PMCID: PMC6303335 DOI: 10.1038/s41467-018-07772-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 11/21/2018] [Indexed: 12/12/2022] Open
Abstract
Soluble cytokine decoy receptors are potent immune modulatory reagents with therapeutic applications. Some virus-encoded secreted cytokine receptors interact with glycosaminoglycans expressed at the cell surface, but the biological significance of this activity in vivo is poorly understood. Here, we show the type I interferon binding protein (IFNα/βBP) encoded by vaccinia and ectromelia viruses requires of this cell binding activity to confer full virulence to these viruses and to retain immunomodulatory activity. Expression of a variant form of the IFNα/βBP that inhibits IFN activity, but does not interact with cell surface glycosaminoglycans, results in highly attenuated viruses with a virulence similar to that of the IFNα/βBP deletion mutant viruses. Transcriptomics analysis and infection of IFN receptor-deficient mice confirmed that the control of IFN activity is the main function of the IFNα/βBP in vivo. We propose that retention of secreted cytokine receptors at the cell surface may largely enhance their immunomodulatory activity.
Collapse
|
48
|
Platanitis E, Decker T. Regulatory Networks Involving STATs, IRFs, and NFκB in Inflammation. Front Immunol 2018; 9:2542. [PMID: 30483250 PMCID: PMC6242948 DOI: 10.3389/fimmu.2018.02542] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 10/16/2018] [Indexed: 01/10/2023] Open
Abstract
Cells engaging in inflammation undergo drastic changes of their transcriptomes. In order to tailor these alterations in gene expression to the requirements of the inflammatory process, tight and coordinate regulation of gene expression by environmental cues, microbial or danger-associated molecules or cytokines, are mandatory. The transcriptional response is set off by signal-regulated transcription factors (SRTFs) at the receiving end of pathways originating at pattern recognition- and cytokine receptors. These interact with a genome that has been set for an appropriate response by prior activity of pioneer or lineage determining transcription factors (LDTFs). The same types of transcription factors are also critical determinants of the changes in chromatin landscapes and transcriptomes that specify potential consequences of inflammation: tissue repair, training, and tolerance. Here we focus on the role of three families of SRTFs in inflammation and its sequels: signal transducers and activators of transcription (STATs), interferon regulatory factors (IRFs), and nuclear factor κB (NFκB). We describe recent findings about their interactions and about their networking with LDTFs. Our aim is to provide a snapshot of a highly dynamic research area.
Collapse
Affiliation(s)
- Ekaterini Platanitis
- Max F. Perutz Laboratories, Department of Microbiology, Immunobiology and Genetics, University of Vienna, Vienna, Austria
| | - Thomas Decker
- Max F. Perutz Laboratories, Department of Microbiology, Immunobiology and Genetics, University of Vienna, Vienna, Austria
| |
Collapse
|
49
|
Novel Class of Viral Ankyrin Proteins Targeting the Host E3 Ubiquitin Ligase Cullin-2. J Virol 2018; 92:JVI.01374-18. [PMID: 30258003 PMCID: PMC6232478 DOI: 10.1128/jvi.01374-18] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 09/11/2018] [Indexed: 02/07/2023] Open
Abstract
Ankyrin repeat (ANK) domains are among the most abundant motifs in eukaryotic proteins. ANK proteins are rare amongst viruses, with the exception of poxviruses, which presumably acquired them from the host via horizontal gene transfer. The architecture of poxvirus ANK proteins is, however, different from that of their cellular counterparts, and this precludes a direct acquisition event. Here we combine bioinformatics analysis and quantitative proteomics to discover a new class of viral ANK proteins with a domain organization that relates to cellular ANK proteins. These noncanonical viral ANK proteins, termed ANK/BC, interact with host Cullin-2 via a C-terminal BC box resembling that of cellular Cullin-2 substrate adaptors such as the von Hippel-Lindau protein. Mutagenesis of the BC box-like sequence abrogates binding to Cullin-2, whereas fusion of this motif to an ANK-only protein confers Cullin-2 association. We demonstrated that these viral ANK/BC proteins are potent immunomodulatory proteins suppressing the activation of the proinflammatory transcription factors NF-κB and interferon (IFN)-responsive factor 3 (IRF-3) and the production of cytokines and chemokines, including interferon, and that association with Cullin-2 is required for optimal inhibitory activity. ANK/BC proteins exist in several orthopoxviruses and cluster into 2 closely related orthologue groups in a phylogenetic lineage that is separate from that of canonical ANK/F-box proteins. Given the existence of cellular proteins with similar architecture, viral ANK/BC proteins may be closely related to the original ANK gene acquired by an ancestral orthopoxvirus. These findings uncover a novel viral strategy to antagonize innate immunity and shed light on the origin of the poxviral ANK protein family.IMPORTANCE Viruses encode multiple proteins aimed at modulating cellular homeostasis and antagonizing the host antiviral response. Most of these genes were originally acquired from the host and subsequently adapted to benefit the virus. ANK proteins are common in eukaryotes but are unusual amongst viruses, with the exception of poxviruses, where they represent one of the largest protein families. We report here the existence of a new class of viral ANK proteins, termed ANK/BC, that provide new insights into the origin of poxvirus ANK proteins. ANK/BC proteins target the host E3 ubiquitin ligase Cullin-2 via a C-terminal BC box domain and are potent suppressors of the production of inflammatory cytokines, including interferon. The existence of cellular ANK proteins whose architecture is similar suggests the acquisition of a host ANK/BC gene by an ancestral orthopoxvirus and its subsequent duplication and adaptation to widen the repertoire of immune evasion strategies.
Collapse
|
50
|
Slfn2 Regulates Type I Interferon Responses by Modulating the NF-κB Pathway. Mol Cell Biol 2018; 38:MCB.00053-18. [PMID: 29866656 DOI: 10.1128/mcb.00053-18] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 05/23/2018] [Indexed: 11/20/2022] Open
Abstract
Although members of the Slfn family have been implicated in the regulation of type I interferon (IFN) responses, the mechanisms by which they mediate their effects remain unknown. In the present study, we provide evidence that targeted disruption of the Slfn2 gene leads to increased transcription of IFN-stimulated genes (ISGs) and enhanced type I IFN-mediated antiviral responses. We demonstrate that Slfn2 interacts with protein phosphatase 6 regulatory subunit 1 (PPP6R1), leading to reduced type I IFN-induced activation of nuclear factor kappa B (NF-κB) signaling, resulting in reduced expression of ISGs. Altogether, these data suggest a novel mechanism by which Slfn2 controls ISG expression and provide evidence for a critical role for Slfn2 in the regulation of IFN-mediated biological responses.
Collapse
|