1
|
Swaraj S, Tripathi S. Interference without interferon: interferon-independent induction of interferon-stimulated genes and its role in cellular innate immunity. mBio 2024; 15:e0258224. [PMID: 39302126 PMCID: PMC11481898 DOI: 10.1128/mbio.02582-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024] Open
Abstract
Interferons (IFNs) are multifaceted proteins that play pivotal roles in orchestrating robust antiviral immune responses and modulating the intricate landscape of host immunity. The major signaling pathway activated by IFNs is the JAK/STAT (Janus kinase/signal transducer and activator of transcription) pathway, which leads to the transcription of a battery of genes, collectively known as IFN-stimulated genes (ISGs). While the well-established role of IFNs in coordinating the innate immune response against viral infections is widely acknowledged, recent years have provided a more distinct comprehension of the functional significance attributed to non-canonical, IFN-independent induction of ISGs. In this review, we summarize the non-conventional signaling pathways of ISG induction. These alternative pathways offer new avenues for developing antiviral strategies or immunomodulation in various diseases.
Collapse
Affiliation(s)
- Shachee Swaraj
- Emerging Viral Pathogens Laboratory, Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru, India
- Microbiology & Cell Biology Department, Biological Sciences Division, Indian Institute of Science, Bengaluru, India
| | - Shashank Tripathi
- Emerging Viral Pathogens Laboratory, Centre for Infectious Disease Research, Indian Institute of Science, Bengaluru, India
- Microbiology & Cell Biology Department, Biological Sciences Division, Indian Institute of Science, Bengaluru, India
| |
Collapse
|
2
|
Ghanta PP, Dang CM, Nelson CM, Feaster DJ, Forrest DW, Tookes H, Pahwa RN, Pallikkuth S, Pahwa SG. Soluble Plasma Proteins of Tumor Necrosis Factor and Immunoglobulin Superfamilies Reveal New Insights into Immune Regulation in People with HIV and Opioid Use Disorder. Vaccines (Basel) 2024; 12:520. [PMID: 38793771 PMCID: PMC11125794 DOI: 10.3390/vaccines12050520] [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: 05/02/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024] Open
Abstract
People with HIV (PWH) frequently suffer from Opioid (OP) Use Disorder (OUD). In an investigation of the impact of OUD on underlying immune dysfunction in PWH, we previously reported that OP use exacerbates inflammation in virally controlled PWH followed in the Infectious Diseases Elimination Act (IDEA) Syringe Services Program (SSP). Unexpectedly, Flu vaccination-induced antibody responses in groups with OUD were superior to PWH without OUD. Here, we investigated the profile of 48 plasma biomarkers comprised of TNF and Ig superfamily (SF) molecules known to impact interactions between T and B cells in 209 participants divided into four groups: (1) HIV+OP+, (2) HIV-OP+, (3) HIV+OP-, and (4) HIV-OP-. The differential expression of the top eight molecules ranked by median values in individual Groups 1-3 in comparison to Group 4 was highly significant. Both OP+ groups 1 and 2 had higher co-stimulatory TNF SF molecules, including 4-1BB, OX-40, CD40, CD30, and 4-1BBL, which were found to positively correlate with Flu Ab titers. In contrast, HIV+OP- exhibited a profile dominant in Ig SF molecules, including PDL-2, CTLA-4, and Perforin, with PDL-2 showing a negative correlation with Flu vaccine titers. These findings are relevant to vaccine development in the fields of HIV and OUD.
Collapse
Affiliation(s)
- Priya P. Ghanta
- Department of Medicine, Division of Infectious Diseases, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (P.P.G.); (D.W.F.); (H.T.)
| | - Christine M. Dang
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (C.M.D.); (R.N.P.); (S.P.)
| | - C. Mindy Nelson
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (C.M.N.); (D.J.F.)
| | - Daniel J. Feaster
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (C.M.N.); (D.J.F.)
| | - David W. Forrest
- Department of Medicine, Division of Infectious Diseases, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (P.P.G.); (D.W.F.); (H.T.)
| | - Hansel Tookes
- Department of Medicine, Division of Infectious Diseases, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (P.P.G.); (D.W.F.); (H.T.)
| | - Rajendra N. Pahwa
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (C.M.D.); (R.N.P.); (S.P.)
| | - Suresh Pallikkuth
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (C.M.D.); (R.N.P.); (S.P.)
| | - Savita G. Pahwa
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (C.M.D.); (R.N.P.); (S.P.)
| |
Collapse
|
3
|
Sanchez S, Dangi T, Awakoaiye B, Irani N, Fourati S, Richner J, Penaloza-MacMaster P. Time-dependent enhancement of mRNA vaccines by 4-1BB costimulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.01.582992. [PMID: 38496467 PMCID: PMC10942304 DOI: 10.1101/2024.03.01.582992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
mRNA vaccines have demonstrated efficacy against COVID-19. However, concerns regarding waning immunity and breakthrough infections have motivated the development of next-generation vaccines with enhanced efficacy. In this study, we investigated the impact of 4-1BB costimulation on immune responses elicited by mRNA vaccines in mice. We first vaccinated mice with an mRNA vaccine encoding the SARS-CoV-2 spike antigen like the Moderna and Pfizer-BioNTech vaccines, followed by administration of 4-1BB costimulatory antibodies at various times post-vaccination. Administering 4-1BB costimulatory antibodies during the priming phase did not enhance immune responses. However, administering 4-1BB costimulatory antibodies after 96 hours elicited a significant improvement in CD8 T cell responses, leading to enhanced protection against breakthrough infections. A similar improvement in immune responses was observed with multiple mRNA vaccines, including vaccines against common cold coronavirus, human immunodeficiency virus (HIV), and arenavirus. These findings demonstrate a time-dependent effect by 4-1BB costimulation and provide insights for developing improved mRNA vaccines.
Collapse
Affiliation(s)
- Sarah Sanchez
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Tanushree Dangi
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Bakare Awakoaiye
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Nahid Irani
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Slim Fourati
- Department of Medicine, Division of Allergy and Immunology, Feinberg School of Medicine and Center for Human Immunobiology, Northwestern University, Chicago, IL 60611, USA
| | - Justin Richner
- Department of Microbiology & Immunology, University of Illinois Chicago College of Medicine, Chicago, IL 60612, USA
| | - Pablo Penaloza-MacMaster
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
4
|
Taleb E, Zucker R, Wagner-Kolasko G, Raviv Y. Atypical Presentation of Monkeypox Virus Infection in a Postvaccinated Altered Immunocompetent Patient-A Case-Report. Sex Transm Dis 2023; 50:59-61. [PMID: 36219768 DOI: 10.1097/olq.0000000000001723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Abstract
We herein report a case of an altered immunocompetent patient with atypical monkeypox presentations after being vaccinated with JYNNOES vaccine. The initial presentation was of acute bacterial paronychia. This incident suggests the need for individuals at high-risk for monkeypox infection with altered immunity to receive 2 vaccination doses.
Collapse
Affiliation(s)
| | | | | | - Yuval Raviv
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
5
|
Epstein-Barr virus-induced ectopic CD137 expression helps nasopharyngeal carcinoma to escape immune surveillance and enables targeting by chimeric antigen receptors. Cancer Immunol Immunother 2022; 71:2583-2596. [DOI: 10.1007/s00262-022-03183-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 02/21/2022] [Indexed: 12/24/2022]
|
6
|
Luu K, Patwardhan MV, Zeng Q, Wickström SL, Lundqvist A, Schwarz H. Regulatory T Cells Inhibit T Cell Activity by Downregulating CD137 Ligand via CD137 Trogocytosis. Cells 2021; 10:353. [PMID: 33572150 PMCID: PMC7914903 DOI: 10.3390/cells10020353] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/04/2021] [Accepted: 02/04/2021] [Indexed: 12/14/2022] Open
Abstract
CD137 is a costimulatory molecule expressed on activated T cells. CD137 ligand (CD137L) is expressed by antigen presenting cells (APC), which use the CD137-CD137L system to enhance immune responses. It was, therefore, surprising to discover CD137 expression on regulatory T cells (Treg). The function of CD137 in Treg are controversial. While some studies report that CD137 signalling converts Treg to effector T cells (Teff), other studies find that CD137-expressing Treg display a stronger inhibitory activity than CD137- Treg. Here, we describe that CD137 on Treg binds to CD137L on APC, upon which one of the two molecules is transferred via trogocytosis to the other cell, where CD137-CD137L forms a complex that is internalized and deprives APC of the immune-stimulatory CD137L. Truncated forms of CD137 that lack the cytoplasmic domain of CD137 are also able to downregulate CD137L, demonstrating that CD137 signalling is not required. Comparable data have been obtained with human and murine cells, indicating that this mechanism is evolutionarily conserved. These data describe trogocytosis of CD137 and CD137L as a new mechanism employed by Treg to control immune responses by downregulating the immunostimulatory CD137L on APC.
Collapse
Affiliation(s)
- Khang Luu
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (K.L.); (M.V.P.); (Q.Z.)
- NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore
- NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore 117456, Singapore
- Integrative Sciences and Engineering Programme, National University of Singapore, Singapore 117456, Singapore
- Department of Oncology-Pathology, Karolinska Institutet, 171 64 Stockholm, Sweden; (S.L.W.); (A.L.)
| | - Mugdha Vijay Patwardhan
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (K.L.); (M.V.P.); (Q.Z.)
- NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore
- NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore 117456, Singapore
| | - Qun Zeng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (K.L.); (M.V.P.); (Q.Z.)
- NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore
- NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore 117456, Singapore
| | - Stina L. Wickström
- Department of Oncology-Pathology, Karolinska Institutet, 171 64 Stockholm, Sweden; (S.L.W.); (A.L.)
| | - Andreas Lundqvist
- Department of Oncology-Pathology, Karolinska Institutet, 171 64 Stockholm, Sweden; (S.L.W.); (A.L.)
| | - Herbert Schwarz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (K.L.); (M.V.P.); (Q.Z.)
- NUS Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore 117456, Singapore
- NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore 117456, Singapore
- Integrative Sciences and Engineering Programme, National University of Singapore, Singapore 117456, Singapore
| |
Collapse
|
7
|
Lisiero DN, Cheng Z, Tejera MM, Neldner BT, Warrick JW, Wuerzberger-Davis SM, Hoffmann A, Suresh M, Miyamoto S. IκBα Nuclear Export Enables 4-1BB-Induced cRel Activation and IL-2 Production to Promote CD8 T Cell Immunity. THE JOURNAL OF IMMUNOLOGY 2020; 205:1540-1553. [PMID: 32817348 DOI: 10.4049/jimmunol.2000039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 07/20/2020] [Indexed: 12/19/2022]
Abstract
Optimal CD8 T cell immunity is orchestrated by signaling events initiated by TCR recognition of peptide Ag in concert with signals from molecules such as CD28 and 4-1BB. The molecular mechanisms underlying the temporal and spatial signaling dynamics in CD8 T cells remain incompletely understood. In this study, we show that stimulation of naive CD8 T cells with agonistic CD3 and CD28 Abs, mimicking TCR and costimulatory signals, coordinately induces 4-1BB and cRel to enable elevated cytosolic cRel:IκBα complex formation and subsequent 4-1BB-induced IκBα degradation, sustained cRel activation, heightened IL-2 production and T cell expansion. NfkbiaNES/NES CD8 T cells harboring a mutated IκBα nuclear export sequence abnormally accumulate inactive cRel:IκBα complexes in the nucleus following stimulation with agonistic anti-CD3 and anti-CD28 Abs, rendering them resistant to 4-1BB induced signaling and a disrupted chain of events necessary for efficient T cell expansion. Consequently, CD8 T cells in NfkbiaNES/NES mice poorly expand during viral infection, and this can be overcome by exogenous IL-2 administration. Consistent with cell-based data, adoptive transfer experiments demonstrated that the antiviral CD8 T cell defect in NfkbiaNES/NES mice was cell intrinsic. Thus, these results reveal that IκBα, via its unique nuclear export function, enables, rather than inhibits 4-1BB-induced cRel activation and IL-2 production to facilitate optimal CD8 T cell immunity.
Collapse
Affiliation(s)
- Dominique N Lisiero
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, WI 53705
| | - Zhang Cheng
- Department of Microbiology, Immunology, and Molecular Genetics, Institute for Quantitative and Computational Biosciences and Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90025
| | - Melba M Tejera
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706
| | - Brandon T Neldner
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706
| | - Jay W Warrick
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, WI 53705.,Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705; and
| | - Shelly M Wuerzberger-Davis
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, WI 53705
| | - Alexander Hoffmann
- Department of Microbiology, Immunology, and Molecular Genetics, Institute for Quantitative and Computational Biosciences and Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90025
| | - M Suresh
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706;
| | - Shigeki Miyamoto
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin-Madison, Madison, WI 53705; .,University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Wisconsin Institute for Medical Research, Madison, WI 53705
| |
Collapse
|
8
|
Luu K, Shao Z, Schwarz H. The relevance of soluble CD137 in the regulation of immune responses and for immunotherapeutic intervention. J Leukoc Biol 2020; 107:731-738. [PMID: 32052477 DOI: 10.1002/jlb.2mr1119-224r] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 11/10/2019] [Accepted: 12/05/2019] [Indexed: 12/13/2022] Open
Abstract
CD137 is a potent costimulatory receptor. Several agonistic anti-CD137 antibodies are currently in clinical trials for tumor immunotherapy. Soluble forms of CD137 (sCD137) are generated by differential splicing and antagonize the activities of membrane-bound CD137 (mCD137) and of therapeutic CD137 agonists. sCD137 is found in sera of patients suffering from autoimmune diseases where it is a natural regulator of immune responses, and which has therapeutic potential for immune-mediated diseases. This review summarizes the current knowledge on sCD137, highlights its potential role in immunotherapy against cancer and in autoimmune diseases, and presents important issues to be addressed by future research.
Collapse
Affiliation(s)
- Khang Luu
- Department of Physiology, and Immunology Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Graduate School for Integrative Sciences & Engineering, National University of Singapore, Singapore, Singapore
| | - Zhe Shao
- Department of Physiology, and Immunology Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Herbert Schwarz
- Department of Physiology, and Immunology Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Graduate School for Integrative Sciences & Engineering, National University of Singapore, Singapore, Singapore
| |
Collapse
|
9
|
CD137 ligand interacts with CD32a to trigger reverse CD137 ligand signaling. Cell Mol Immunol 2020; 17:1188-1189. [PMID: 32076121 DOI: 10.1038/s41423-020-0370-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 01/17/2020] [Indexed: 12/22/2022] Open
|
10
|
Wu M, Wong HY, Lin JL, Moliner A, Schwarz H. Induction of CD137 expression by viral genes reduces T cell costimulation. J Cell Physiol 2019; 234:21076-21088. [DOI: 10.1002/jcp.28710] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/26/2019] [Accepted: 04/10/2019] [Indexed: 12/28/2022]
Affiliation(s)
- Meihui Wu
- Department of Physiology Yong Loo Lin School of Medicine, National University of Singapore Singapore
- Immunology Programme Life Sciences Institute, National University of Singapore Singapore
| | - Hiu Yi Wong
- Department of Physiology Yong Loo Lin School of Medicine, National University of Singapore Singapore
- Immunology Programme Life Sciences Institute, National University of Singapore Singapore
| | - Jia Le Lin
- Department of Physiology Yong Loo Lin School of Medicine, National University of Singapore Singapore
- Immunology Programme Life Sciences Institute, National University of Singapore Singapore
| | - Annalena Moliner
- Immunology Programme Life Sciences Institute, National University of Singapore Singapore
| | - Herbert Schwarz
- Department of Physiology Yong Loo Lin School of Medicine, National University of Singapore Singapore
- Immunology Programme Life Sciences Institute, National University of Singapore Singapore
| |
Collapse
|
11
|
Abstract
Costimulatory signals initiated by the interaction between the tumor necrosis factor (TNF) ligand and cognate TNF receptor (TNFR) superfamilies promote clonal expansion, differentiation, and survival of antigen-primed CD4+ and CD8+ T cells and have a pivotal role in T-cell-mediated adaptive immunity and diseases. Accumulating evidence in recent years indicates that costimulatory signals via the subset of the TNFR superfamily molecules, OX40 (TNFRSF4), 4-1BB (TNFRSF9), CD27, DR3 (TNFRSF25), CD30 (TNFRSF8), GITR (TNFRSF18), TNFR2 (TNFRSF1B), and HVEM (TNFRSF14), which are constitutive or inducible on T cells, play important roles in protective immunity, inflammatory and autoimmune diseases, and tumor immunotherapy. In this chapter, we will summarize the findings of recent studies on these TNFR family of co-signaling molecules regarding their function at various stages of the T-cell response in the context of infection, inflammation, and cancer. We will also discuss how these TNFR co-signals are critical for immune regulation and have therapeutic potential for the treatment of T-cell-mediated diseases.
Collapse
|
12
|
Wallach D. The Tumor Necrosis Factor Family: Family Conventions and Private Idiosyncrasies. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a028431. [PMID: 28847899 DOI: 10.1101/cshperspect.a028431] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The tumor necrosis factor (TNF) cytokine family and the TNF/nerve growth factor (NGF) family of their cognate receptors together control numerous immune functions, as well as tissue-homeostatic and embryonic-development processes. These diverse functions are dictated by both shared and distinct features of family members, and by interactions of some members with nonfamily ligands and coreceptors. The spectra of their activities are further expanded by the occurrence of the ligands and receptors in both membrane-anchored and soluble forms, by "re-anchoring" of soluble forms to extracellular matrix components, and by signaling initiation via intracellular domains (IDs) of both receptors and ligands. Much has been learned about shared features of the receptors as well as of the ligands; however, we still have only limited knowledge of the mechanistic basis for their functional heterogeneity and for the differences between their functions and those of similarly acting cytokines of other families.
Collapse
Affiliation(s)
- David Wallach
- Department of Biomolecular Sciences, The Weizmann Institute of Science, 76100 Rehovot, Israel
| |
Collapse
|
13
|
Vashist N, Trittel S, Ebensen T, Chambers BJ, Guzmán CA, Riese P. Influenza-Activated ILC1s Contribute to Antiviral Immunity Partially Influenced by Differential GITR Expression. Front Immunol 2018; 9:505. [PMID: 29623077 PMCID: PMC5874297 DOI: 10.3389/fimmu.2018.00505] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 02/26/2018] [Indexed: 01/07/2023] Open
Abstract
Innate lymphoid cells (ILCs) represent diversified subsets of effector cells as well as immune regulators of mucosal immunity and are classified into group 1 ILCs, group 2 ILCs, and group 3 ILCs. Group 1 ILCs encompass natural killer (NK) cells and non-NK ILCs (ILC1s) and mediate their functionality via the rapid production of IFN-γ and TNF-α. The current knowledge of ILC1s mainly associates them to inflammatory processes. Much less is known about their regulation during infection and their capacity to interact with cells of the adaptive immune system. The present study dissected the role of ILC1s during early influenza A virus infection, thereby revealing their impact on the antiviral response. Exploiting in vitro and in vivo H1N1 infection systems, a cross-talk of ILC1s with cells of the innate and the adaptive immunity was demonstrated, which contributes to anti-influenza immunity. A novel association of ILC1 functionality and the expression of the glucocorticoid-induced TNFR-related protein (GITR) was observed, which hints toward a so far undescribed role of GITR in regulating ILC1 responsiveness. Overexpression of GITR inhibits IFN-γ production by ILC1s, whereas partial reduction of GITR expression can reverse this effect, thereby regulating ILC1 functionality. These new insights into ILC1 biology define potential intervention targets to modulate the functional properties of ILC1s, thus contributing toward the development of new immune interventions against influenza.
Collapse
Affiliation(s)
- Neha Vashist
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Department of Medicine, Center for Infectious Medicine, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Stephanie Trittel
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Thomas Ebensen
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Benedict J Chambers
- Department of Medicine, Center for Infectious Medicine, Karolinska Institute, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Carlos A Guzmán
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Peggy Riese
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
14
|
Bathke B, Pätzold J, Kassub R, Giessel R, Lämmermann K, Hinterberger M, Brinkmann K, Chaplin P, Suter M, Hochrein H, Lauterbach H. CD70 encoded by modified vaccinia virus Ankara enhances CD8 T-cell-dependent protective immunity in MHC class II-deficient mice. Immunology 2018; 154:285-297. [PMID: 29281850 PMCID: PMC5980220 DOI: 10.1111/imm.12884] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 11/28/2017] [Accepted: 12/03/2017] [Indexed: 12/13/2022] Open
Abstract
The immunological outcome of infections and vaccinations is largely determined during the initial first days in which antigen-presenting cells instruct T cells to expand and differentiate into effector and memory cells. Besides the essential stimulation of the T-cell receptor complex a plethora of co-stimulatory signals not only ensures a proper T-cell activation but also instils phenotypic and functional characteristics in the T cells appropriate to fight off the invading pathogen. The tumour necrosis factor receptor/ligand pair CD27/CD70 gained a lot of attention because of its key role in regulating T-cell activation, survival, differentiation and maintenance, especially in the course of viral infections and cancer. We sought to investigate the role of CD70 co-stimulation for immune responses induced by the vaccine vector modified vaccinia virus Ankara-Bavarian Nordic® (MVA-BN® ). Short-term blockade of CD70 diminished systemic CD8 T-cell effector and memory responses in mice. The dependence on CD70 became even more apparent in the lungs of MHC class II-deficient mice. Importantly, genetically encoded CD70 in MVA-BN® not only increased CD8 T-cell responses in wild-type mice but also substituted for CD4 T-cell help. MHC class II-deficient mice that were immunized with recombinant MVA-CD70 were fully protected against a lethal virus infection, whereas MVA-BN® -immunized mice failed to control the virus. These data are in line with CD70 playing an important role for vaccine-induced CD8 T-cell responses and prove the potency of integrating co-stimulatory molecules into the MVA-BN® backbone.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Mark Suter
- Vetsuisse Fakultät, Dekanat, Bereich Immunologie, Universität Zürich, Zurich, Switzerland
| | | | | |
Collapse
|
15
|
Zhu HQ, Gao FH. The Molecular Mechanisms of Regulation on USP2's Alternative Splicing and the Significance of Its Products. Int J Biol Sci 2017; 13:1489-1496. [PMID: 29230097 PMCID: PMC5723915 DOI: 10.7150/ijbs.21637] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 09/10/2017] [Indexed: 01/06/2023] Open
Abstract
Ubiquitin-specific protease 2 (USP2) has a regulatory function in cell growth or death and is involved in the pathogenesis of various diseases. USP2 gene can generate 7 splicing variants through alternative splicing, and 5 variants respectively as USP2-201, USP2-202, USP2-204, USP2-205, USP2-206 can encode proteins. The influence of circadian rhythm, nutrition and androgen on specific signaling molecules or cytokines can regulate the alternative splicing of USP2. Specifically, PKC activator, IL-1β, TNF-α, PDGF-BB, TGF-β1 are all regulatory factors for USP2's alternative splicing. USP2-201 plays a crucial role in cell cycle progression, and is also of great significance in EGFR recycling. USP2-202 can activate apoptosis signaling pathway to participate in cell apoptosis, and USP2-204 can induce cell anti-virus reaction to decrease. In general, we collect and summarize the factors involved in the alternative splicing of USP2 in this review to further understand the mechanism behind the USP2's alternative splicing.
Collapse
Affiliation(s)
| | - Feng-Hou Gao
- Department of Oncology, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhi Zao Ju Rd, Shanghai, China
| |
Collapse
|
16
|
Ward-Kavanagh LK, Lin WW, Šedý JR, Ware CF. The TNF Receptor Superfamily in Co-stimulating and Co-inhibitory Responses. Immunity 2017; 44:1005-19. [PMID: 27192566 DOI: 10.1016/j.immuni.2016.04.019] [Citation(s) in RCA: 314] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Indexed: 02/08/2023]
Abstract
Cytokines related to tumor necrosis factor (TNF) provide a communication network essential for coordinating multiple cell types into an effective host defense system against pathogens and malignant cells. The pathways controlled by the TNF superfamily differentiate both innate and adaptive immune cells and modulate stromal cells into microenvironments conducive to host defenses. Members of the TNF receptor superfamily activate diverse cellular functions from the production of type 1 interferons to the modulation of survival of antigen-activated T cells. Here, we focus attention on the subset of TNF superfamily receptors encoded in the immune response locus in chromosomal region 1p36. Recent studies have revealed that these receptors use diverse mechanisms to either co-stimulate or restrict immune responses. Translation of the fundamental mechanisms of TNF superfamily is leading to the design of therapeutics that can alter pathogenic processes in several autoimmune diseases or promote immunity to tumors.
Collapse
Affiliation(s)
- Lindsay K Ward-Kavanagh
- Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Wai Wai Lin
- Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - John R Šedý
- Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Carl F Ware
- Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
17
|
Shen YL, Gan Y, Gao HF, Fan YC, Wang Q, Yuan H, Song YF, Wang JD, Tu H. TNFSF9 exerts an inhibitory effect on hepatocellular carcinoma. J Dig Dis 2017; 18:395-403. [PMID: 28547807 DOI: 10.1111/1751-2980.12489] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 05/07/2017] [Accepted: 05/23/2017] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Tumor necrosis factor superfamily member 9 (TNFSF9), also known as 4-1BBL and CD137L, has been implicated in cancer immunotherapy due to its function as a T-cell co-stimulator. We aimed to investigate the role of TNFSF9 in the cancer pathogenesis in hepatocellular carcinoma (HCC). METHODS TNFSF9 expression was examined by immunohistochemistry in 106 pairs of HCC and adjacent non-tumorous tissues, and by quantitative polymerase chain reaction and Western blot in HCC cell lines. The impact of TNFSF9 on the proliferation, migration and invasion of HCC cells was determined using the 3-(4,5-diethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt (MTS) and transwell assays in vitro. We also assessed the influence of TNFSF9 on the growth and metastasis of HCC tumors in an orthotopic mouse model of human HCC. RESULTS TNFSF9 expression was downregulated in approximately 70% of HCC tissues. A decreased expression of TNFSF9 was also consistently observed in all the four HCC cell lines. Either the overexpression of TNFSF9 or treatment with recombinant TNFSF9 protein could significantly inhibit the proliferation, migration and invasion of Huh7 and SMMC-7721 HCC cells in vitro. The inhibitory effect of TNFSF9 on HCC was further confirmed in vivo. Mice orthotopically transplanted with TNFSF9-overexpressing Huh7 cells developed significantly smaller tumors with less intrahepatic metastasis and distant metastasis compared with the control group. CONCLUSIONS TNFSF9 may be a tumor suppressor in HCC. Based on its immune stimulatory aspect and the tumor inhibition property, TNFSF9 may be a promising therapeutic target for HCC.
Collapse
Affiliation(s)
- Yu Ling Shen
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Head and Neck Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yu Gan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hai Feng Gao
- Department of Immunology, Key Laboratory of Medical Molecular Virology of MOE/MOH, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ying Chao Fan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qing Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hui Yuan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Fang Song
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jia Dong Wang
- Department of Head and Neck Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hong Tu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
18
|
Flies AS, Blackburn NB, Lyons AB, Hayball JD, Woods GM. Comparative Analysis of Immune Checkpoint Molecules and Their Potential Role in the Transmissible Tasmanian Devil Facial Tumor Disease. Front Immunol 2017; 8:513. [PMID: 28515726 PMCID: PMC5413580 DOI: 10.3389/fimmu.2017.00513] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Accepted: 04/18/2017] [Indexed: 12/13/2022] Open
Abstract
Immune checkpoint molecules function as a system of checks and balances that enhance or inhibit immune responses to infectious agents, foreign tissues, and cancerous cells. Immunotherapies that target immune checkpoint molecules, particularly the inhibitory molecules programmed cell death 1 and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), have revolutionized human oncology in recent years, yet little is known about these key immune signaling molecules in species other than primates and rodents. The Tasmanian devil facial tumor disease is caused by transmissible cancers that have resulted in a massive decline in the wild Tasmanian devil population. We have recently demonstrated that the inhibitory checkpoint molecule PD-L1 is upregulated on Tasmanian devil (Sarcophilus harrisii) facial tumor cells in response to the interferon-gamma cytokine. As this could play a role in immune evasion by tumor cells, we performed a thorough comparative analysis of checkpoint molecule protein sequences among Tasmanian devils and eight other species. We report that many of the key signaling motifs and ligand-binding sites in the checkpoint molecules are highly conserved across the estimated 162 million years of evolution since the last common ancestor of placental and non-placental mammals. Specifically, we discovered that the CTLA-4 (MYPPPY) ligand-binding motif and the CTLA-4 (GVYVKM) inhibitory domain are completely conserved across all nine species used in our comparative analysis, suggesting that the function of CTLA-4 is likely conserved in these species. We also found that cysteine residues for intra- and intermolecular disulfide bonds were also highly conserved. For instance, all 20 cysteine residues involved in disulfide bonds in the human 4-1BB molecule were also present in devil 4-1BB. Although many key sequences were conserved, we have also identified immunoreceptor tyrosine-based inhibitory motifs (ITIMs) and immunoreceptor tyrosine-based switch motifs (ITSMs) in genes and protein domains that have not been previously reported in any species. This checkpoint molecule analysis and review of salient features for each of the molecules presented here can serve as road map for the development of a Tasmanian devil facial tumor disease immunotherapy. Finally, the strategies can be used as a guide for veterinarians, ecologists, and other researchers willing to venture into the nascent field of wild immunology.
Collapse
Affiliation(s)
- Andrew S. Flies
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
- Department of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Nicholas B. Blackburn
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
- School of Medicine, South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Alan Bruce Lyons
- School of Medicine, University of Tasmania, Hobart, TAS, Australia
| | - John D. Hayball
- Department of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
- Discipline of Obstetrics and Gynaecology, School of Medicine, Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
| | - Gregory M. Woods
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| |
Collapse
|
19
|
Forsythoside A Inhibits BVDV Replication via TRAF2-Dependent CD28-4-1BB Signaling in Bovine PBMCs. PLoS One 2016; 11:e0162791. [PMID: 27617959 PMCID: PMC5019491 DOI: 10.1371/journal.pone.0162791] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 08/28/2016] [Indexed: 01/02/2023] Open
Abstract
Bovine viral diarrhea virus (BVDV), the causative agent of bovine viral diarrhea/mucosal disease (BVD/MD), is an important pathogen of cattle and other wild animals throughout the world. BVDV infection typically leads to an impaired immune response in cattle. In the present study, we investigated the effect of Forsythoside A (FTA) on BVDV infection of bovine peripheral blood mononuclear cells (PBMCs). We found that Forsythoside A could not only promote proliferation of PBMCs and T cells activation but also inhibit the replication of BVDV as well as apoptosis induced by BVDV. FTA treatment could counteract the BVDV-induced overproduction of IFN-γ to maintain the immune homeostasis in bovine PBMCs. At same time, FTA can enhance the secretion of IL-2. What's more, BVDV promotes the expression of CD28, 4-1BB and TRAF-2, which can be modulated by FTA. Our data suggest that FTA protects PBMCs from BVDV infection possibly via TRAF2-dependent CD28-4-1BB signaling, which may activate PBMCs in response to BVDV infection. Therefore, this aids in the development of an effective adjuvant for vaccines against BVDV and other specific FTA-based therapies for preventing BVDV infection.
Collapse
|
20
|
Wang W, Xu L, Brandsma JH, Wang Y, Hakim MS, Zhou X, Yin Y, Fuhler GM, van der Laan LJW, van der Woude CJ, Sprengers D, Metselaar HJ, Smits R, Poot RA, Peppelenbosch MP, Pan Q. Convergent Transcription of Interferon-stimulated Genes by TNF-α and IFN-α Augments Antiviral Activity against HCV and HEV. Sci Rep 2016; 6:25482. [PMID: 27150018 PMCID: PMC4858707 DOI: 10.1038/srep25482] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 04/19/2016] [Indexed: 01/05/2023] Open
Abstract
IFN-α has been used for decades to treat chronic hepatitis B and C, and as an off-label treatment for some cases of hepatitis E virus (HEV) infection. TNF-α is another important cytokine involved in inflammatory disease, which can interact with interferon signaling. Because interferon-stimulated genes (ISGs) are the ultimate antiviral effectors of the interferon signaling, this study aimed to understand the regulation of ISG transcription and the antiviral activity by IFN-α and TNF-α. In this study, treatment of TNF-α inhibited replication of HCV by 71 ± 2.4% and HEV by 41 ± 4.9%. Interestingly, TNF-α induced the expression of a panel of antiviral ISGs (2-11 fold). Blocking the TNF-α signaling by Humira abrogated ISG induction and its antiviral activity. Chip-seq data analysis and mutagenesis assay further revealed that the NF-κB protein complex, a key downstream element of TNF-α signaling, directly binds to the ISRE motif in the ISG promoters and thereby drives their transcription. This process is independent of interferons and JAK-STAT cascade. Importantly, when combined with IFN-α, TNF-α works cooperatively on ISG induction, explaining their additive antiviral effects. Thus, our study reveals a novel mechanism of convergent transcription of ISGs by TNF-α and IFN-α, which augments their antiviral activity against HCV and HEV.
Collapse
Affiliation(s)
- Wenshi Wang
- Department of Gastroenterology and Hepatology, Postgraduate School Molecular Medicine, Erasmus MC-University Medical Center, Rotterdam, 3015 CE, The Netherlands
| | - Lei Xu
- Department of Gastroenterology and Hepatology, Postgraduate School Molecular Medicine, Erasmus MC-University Medical Center, Rotterdam, 3015 CE, The Netherlands
| | - Johannes H Brandsma
- Department of Cell Biology, Medical Genetics Cluster, Erasmus MC-University Medical Center, Rotterdam, 3015 CE, The Netherlands
| | - Yijin Wang
- Department of Gastroenterology and Hepatology, Postgraduate School Molecular Medicine, Erasmus MC-University Medical Center, Rotterdam, 3015 CE, The Netherlands
| | - Mohamad S Hakim
- Department of Gastroenterology and Hepatology, Postgraduate School Molecular Medicine, Erasmus MC-University Medical Center, Rotterdam, 3015 CE, The Netherlands.,Department of Microbiology, Faculty of Medicine, Gadjah Mada University, Yogyakarta, Indonesia
| | - Xinying Zhou
- Department of Gastroenterology and Hepatology, Postgraduate School Molecular Medicine, Erasmus MC-University Medical Center, Rotterdam, 3015 CE, The Netherlands
| | - Yuebang Yin
- Department of Gastroenterology and Hepatology, Postgraduate School Molecular Medicine, Erasmus MC-University Medical Center, Rotterdam, 3015 CE, The Netherlands
| | - Gwenny M Fuhler
- Department of Gastroenterology and Hepatology, Postgraduate School Molecular Medicine, Erasmus MC-University Medical Center, Rotterdam, 3015 CE, The Netherlands
| | - Luc J W van der Laan
- Department of Surgery, Postgraduate School Molecular Medicine, Erasmus MC-University Medical Center, Rotterdam, 3015 CE, The Netherlands
| | - C Janneke van der Woude
- Department of Gastroenterology and Hepatology, Postgraduate School Molecular Medicine, Erasmus MC-University Medical Center, Rotterdam, 3015 CE, The Netherlands
| | - Dave Sprengers
- Department of Gastroenterology and Hepatology, Postgraduate School Molecular Medicine, Erasmus MC-University Medical Center, Rotterdam, 3015 CE, The Netherlands
| | - Herold J Metselaar
- Department of Gastroenterology and Hepatology, Postgraduate School Molecular Medicine, Erasmus MC-University Medical Center, Rotterdam, 3015 CE, The Netherlands
| | - Ron Smits
- Department of Gastroenterology and Hepatology, Postgraduate School Molecular Medicine, Erasmus MC-University Medical Center, Rotterdam, 3015 CE, The Netherlands
| | - Raymond A Poot
- Department of Cell Biology, Medical Genetics Cluster, Erasmus MC-University Medical Center, Rotterdam, 3015 CE, The Netherlands
| | - Maikel P Peppelenbosch
- Department of Gastroenterology and Hepatology, Postgraduate School Molecular Medicine, Erasmus MC-University Medical Center, Rotterdam, 3015 CE, The Netherlands
| | - Qiuwei Pan
- Department of Gastroenterology and Hepatology, Postgraduate School Molecular Medicine, Erasmus MC-University Medical Center, Rotterdam, 3015 CE, The Netherlands
| |
Collapse
|
21
|
Dharmadhikari B, Wu M, Abdullah NS, Rajendran S, Ishak ND, Nickles E, Harfuddin Z, Schwarz H. CD137 and CD137L signals are main drivers of type 1, cell-mediated immune responses. Oncoimmunology 2016; 5:e1113367. [PMID: 27141396 PMCID: PMC4839363 DOI: 10.1080/2162402x.2015.1113367] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 10/22/2015] [Accepted: 10/22/2015] [Indexed: 10/22/2022] Open
Abstract
CD137 is expressed on activated T cells and NK cells, among others, and is a potent co-stimulator of antitumor immune responses. CD137 ligand (CD137L) is expressed by antigen presenting cells (APC), and CD137L reverse signaling into APC enhances their activity. CD137-CD137L interactions as main driver of type 1, cell-mediated immune responses explains the puzzling observation that CD137 agonists which enhance antitumor immune responses also ameliorate autoimmune diseases. Upon co-stimulation by CD137, Th1 CD4+ T cells together with Tc1 CD8+ T cells and NK cells inhibit other T cell subsets, thereby promoting antitumor responses and mitigating non-type 1 auto-immune diseases.
Collapse
Affiliation(s)
- Bhushan Dharmadhikari
- Department of Physiology, and Immunology Programme, National University of Singapore, Singapore
| | - Meihui Wu
- Department of Physiology, and Immunology Programme, National University of Singapore, Singapore
| | - Nur Sharalyn Abdullah
- Department of Physiology, and Immunology Programme, National University of Singapore, Singapore
| | - Sakthi Rajendran
- Department of Physiology, and Immunology Programme, National University of Singapore, Singapore
| | - Nur Diana Ishak
- Department of Physiology, and Immunology Programme, National University of Singapore, Singapore
| | - Emily Nickles
- Department of Physiology, and Immunology Programme, National University of Singapore, Singapore
| | - Zulkarnain Harfuddin
- Department of Physiology, and Immunology Programme, National University of Singapore, Singapore
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore
| | - Herbert Schwarz
- Department of Physiology, and Immunology Programme, National University of Singapore, Singapore
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore
| |
Collapse
|
22
|
Sriram U, Haldar B, Cenna JM, Gofman L, Potula R. Methamphetamine mediates immune dysregulation in a murine model of chronic viral infection. Front Microbiol 2015; 6:793. [PMID: 26322025 PMCID: PMC4531300 DOI: 10.3389/fmicb.2015.00793] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 07/21/2015] [Indexed: 02/03/2023] Open
Abstract
Methamphetamine (METH) is a highly addictive psychostimulant that not only affects the brain and cognitive functions but also greatly impacts the host immune system, rendering the body susceptible to infections and exacerbating the severity of disease. Although there is gathering evidence about METH abuse and increased incidence of HIV and other viral infections, not much is known about the effects on the immune system in a chronic viral infection setting. We have used the lymphocytic choriomeningitis virus (LCMV) chronic mouse model of viral infection in a chronic METH environment and demonstrate that METH significantly increases CD3 marker on splenocytes and programmed death-1 (PD-1) expression on T cells, a cell surface signaling molecule known to inhibit T cell function and cause exhaustion in a lymphoid organ. Many of these METH effects were more pronounced during early stage of infection, which are gradually attenuated during later stages of infection. An essential cytokine for T-lymphocyte homeostasis, Interleukin-2 (IL-2) in serum was prominently reduced in METH-exposed infected mice. In addition, the serum pro-inflammatory (TNF, IL12 p70, IL1β, IL-6, and KC-GRO) and Th2 (IL-2, IL-10, and IL-4) cytokine profiles were also altered in the presence of METH. Interestingly CXCR3, an inflammatory chemokine receptor, showed significant increase in the METH treated LCMV infected mice. Similarly, compared to only infected mice, epidermal growth factor receptor (EGFR) in METH exposed LCMV infected mice were up regulated. Collectively, our data suggest that METH alters systemic, peripheral immune responses and modulates key markers on T cells involved in pathogenesis of chronic viral infection.
Collapse
Affiliation(s)
- Uma Sriram
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine Philadelphia, PA, USA
| | - Bijayesh Haldar
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine Philadelphia, PA, USA
| | - Jonathan M Cenna
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine Philadelphia, PA, USA
| | - Larisa Gofman
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine Philadelphia, PA, USA
| | - Raghava Potula
- Department of Pathology and Laboratory Medicine, Temple University School of Medicine Philadelphia, PA, USA ; Center for Substance Abuse Research, Temple University School of Medicine Philadelphia, PA, USA
| |
Collapse
|
23
|
Linch SN, McNamara MJ, Redmond WL. OX40 Agonists and Combination Immunotherapy: Putting the Pedal to the Metal. Front Oncol 2015; 5:34. [PMID: 25763356 PMCID: PMC4329814 DOI: 10.3389/fonc.2015.00034] [Citation(s) in RCA: 183] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 01/30/2015] [Indexed: 12/24/2022] Open
Abstract
Recent studies have highlighted the therapeutic efficacy of immunotherapy, a class of cancer treatments that utilize the patient’s own immune system to destroy cancerous cells. Within a tumor the presence of a family of negative regulatory molecules, collectively known as “checkpoint inhibitors,” can inhibit T cell function to suppress anti-tumor immunity. Checkpoint inhibitors, such as CTLA-4 and PD-1, attenuate T cell proliferation and cytokine production. Targeted blockade of CTLA-4 or PD-1 with antagonist monoclonal antibodies (mAbs) releases the “brakes” on T cells to boost anti-tumor immunity. Generating optimal “killer” CD8 T cell responses also requires T cell receptor activation plus co-stimulation, which can be provided through ligation of tumor necrosis factor receptor family members, including OX40 (CD134) and 4-1BB (CD137). OX40 is of particular interest as treatment with an activating (agonist) anti-OX40 mAb augments T cell differentiation and cytolytic function leading to enhanced anti-tumor immunity against a variety of tumors. When used as single agents, these drugs can induce potent clinical and immunologic responses in patients with metastatic disease. However, each of these agents only benefits a subset of patients, highlighting the critical need for more effective combinatorial therapeutic strategies. In this review, we will discuss our current understanding of the cellular and molecular mechanisms by which OX40 agonists synergize with checkpoint inhibitor blockade to augment T cell-mediated anti-tumor immunity and the potential opportunities for clinical translation of combinatorial immunotherapeutic strategies.
Collapse
Affiliation(s)
- Stefanie N Linch
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Portland Medical Center , Portland, OR , USA
| | - Michael J McNamara
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Portland Medical Center , Portland, OR , USA
| | - William L Redmond
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Portland Medical Center , Portland, OR , USA
| |
Collapse
|