1
|
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
|
2
|
Stoler-Barak L, Harris E, Peres A, Hezroni H, Kuka M, Di Lucia P, Grenov A, Gurwicz N, Kupervaser M, Yip BH, Iannacone M, Yaari G, Crispino JD, Shulman Z. B cell class switch recombination is regulated by DYRK1A through MSH6 phosphorylation. Nat Commun 2023; 14:1462. [PMID: 36927854 PMCID: PMC10020581 DOI: 10.1038/s41467-023-37205-5] [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: 06/21/2022] [Accepted: 03/06/2023] [Indexed: 03/18/2023] Open
Abstract
Protection from viral infections depends on immunoglobulin isotype switching, which endows antibodies with effector functions. Here, we find that the protein kinase DYRK1A is essential for B cell-mediated protection from viral infection and effective vaccination through regulation of class switch recombination (CSR). Dyrk1a-deficient B cells are impaired in CSR activity in vivo and in vitro. Phosphoproteomic screens and kinase-activity assays identify MSH6, a DNA mismatch repair protein, as a direct substrate for DYRK1A, and deletion of a single phosphorylation site impaired CSR. After CSR and germinal center (GC) seeding, DYRK1A is required for attenuation of B cell proliferation. These findings demonstrate DYRK1A-mediated biological mechanisms of B cell immune responses that may be used for therapeutic manipulation in antibody-mediated autoimmunity.
Collapse
Affiliation(s)
- Liat Stoler-Barak
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Ethan Harris
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Ayelet Peres
- Faculty of Engineering, Bar Ilan University, Ramat Gan, 52900, Israel
| | - Hadas Hezroni
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Mirela Kuka
- Vita-Salute San Raffaele University and Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Pietro Di Lucia
- Vita-Salute San Raffaele University and Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Amalie Grenov
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Neta Gurwicz
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Meital Kupervaser
- De Botton Institute for Proteomics, Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Bon Ham Yip
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Matteo Iannacone
- Vita-Salute San Raffaele University and Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Gur Yaari
- Faculty of Engineering, Bar Ilan University, Ramat Gan, 52900, Israel
| | - John D Crispino
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Ziv Shulman
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, 7610001, Israel.
| |
Collapse
|
3
|
Limbic Encephalitis Brain Damage Induced by Cocal Virus in Adult Mice Is Reduced by Environmental Enrichment: Neuropathological and Behavioral Studies. Viruses 2020; 13:v13010048. [PMID: 33396704 PMCID: PMC7824630 DOI: 10.3390/v13010048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 12/14/2020] [Indexed: 12/26/2022] Open
Abstract
We previously demonstrated, using the Piry virus model, that environmental enrichment promotes higher T-cell infiltration, fewer microglial changes, and faster central nervous system (CNS) virus clearance in adult mice. However, little is known about disease progression, behavioral changes, CNS cytokine concentration, and neuropathology in limbic encephalitis in experimental models. Using Cocal virus, we infected C57Bl6 adult mice and studied the neuroanatomical distribution of viral antigens in correlation with the microglial morphological response, measured the CNS cytokine concentration, and assessed behavioral changes. C57Bl6 adult mice were maintained in an impoverished environment (IE) or enriched environment (EE) for four months and then subjected to the open field test. Afterwards, an equal volume of normal or virus-infected brain homogenate was nasally instilled. The brains were processed to detect viral antigens and microglial morphological changes using selective immunolabeling. We demonstrated earlier significant weight loss and higher mortality in IE mice. Additionally, behavioral analysis revealed a significant influence of the environment on locomotor and exploratory activity that was associated with less neuroinvasion and a reduced microglial response. Thus, environmental enrichment was associated with a more effective immune response in a mouse model of limbic encephalitis, allowing faster viral clearance/decreased viral dissemination, reduced disease progression, and less CNS damage.
Collapse
|
4
|
Goncalves-Alves E, Saferding V, Schliehe C, Benson R, Kurowska-Stolarska M, Brunner JS, Puchner A, Podesser BK, Smolen JS, Redlich K, Bonelli M, Brewer J, Bergthaler A, Steiner G, Blüml S. MicroRNA-155 Controls T Helper Cell Activation During Viral Infection. Front Immunol 2019; 10:1367. [PMID: 31275315 PMCID: PMC6593301 DOI: 10.3389/fimmu.2019.01367] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/29/2019] [Indexed: 11/27/2022] Open
Abstract
MicroRNA (miR) 155 has been implicated in the regulation of innate and adaptive immunity as well as autoimmune processes. Importantly, it has been shown to regulate several antiviral responses, but its contribution to the immune response against cytopathic viruses such as vesicular stomatitis virus (VSV) infections is not known. Using transgenic/recombinant VSV expressing ovalbumin, we show that miR-155 is crucially involved in regulating the T helper cell response against this virus. Our experiments indicate that miR-155 in CD4+ T cells controls their activation, proliferation, and cytokine production in vitro and in vivo upon immunization with OVA as well as during VSV viral infection. Using intravital multiphoton microscopy we analyzed the interaction of antigen presenting cells (APCs) and T cells after OVA immunization and found impaired complex formation when using miR-155 deficient CD4+ T cells compared to wildtype CD4+ T cells ex vivo. In contrast, miR-155 was dispensable for the maturation of myeloid APCs and for their T cell stimulatory capacity. Our data provide the first evidence that miR-155 is required for efficient CD4+ T cell activation during anti-viral defense by allowing robust APC-T cell interaction required for activation and cytokine production of virus specific T cells.
Collapse
Affiliation(s)
| | - Victoria Saferding
- Department of Rheumatology, Medical University Vienna, Vienna, Austria.,Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Vienna, Austria
| | - Christopher Schliehe
- CeMM - Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Robert Benson
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | | | - Julia Stefanie Brunner
- Institute for Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria.,Christian Doppler Laboratory for Arginine Metabolism in Rheumatoid Arthritis and Multiple Sclerosis, Vienna, Austria
| | - Antonia Puchner
- Department of Rheumatology, Medical University Vienna, Vienna, Austria
| | - Bruno K Podesser
- Department of Biomedical Research, Medical University of Vienna, Vienna, Austria
| | - Josef S Smolen
- Department of Rheumatology, Medical University Vienna, Vienna, Austria
| | - Kurt Redlich
- Department of Rheumatology, Medical University Vienna, Vienna, Austria
| | - Michael Bonelli
- Department of Rheumatology, Medical University Vienna, Vienna, Austria
| | - James Brewer
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Andreas Bergthaler
- CeMM - Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Günter Steiner
- Department of Rheumatology, Medical University Vienna, Vienna, Austria
| | - Stephan Blüml
- Department of Rheumatology, Medical University Vienna, Vienna, Austria.,Christian Doppler Laboratory for Arginine Metabolism in Rheumatoid Arthritis and Multiple Sclerosis, Vienna, Austria
| |
Collapse
|
5
|
Huang Q, Hu J, Tang J, Xu L, Ye L. Molecular Basis of the Differentiation and Function of Virus Specific Follicular Helper CD4 + T Cells. Front Immunol 2019; 10:249. [PMID: 30828337 PMCID: PMC6384271 DOI: 10.3389/fimmu.2019.00249] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 01/29/2019] [Indexed: 12/12/2022] Open
Abstract
During viral infection, virus-specific follicular helper T cells provide important help to cognate B cells for their survival, consecutive proliferation and mutation and eventual differentiation into memory B cells and antibody-secreting plasma cells. Similar to Tfh cells generated in other conditions, the differentiation of virus-specific Tfh cells can also be characterized as a process involved multiple factors and stages, however, which also exhibits distinct features. Here, we mainly focus on the current understanding of Tfh fate commitment, functional maturation, lineage maintenance and memory transition and formation in the context of viral infection.
Collapse
Affiliation(s)
- Qizhao Huang
- Cancer Center, The General Hospital of Western Theater Command, Chengdu, China.,Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Jianjun Hu
- Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Jianfang Tang
- Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Lifan Xu
- Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Lilin Ye
- Institute of Immunology, Third Military Medical University, Chongqing, China
| |
Collapse
|
6
|
Nazerai L, Schøller AS, Rasmussen POS, Buus S, Stryhn A, Christensen JP, Thomsen AR. A New In Vivo Model to Study Protective Immunity to Zika Virus Infection in Mice With Intact Type I Interferon Signaling. Front Immunol 2018; 9:593. [PMID: 29623081 PMCID: PMC5874300 DOI: 10.3389/fimmu.2018.00593] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 03/09/2018] [Indexed: 12/12/2022] Open
Abstract
The association between recent Zika virus (ZIKV) infection and neurological complications, microcephaly in the fetus, and Guillain–Barré syndrome in adults underscores the necessity for a protective vaccine. Rational vaccine development requires an in-depth understanding of the mechanisms which could protect against infection with this virus. However, so far, such an analysis has been hampered by the absence of a suitable small animal model. Unlike the situation in humans, ZIKV only replicates effectively in the peripheral organs of mice, if type I IFN signaling is interrupted. As type I IFN also impacts the adaptive immune response, mice with such a defect are not optimal for a comprehensive immunological analysis. In this report, we show that even in wild-type (WT) mice i.c. infection with low doses of virus causes marked local virus replication and lethal encephalitis in naïve mice. Furthermore, peripheral infection of WT mice with low doses of virus induces a significant immune response, which provides long-lasting protection of WT mice from a fatal outcome of subsequent i.c. challenge. Therefore, combining peripheral priming with later i.c. challenge represents a new approach for studying the adaptive immune response to ZIKV in mice with an intact type I IFN response. In this study, we focused on the mechanisms underlying resistance to reinfection. Using a combination of adoptive transfer, antibody-based cell depletion, and gene targeting, we show that the key protective factor in type I IFN replete mice is humoral immunity. CD8 T cells are not essential in mice with preformed specific antibodies, but under conditions where initial antibody levels are low, effector CD8 T cells may play a role as a back-up system. These results have important implications for our understanding of natural immunity to ZIKV infection and for Zika vaccine design.
Collapse
Affiliation(s)
- Loulieta Nazerai
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Amalie Skak Schøller
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | | | - Søren Buus
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Anette Stryhn
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | | | - Allan Randrup Thomsen
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
7
|
Balathasan L, Tang VA, Yadollahi B, Brun J, Labelle M, Lefebvre C, Swift SL, Stojdl DF. Activating Peripheral Innate Immunity Enables Safe and Effective Oncolytic Virotherapy in the Brain. MOLECULAR THERAPY-ONCOLYTICS 2017; 7:45-56. [PMID: 29062886 PMCID: PMC5645178 DOI: 10.1016/j.omto.2017.09.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 09/08/2017] [Indexed: 11/29/2022]
Abstract
The oncolytic mutant vesicular stomatitis virus VSVΔ51 achieves robust efficacy in multiple extracranial tumor models. Yet for malignancies of the brain, direct intratumoral infusion of VSVΔ51 causes lethal virus-induced neuropathology. Here, we have developed a novel therapeutic regime that uses peripheral immunization with a single sub-lethal dose of VSVΔ51 to establish an acute anti-viral state that enables the safe intracranial (IC) infusion of an otherwise lethal dose of VSVΔ51 within just 6 hr. Although type I interferons alone appeared insufficient to explain this protective phenotype, serum isolated at early time points from primed animals conferred protection against an IC dose of virus. Adaptive immune populations had minimal contributions. Finally, the therapeutic utility of this novel strategy was demonstrated by peripherally priming and intracranially treating mice bearing aggressive CT2A syngeneic astrocytomas with VSVΔ51. Approximately 25% of animals achieved complete regression of established tumors, with no signs of virus-induced neurological impairment. This approach may harness an early warning system in the brain that has evolved to protect the host against otherwise lethal neurotropic viral infections. We have exploited this protective mechanism to safely and efficaciously treat brain tumors with an otherwise neurotoxic virus, potentially widening the available treatment options for oncolytic virotherapy in the brain.
Collapse
Affiliation(s)
- Lukxmi Balathasan
- Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON K1H 8L1, Canada
| | - Vera A Tang
- Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON K1H 8L1, Canada
| | - Beta Yadollahi
- Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON K1H 8L1, Canada.,Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada
| | - Jan Brun
- Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON K1H 8L1, Canada
| | - Melanie Labelle
- Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON K1H 8L1, Canada
| | - Charles Lefebvre
- Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON K1H 8L1, Canada
| | - Stephanie L Swift
- Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON K1H 8L1, Canada
| | - David F Stojdl
- Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON K1H 8L1, Canada.,Department of Biology, Microbiology and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5, Canada.,Department of Pediatrics, University of Ottawa, 75 Laurier Ave. E., Ottawa, ON K1N 6N5, Canada
| |
Collapse
|
8
|
Tokarz-Deptuła B, Malinowska M, Adamiak M, Deptuła W. Coronins and their role in immunological phenomena. Cent Eur J Immunol 2017; 41:435-441. [PMID: 28450807 PMCID: PMC5382889 DOI: 10.5114/ceji.2016.65143] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 01/06/2016] [Indexed: 02/05/2023] Open
Abstract
Coronins are a large family of proteins occurring in many eukaryotes. In mammals, seven coronin genes have been identified, evidencing that coronins 1 to 6 present classic coronin structure, while coronin 7 is a tandem coronin particle, without a spiral domain, although the best characterised coronin, in terms of both structure and function, is the mammalian coronin 1. It has been proven that they are related to regulation of actin dynamics, e.g. as a result of interaction with the complex of proteins Arp2/3. These proteins also modulate the activity of immune system cells, including lymphocyte T and B cells, neutrophils and macrophages. They are involved in bacterial infections with Mycobacterium tuberculosis, M. leprae and Helicobacter pylori and participate in the response to viral infections, e.g. infections of lymphocytic choriomeningitis virus (LCMV) and vesicular stomatitis Indiana virus (VSV). Also their involvement in autoimmune diseases such as lupus erythematosus has been recorded.
Collapse
Affiliation(s)
| | | | - Mateusz Adamiak
- Department of Immunology, Faculty of Biology, University of Szczecin, Poland
| | - Wiesław Deptuła
- Department of Immunology, Faculty of Biology, University of Szczecin, Poland
| |
Collapse
|
9
|
Falls T, Roy DG, Bell JC, Bourgeois-Daigneault MC. Murine Tumor Models for Oncolytic Rhabdo-Virotherapy. ILAR J 2017; 57:73-85. [PMID: 27034397 DOI: 10.1093/ilar/ilv048] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The preclinical optimization and validation of novel treatments for cancer therapy requires the use of laboratory animals. Although in vitro experiments using tumor cell lines and ex vivo treatment of patient tumor samples provide a remarkable first-line tool for the initial study of tumoricidal potential, tumor-bearing animals remain the primary option to study delivery, efficacy, and safety of therapies in the context of a complete tumor microenvironment and functional immune system. In this review, we will describe the use of murine tumor models for oncolytic virotherapy using vesicular stomatitis virus. We will discuss studies using immunocompetent and immunodeficient models with respect to toxicity and therapeutic treatments, as well as the various techniques and tools available to study cancer therapy with Rhabdoviruses.
Collapse
Affiliation(s)
- Theresa Falls
- Theresa Falls is a research technician at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada. Dominic Guy Roy is a Ph.D candidate at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and a Ph.D candidate in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada. John Cameron Bell is a senior researcher at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and professor in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada. Marie-Claude Bourgeois-Daigneault is a postdoctoral fellow at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and a postdoctoral fellow in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada
| | - Dominic Guy Roy
- Theresa Falls is a research technician at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada. Dominic Guy Roy is a Ph.D candidate at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and a Ph.D candidate in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada. John Cameron Bell is a senior researcher at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and professor in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada. Marie-Claude Bourgeois-Daigneault is a postdoctoral fellow at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and a postdoctoral fellow in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada
| | - John Cameron Bell
- Theresa Falls is a research technician at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada. Dominic Guy Roy is a Ph.D candidate at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and a Ph.D candidate in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada. John Cameron Bell is a senior researcher at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and professor in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada. Marie-Claude Bourgeois-Daigneault is a postdoctoral fellow at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and a postdoctoral fellow in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada
| | - Marie-Claude Bourgeois-Daigneault
- Theresa Falls is a research technician at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada. Dominic Guy Roy is a Ph.D candidate at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and a Ph.D candidate in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada. John Cameron Bell is a senior researcher at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and professor in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada. Marie-Claude Bourgeois-Daigneault is a postdoctoral fellow at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and a postdoctoral fellow in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada
| |
Collapse
|
10
|
Access of protective antiviral antibody to neuronal tissues requires CD4 T-cell help. Nature 2016; 533:552-6. [PMID: 27225131 PMCID: PMC4883597 DOI: 10.1038/nature17979] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Accepted: 04/05/2016] [Indexed: 12/24/2022]
Abstract
Circulating antibodies can access most tissues to mediate surveillance and elimination of invading pathogens. Immunopriviledged tissues such as the brain and the peripheral nervous system, are shielded from plasma proteins, by the blood-brain barrier1 and blood nerve barrier2, respectively. Yet, circulating antibodies must somehow gain access to these tissues in order to mediate their antimicrobial functions. Here, we examine the mechanism by which antibodies gain access to neuronal tissues to control infection. Using mouse model of genital herpes infection, we demonstrate that both antibodies and CD4 T cells are required to protect the host following immunization at a distal site. We show that memory CD4 T cells migrate to the dorsal root ganglia (DRG) and spinal cord in response to HSV-2 infection. Once inside these neuronal tissues, CD4 T cells secrete interferon (IFN)-γ and mediate local increase in vascular permeability, enabling antibody access for viral control. A similar requirement for CD4 T cells for antibody access to the brain was observed following intranasal challenge with vesicular stomatitis virus. Our results reveal a previously unappreciated role of CD4 help in mobilizing antibodies to the peripheral sites of infection where they help to limit viral spread.
Collapse
|
11
|
Andreasen LV, Hansen LB, Andersen P, Agger EM, Dietrich J. Aluminium hydroxide potentiates a protective Th1 biased immune response against polio virus that allows for dose sparing in mice and rats. Vaccine 2015; 33:1873-9. [DOI: 10.1016/j.vaccine.2015.02.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 12/08/2014] [Accepted: 02/04/2015] [Indexed: 01/27/2023]
|
12
|
Spanier J, Lienenklaus S, Paijo J, Kessler A, Borst K, Heindorf S, Baker DP, Kröger A, Weiss S, Detje CN, Staeheli P, Kalinke U. Concomitant TLR/RLH signaling of radioresistant and radiosensitive cells is essential for protection against vesicular stomatitis virus infection. THE JOURNAL OF IMMUNOLOGY 2014; 193:3045-54. [PMID: 25127863 DOI: 10.4049/jimmunol.1400959] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Several studies indicated that TLR as well as retinoic acid-inducible gene I-like helicase (RLH) signaling contribute to vesicular stomatitis virus (VSV)-mediated triggering of type I IFN (IFN-I) responses. Nevertheless, TLR-deficient MyD88(-/-)Trif(-/-) mice and RLH-deficient caspase activation and recruitment domain adaptor inducing IFN-β (Cardif)(-/-) mice showed only marginally enhanced susceptibility to lethal VSV i.v. infection. Therefore, we addressed whether concomitant TLR and RLH signaling, or some other additional mechanism, played a role. To this end, we generated MyD88(-/-)Trif(-/-)Cardif(-/-) (MyTrCa(-/-)) mice that succumbed to low-dose i.v. VSV infection with similar kinetics as IFN-I receptor-deficient mice. Three independent approaches (i.e., analysis of IFN-α/β serum levels, experiments with IFN-β reporter mice, and investigation of local IFN-stimulated gene induction) revealed that MyTrCa(-/-) mice did not mount IFN-I responses following VSV infection. Of note, treatment with rIFN-α protected the animals, qualifying MyTrCa(-/-) mice as a model to study the contribution of different immune cell subsets to the production of antiviral IFN-I. Upon adoptive transfer of wild-type plasmacytoid dendritic cells and subsequent VSV infection, MyTrCa(-/-) mice displayed significantly reduced viral loads in peripheral organs and showed prolonged survival. On the contrary, adoptive transfer of wild-type myeloid dendritic cells did not have such effects. Analysis of bone marrow chimeric mice revealed that TLR and RLH signaling of radioresistant and radiosensitive cells was required for efficient protection. Thus, upon VSV infection, plasmacytoid dendritic cell-derived IFN-I primarily protects peripheral organs, whereas concomitant TLR and RLH signaling of radioresistant stroma cells as well as of radiosensitive immune cells is needed to effectively protect against lethal disease.
Collapse
Affiliation(s)
- Julia Spanier
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research, Braunschweig, and the Hannover Medical School, 30625 Hannover, Germany
| | - Stefan Lienenklaus
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Jennifer Paijo
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research, Braunschweig, and the Hannover Medical School, 30625 Hannover, Germany
| | - Annett Kessler
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research, Braunschweig, and the Hannover Medical School, 30625 Hannover, Germany
| | - Katharina Borst
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research, Braunschweig, and the Hannover Medical School, 30625 Hannover, Germany
| | - Sabrina Heindorf
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research, Braunschweig, and the Hannover Medical School, 30625 Hannover, Germany
| | | | - Andrea Kröger
- Research Group on Innate Immunity and Infection, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; and
| | - Siegfried Weiss
- Department of Molecular Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Claudia N Detje
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research, Braunschweig, and the Hannover Medical School, 30625 Hannover, Germany
| | - Peter Staeheli
- Department of Virology, Institute for Medical Microbiology and Hygiene, Albert-Ludwigs-Universität, 79104 Freiburg, Germany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research, Braunschweig, and the Hannover Medical School, 30625 Hannover, Germany;
| |
Collapse
|
13
|
Dietrich J, Andreasen LV, Andersen P, Agger EM. Inducing dose sparing with inactivated polio virus formulated in adjuvant CAF01. PLoS One 2014; 9:e100879. [PMID: 24956110 PMCID: PMC4067388 DOI: 10.1371/journal.pone.0100879] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 05/30/2014] [Indexed: 11/25/2022] Open
Abstract
The development of new low cost inactivated polio virus based vaccines (IPV) is a high priority, and will be required to eradicate polio. In addition, such a vaccine constitutes the only realistic polio vaccine in the post-eradication era. One way to reduce the cost of a vaccine is to increase immunogenicity by use of adjuvants. The CAF01 adjuvant has previously been shown to be a safe and potent adjuvant with several antigens, and here we show that in mice IPV formulated with CAF01 induced increased systemic protective immunity measured by binding and neutralization antibody titers in serum. CAF01 also influenced the kinetics of both the cellular and humoral response against IPV to produce a faster, as well as a stronger, response, dominated by IgG2a, IgG2b, and IgG2c isotypes as well as IPV specific T cells secreting IFN-γ/IL-2. Finally, as intestinal immunity is also a priority of polio vaccines, we present a vaccine strategy based on simultaneous priming at an intradermal and an intramuscular site that generate intestinal immune responses against polio virus. Taken together, the IPV-CAF01 formulation constitutes a new promising vaccine against polio with the ability to generate strong humoral and cellular immunity against the polio virus.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/pharmacology
- Animals
- Antibodies, Viral/immunology
- Chemistry, Pharmaceutical
- Dose-Response Relationship, Immunologic
- Female
- Immunity, Cellular/drug effects
- Immunity, Cellular/immunology
- Immunoglobulin A/immunology
- Immunoglobulin G/metabolism
- Injections, Intradermal
- Injections, Intramuscular
- Intestinal Mucosa/metabolism
- Intestines/drug effects
- Mice, Inbred C57BL
- Neutralization Tests
- Poliovirus/drug effects
- Poliovirus/immunology
- Poliovirus Vaccine, Inactivated/administration & dosage
- Poliovirus Vaccine, Inactivated/immunology
- Vaccination
Collapse
Affiliation(s)
- Jes Dietrich
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
- * E-mail:
| | - Lars Vibe Andreasen
- Department of Vaccine Development, Statens Serum Institut, Copenhagen, Denmark
| | - Peter Andersen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Else Marie Agger
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| |
Collapse
|
14
|
Quantification of B16 melanoma cells in lungs using triplex Q-PCR--a new approach to evaluate melanoma cell metastasis and tumor control. PLoS One 2014; 9:e87831. [PMID: 24498205 PMCID: PMC3909236 DOI: 10.1371/journal.pone.0087831] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 12/30/2013] [Indexed: 01/14/2023] Open
Abstract
Skin cancer is the most common type of all cancers. However, it comprises several different types of cancers, one of which is malignant melanoma. Even though melanomas only make up about 5% of skin cancers, they are responsible for the majority of skin cancer deaths due to the poor chance of survival once the tumor has metastasized. In the present study, we have developed a new assay for quantitative analysis of B16 melanoma metastasis in the lungs. We have used a triplex Q-PCR to determine the expression of the melanoma genes GP100/Pmel and tyrosinase-related protein 2 (TRP-2), and found that B16.F10gp cells were detectable in the lungs as early as 2 hours after intravenous challenge with ≥104 tumor cells. When investigating the gene expression as a function of time, we observed a gradual decrease from 2–24 hours post tumor challenge followed by an increase of approximately 2 log10 on day 11. The early decrease was accelerated in the presence of activated NK cells. To further evaluate our assay, we also investigated the level of metastasis in the context of vaccination with replication defective adenoviral vectors, Ad-Ii-GP and Ad-GP, previously found to significantly delay the outgrowth of subcutaneous melanomas. Results obtained using Q-PCR were compared to conventional counting of metastatic foci under a dissection microscope. A marked reduction in gene expression was observed in the lungs after vaccination with both vectors; however, Ad-Ii-GP showed the highest protection, and matching results were obtained by enumeration of visible tumor nodules on the lung surfaces. Finally, we could show that inhibition of tumor metastasis required antigen-specific CD8 T cells and IFNγ, but not perforin. In conclusion, the presented results validate triplex Q-PCR as a fast, objective, and quantitative method for analysis of melanoma metastasis in the lungs.
Collapse
|
15
|
Steel CD, Breving K, Tavakoli S, Kim WK, Sanford LD, Ciavarra RP. Role of peripheral immune response in microglia activation and regulation of brain chemokine and proinflammatory cytokine responses induced during VSV encephalitis. J Neuroimmunol 2013; 267:50-60. [PMID: 24369299 DOI: 10.1016/j.jneuroim.2013.12.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 11/25/2013] [Accepted: 12/04/2013] [Indexed: 11/25/2022]
Abstract
We report herein that neuroinvasion by vesicular stomatitis virus (VSV) activates microglia and induces a peripheral dendritic cell (DC)-dependent inflammatory response in the central nervous system (CNS). VSV neuroinvasion rapidly induces multiple brain chemokine and proinflammatory cytokine mRNAs that display bimodal kinetics. Peripheral DC ablation or T cell depletion suppresses the second wave of this response demonstrating that infiltrating T cells are primarily responsible for the bimodal characteristics of this response. The robust infiltrate associated with VSV encephalitis likely depends on sustained production of brain CCL19 and CCR7 expression on infiltrating inflammatory cells.
Collapse
Affiliation(s)
- Christina D Steel
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA 23501, United States
| | - Kimberly Breving
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA 23501, United States
| | - Susan Tavakoli
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA 23501, United States
| | - Woong-Ki Kim
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA 23501, United States
| | - Larry D Sanford
- Department of Pathology and Anatomy, Eastern Virginia Medical School, Norfolk, VA 23501, United States
| | - Richard P Ciavarra
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA 23501, United States.
| |
Collapse
|
16
|
Tchang VSY, Mekker A, Siegmund K, Karrer U, Pieters J. Diverging role for coronin 1 in antiviral CD4+ and CD8+ T cell responses. Mol Immunol 2013; 56:683-92. [DOI: 10.1016/j.molimm.2013.05.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 04/24/2013] [Accepted: 05/04/2013] [Indexed: 11/24/2022]
|
17
|
Blackham AU, Northrup SA, Willingham M, Sirintrapun J, Russell GB, Lyles DS, Stewart JH. Molecular determinants of susceptibility to oncolytic vesicular stomatitis virus in pancreatic adenocarcinoma. J Surg Res 2013; 187:412-26. [PMID: 24252853 DOI: 10.1016/j.jss.2013.10.032] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 10/03/2013] [Accepted: 10/17/2013] [Indexed: 11/26/2022]
Abstract
BACKGROUND M protein mutant vesicular stomatitis virus (M51R-VSV) has oncolytic properties against many cancers. However, some cancer cells are resistant to M51R-VSV. Herein, we evaluate the molecular determinants of vesicular stomatitis virus (VSV) resistance in pancreatic adenocarcinoma cells. METHODS Cell viability and the effect of β-interferon (IFN) were analyzed using 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium assay. Gene expression was evaluated via microarray analysis. Cell infectability was measured by flow cytometry. Xenografts were established in athymic nude mice and treated with intratumoral M51R-VSV. RESULTS Four of five pancreatic cancer cell lines were sensitive to M51R-VSV, whereas Panc 03.27 cells remained resistant (81 ± 3% viability 72 h after single-cycle infection). Comparing sensitive MiaPaCa2 cells with resistant Panc 03.27 cells, significant differences in gene expression were found relating to IFN signaling (P = 2 × 10(-5)), viral entry (P = 3 × 10(-4)), and endocytosis (P = 7 × 10(-4)). MiaPaCa2 cells permitted high levels of VSV infection, whereas Panc 03.27 cells were capable of resisting VSV cell entry even at high multiplicities of infection. Extrinsic β-IFN overcame apparent defects in IFN-mediated pathways in MiaPaCa2 cells conferring VSV resistance. In contrast, β-IFN decreased cell viability in Panc 3.27 cells, suggesting intact antiviral mechanisms. VSV-treated xenografts exhibited reduced tumor growth relative to controls in both MiaPaCa2 (1423 ± 345% versus 164 ± 136%; P < 0.001) and Panc 3.27 (979 ± 153% versus 50 ± 56%; P = 0.002) tumors. Significant lymphocytic infiltration was seen in M51R-VSV-treated Panc 03.27 xenografts. CONCLUSIONS Inhibition of VSV endocytosis and intact IFN-mediated defenses are responsible for M51R-VSV resistance in pancreatic adenocarcinoma cells. M51R-VSV treatment appears to induce antitumor cellular immunity in vivo, which may expand its clinical efficacy.
Collapse
Affiliation(s)
- Aaron U Blackham
- Division of Surgical Sciences, Department of General Surgery, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Scott A Northrup
- Division of Surgical Sciences, Department of General Surgery, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Mark Willingham
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Joseph Sirintrapun
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Greg B Russell
- Division of Public Health Sciences, Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Douglas S Lyles
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - John H Stewart
- Division of Surgical Sciences, Department of General Surgery, Wake Forest School of Medicine, Winston-Salem, North Carolina.
| |
Collapse
|
18
|
Hastie E, Cataldi M, Marriott I, Grdzelishvili VZ. Understanding and altering cell tropism of vesicular stomatitis virus. Virus Res 2013; 176:16-32. [PMID: 23796410 DOI: 10.1016/j.virusres.2013.06.003] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2013] [Revised: 06/06/2013] [Accepted: 06/07/2013] [Indexed: 12/18/2022]
Abstract
Vesicular stomatitis virus (VSV) is a prototypic nonsegmented negative-strand RNA virus. VSV's broad cell tropism makes it a popular model virus for many basic research applications. In addition, a lack of preexisting human immunity against VSV, inherent oncotropism and other features make VSV a widely used platform for vaccine and oncolytic vectors. However, VSV's neurotropism that can result in viral encephalitis in experimental animals needs to be addressed for the use of the virus as a safe vector. Therefore, it is very important to understand the determinants of VSV tropism and develop strategies to alter it. VSV glycoprotein (G) and matrix (M) protein play major roles in its cell tropism. VSV G protein is responsible for VSV broad cell tropism and is often used for pseudotyping other viruses. VSV M affects cell tropism via evasion of antiviral responses, and M mutants can be used to limit cell tropism to cell types defective in interferon signaling. In addition, other VSV proteins and host proteins may function as determinants of VSV cell tropism. Various approaches have been successfully used to alter VSV tropism to benefit basic research and clinically relevant applications.
Collapse
Affiliation(s)
- Eric Hastie
- Department of Biology, University of North Carolina at Charlotte, 9201 University City Boulevard, Charlotte, NC 28223, United States
| | | | | | | |
Collapse
|
19
|
Eisenstein S, Coakley BA, Briley-Saebo K, Ma G, Chen HM, Meseck M, Ward S, Divino C, Woo S, Chen SH, Pan PY. Myeloid-derived suppressor cells as a vehicle for tumor-specific oncolytic viral therapy. Cancer Res 2013; 73:5003-15. [PMID: 23536556 DOI: 10.1158/0008-5472.can-12-1597] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
One of the several impediments to effective oncolytic virus therapy of cancer remains a lack of tumor-specific targeting. Myeloid-derived suppressor cells (MDSC) are immature myeloid cells induced by tumor factors in tumor-bearing hosts. The biodistribution kinetics of MDSC and other immune cell types in a murine hepatic colon cancer model was investigated through the use of tracking markers and MRI. MDSCs were superior to other immune cell types in preferential migration to tumors in comparison with other tissues. On the basis of this observation, we engineered a strain of vesicular stomatitis virus (VSV), an oncolytic rhabdovirus that bound MDSCs and used them as a delivery vehicle. Improving VSV-binding efficiency to MDSCs extended the long-term survival of mice bearing metastatic colon tumors compared with systemic administration of wild-type VSV alone. Survival was further extended by multiple injections of the engineered virus without significant toxicity. Notably, direct tumor killing was accentuated by promoting MDSC differentiation towards the classically activated M1-like phenotype. Our results offer a preclinical proof-of-concept for using MDSCs to facilitate and enhance the tumor-killing activity of tumor-targeted oncolytic therapeutics.
Collapse
Affiliation(s)
- Samuel Eisenstein
- Department of Oncological Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Moseman EA, Iannacone M, Bosurgi L, Tonti E, Chevrier N, Tumanov A, Fu YX, Hacohen N, von Andrian UH. B cell maintenance of subcapsular sinus macrophages protects against a fatal viral infection independent of adaptive immunity. Immunity 2012; 36:415-26. [PMID: 22386268 DOI: 10.1016/j.immuni.2012.01.013] [Citation(s) in RCA: 124] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2011] [Revised: 12/14/2011] [Accepted: 01/24/2012] [Indexed: 01/12/2023]
Abstract
Neutralizing antibodies have been thought to be required for protection against acutely cytopathic viruses, such as the neurotropic vesicular stomatitis virus (VSV). Utilizing mice that possess B cells but lack antibodies, we show here that survival upon subcutaneous (s.c.) VSV challenge was independent of neutralizing antibody production or cell-mediated adaptive immunity. However, B cells were absolutely required to provide lymphotoxin (LT) α1β2, which maintained a protective subcapsular sinus (SCS) macrophage phenotype within virus draining lymph nodes (LNs). Macrophages within the SCS of B cell-deficient LNs, or of mice that lack LTα1β2 selectively in B cells, displayed an aberrant phenotype, failed to replicate VSV, and therefore did not produce type I interferons, which were required to prevent fatal VSV invasion of intranodal nerves. Thus, although B cells are essential for survival during VSV infection, their contribution involves the provision of innate differentiation and maintenance signals to macrophages, rather than adaptive immune mechanisms.
Collapse
Affiliation(s)
- E Ashley Moseman
- Immune Disease Institute and Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Swain SL, McKinstry KK, Strutt TM. Expanding roles for CD4⁺ T cells in immunity to viruses. Nat Rev Immunol 2012; 12:136-48. [PMID: 22266691 PMCID: PMC3764486 DOI: 10.1038/nri3152] [Citation(s) in RCA: 656] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
CD4+ T cells are orchestrators, regulators and direct effectors of antiviral immunity. Neutralizing antibodies provide protection against many viral pathogens, and CD4+ T cells can help B cells to generate stronger and longer-lived antibody responses. CD4+ T cells help antiviral CD8+ T cells in two main ways: they maximize CD8+ T cell population expansion during a primary immune response and also facilitate the generation of virus-specific memory CD8+ T cell populations. In addition to their helper functions, CD4+ T cells contribute directly to viral clearance. They secrete cytokines with antiviral activities and, in some circumstances, can eliminate infected cells through cytotoxic killing. Memory CD4+ T cells provide superior protection during re-infection with a virus. Compared with new effector CD4+ T cells, memory CD4+ T cells have enhanced helper and effector functions and can rapidly trigger innate immune defence mechanisms early in the infection.
Immunity to viruses is typically associated with the development of cytotoxic CD8+ T cells. However, CD4+ T cells are also important for protection during viral infection. Here, the authors describe the various ways in which different CD4+T cell subsets can contribute to the antiviral immune response. Viral pathogens often induce strong effector CD4+ T cell responses that are best known for their ability to help B cell and CD8+ T cell responses. However, recent studies have uncovered additional roles for CD4+ T cells, some of which are independent of other lymphocytes, and have described previously unappreciated functions for memory CD4+ T cells in immunity to viruses. Here, we review the full range of antiviral functions of CD4+ T cells, discussing the activities of these cells in helping other lymphocytes and in inducing innate immune responses, as well as their direct antiviral roles. We suggest that all of these functions of CD4+ T cells are integrated to provide highly effective immune protection against viral pathogens.
Collapse
Affiliation(s)
- Susan L Swain
- Department of Pathology, University of Massachusetts Medical School, 55 Lake Avenue N, Worcester, Massachusetts 01655, USA.
| | | | | |
Collapse
|
22
|
Abstract
Members of the family Rhabdoviridae are single-stranded RNA viruses and globally important pathogens of wild and cultured fish and thus relatively well studied in their respective hosts or other model systems. Here, we review the protective immune mechanisms that fish mount in response to rhabdovirus infections. Teleost fish possess the principal components of innate and adaptive immunity found in other vertebrates. Neutralizing antibodies are critical for long-term protection from fish rhabdoviruses, but several studies also indicate a role for cell-mediated immunity. Survival of acute rhabdoviral infection is also dependent on innate immunity, particularly the interferon (IFN) system that is rapidly induced in response to infection. Paradoxically, rhabdoviruses are sensitive to the effects of IFN but virulent rhabdoviruses can continue to replicate owing to the abilities of the matrix (M) protein to mediate host-cell shutoff and the non‑virion (NV) protein to subvert programmed cell death and suppress functional IFN. While many basic features of the fish immune response to rhabdovirus infections are becoming better understood, much less is known about how factors in the environment affect the ecology of rhabdovirus infections in natural populations of aquatic animals.
Collapse
|
23
|
Heiber JF, Xu XX, Barber GN. Potential of vesicular stomatitis virus as an oncolytic therapy for recurrent and drug-resistant ovarian cancer. CHINESE JOURNAL OF CANCER 2011; 30:805-14. [PMID: 22059911 PMCID: PMC4013328 DOI: 10.5732/cjc.011.10205] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In the last decade, we have gained significant understanding of the mechanism by which vesicular stomatitis virus (VSV) specifically kills cancer cells. Dysregulation of translation and defective innate immunity are both thought to contribute to VSV oncolysis. Safety and efficacy are important objectives to consider in evaluating VSV as a therapy for malignant disease. Ongoing efforts may enable VSV virotherapy to be considered in the near future to treat drug-resistant ovarian cancer when other options have been exhausted. In this article, we review the development of VSV as a potential therapeutic approach for recurrent or drug-resistant ovarian cancer.
Collapse
Affiliation(s)
- Joshua F Heiber
- University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | | | | |
Collapse
|
24
|
Vesicular stomatitis virus expressing tumor suppressor p53 is a highly attenuated, potent oncolytic agent. J Virol 2011; 85:10440-50. [PMID: 21813611 DOI: 10.1128/jvi.05408-11] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Vesicular stomatitis virus (VSV), a negative-strand RNA rhabdovirus, preferentially replicates in and eradicates transformed versus nontransformed cells and is thus being considered for use as a potential anticancer treatment. The genetic malleability of VSV also affords an opportunity to develop more potent agents that exhibit increased therapeutic activity. The tumor suppressor p53 has been shown to exert potent antitumor properties, which may in part involve stimulating host innate immune responses to malignancies. To evaluate whether VSV expressing p53 exhibited enhanced oncolytic action, the murine p53 (mp53) gene was incorporated into recombinant VSVs with or without a functional viral M gene-encoded protein that could either block (VSV-mp53) or enable [VSV-M(mut)-mp53] host mRNA export following infection of susceptible cells. Our results indicated that VSV-mp53 and VSV-M(mut)-mp53 expressed high levels of functional p53 and retained the ability to lyse transformed versus normal cells. In addition, we observed that VSV-ΔM-mp53 was extremely attenuated in vivo due to p53 activating innate immune genes, such as type I interferon (IFN). Significantly, immunocompetent animals with metastatic mammary adenocarcinoma exhibited increased survival following treatment with a single inoculation of VSV-ΔM-mp53, the mechanisms of which involved enhanced CD49b+ NK and tumor-specific CD8+ T cell responses. Our data indicate that VSV incorporating p53 could provide a safe, effective strategy for the design of VSV oncolytic therapeutics and VSV-based vaccines.
Collapse
|
25
|
Subcapsular sinus macrophages prevent CNS invasion on peripheral infection with a neurotropic virus. Nature 2010; 465:1079-83. [PMID: 20577213 PMCID: PMC2892812 DOI: 10.1038/nature09118] [Citation(s) in RCA: 264] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2010] [Accepted: 04/22/2010] [Indexed: 12/18/2022]
Abstract
Lymph nodes (LNs) capture microorganisms that breach the body’s external barriers and enter draining lymphatics, limiting the systemic spread of pathogens1. Recent work has shown that CD11b+CD169+ macrophages, which populate the subcapsular sinus (SCS) of LNs, are critical for clearance of viruses from the lymph and for initiating antiviral humoral immune responses2,3,4. Using vesicular stomatitis virus (VSV), a relative of rabies virus transmitted by insect bites, we show here that SCS macrophages perform a third vital function: they prevent lymph-borne neurotropic viruses from infecting the CNS. Upon local depletion of LN macrophages, ~60% of mice developed ascending paralysis and died 7–10 days after subcutaneous infection with a small dose of VSV, while macrophage-sufficient animals remained asymptomatic and cleared the virus. VSV gained access to the nervous system via peripheral nerves in macrophage-depleted LNs. In contrast, within macrophage-sufficient LNs VSV replicated preferentially within SCS macrophages but not in adjacent nerves. Removal of SCS macrophages did not compromise adaptive immune responses against VSV, but reduced type I interferon (IFN-I) production within infected LNs. VSV-infected macrophages recruited IFN-I producing plasmacytoid dendritic cells to the SCS and additionally were a major source of IFN-I themselves. Experiments in bone marrow chimeric mice revealed that IFN-I must act on both hematopoietic and stromal compartments, including the intranodal nerves, to prevent lethal VSV infection. These results identify SCS macrophages as crucial gatekeepers to the CNS that prevent fatal viral neuroinvasion upon peripheral infection.
Collapse
|
26
|
Tsuji-Kawahara S, Chikaishi T, Takeda E, Kato M, Kinoshita S, Kajiwara E, Takamura S, Miyazawa M. Persistence of viremia and production of neutralizing antibodies differentially regulated by polymorphic APOBEC3 and BAFF-R loci in friend virus-infected mice. J Virol 2010; 84:6082-95. [PMID: 20375169 PMCID: PMC2876660 DOI: 10.1128/jvi.02516-09] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Accepted: 03/26/2010] [Indexed: 02/03/2023] Open
Abstract
Several host genes control retroviral replication and pathogenesis through the regulation of immune responses to viral antigens. The Rfv3 gene influences the persistence of viremia and production of virus-neutralizing antibodies in mice infected with Friend mouse retrovirus complex (FV). This locus has been mapped within a narrow segment of mouse chromosome 15 harboring the APOBEC3 and BAFF-R loci, both of which show functional polymorphisms among different strains of mice. The exon 5-lacking product of the APOBEC3 allele expressed in FV-resistant C57BL/6 (B6) mice directly restricts viral replication, and mice lacking the B6-derived APOBEC3 exhibit exaggerated pathology and reduced production of neutralizing antibodies. However, the mechanisms by which the polymorphisms at the APOBEC3 locus affect the production of neutralizing antibodies remain unclear. Here we show that the APOBEC3 genotypes do not directly affect the B-cell repertoire, and mice lacking B6-derived APOBEC3 still produce FV-neutralizing antibodies in the presence of primed T helper cells. Instead, higher viral loads at a very early stage of FV infection caused by either a lack of the B6-derived APOBEC3 or a lack of the wild-type BAFF-R resulted in slower production of neutralizing antibodies. Indeed, B cells were hyperactivated soon after infection in the APOBEC3- or BAFF-R-deficient mice. In contrast to mice deficient in the B6-derived APOBEC3, which cleared viremia by 4 weeks after FV infection, mice lacking the functional BAFF-R allele exhibited sustained viremia, indicating that the polymorphisms at the BAFF-R locus may better explain the Rfv3-defining phenotype of persistent viremia.
Collapse
Affiliation(s)
- Sachiyo Tsuji-Kawahara
- Departments of Immunology, Dermatology, Kinki University School of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan, UMN Pharma Inc., Yokohama 222-0033, Japan
| | - Tomomi Chikaishi
- Departments of Immunology, Dermatology, Kinki University School of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan, UMN Pharma Inc., Yokohama 222-0033, Japan
| | - Eri Takeda
- Departments of Immunology, Dermatology, Kinki University School of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan, UMN Pharma Inc., Yokohama 222-0033, Japan
| | - Maiko Kato
- Departments of Immunology, Dermatology, Kinki University School of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan, UMN Pharma Inc., Yokohama 222-0033, Japan
| | - Saori Kinoshita
- Departments of Immunology, Dermatology, Kinki University School of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan, UMN Pharma Inc., Yokohama 222-0033, Japan
| | - Eiji Kajiwara
- Departments of Immunology, Dermatology, Kinki University School of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan, UMN Pharma Inc., Yokohama 222-0033, Japan
| | - Shiki Takamura
- Departments of Immunology, Dermatology, Kinki University School of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan, UMN Pharma Inc., Yokohama 222-0033, Japan
| | - Masaaki Miyazawa
- Departments of Immunology, Dermatology, Kinki University School of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan, UMN Pharma Inc., Yokohama 222-0033, Japan
| |
Collapse
|
27
|
Bartholdy C, Christensen JE, Grujic M, Christensen JP, Thomsen AR. T-cell intrinsic expression of MyD88 is required for sustained expansion of the virus-specific CD8+ T-cell population in LCMV-infected mice. J Gen Virol 2009; 90:423-431. [PMID: 19141452 DOI: 10.1099/vir.0.004960-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Acute infection with lymphocytic choriomeningitis virus (LCMV) normally results in robust clonal expansion of virus-specific CD8(+) T cells, which in turn control the primary infection. However, similar infection of myeloid differentiation factor 88 (MyD88)-deficient mice leads to a markedly impaired T-cell response and chronic infection. It has been found previously that impairment of the innate immune response is not sufficient to explain this profound change in outcome. Using adoptive transfer of CD8(+) T cells, this study demonstrated unequivocally that T-cell expression of MyD88 is critical for a normal T-cell response to LCMV. In addition, it was found that expression of MyD88 is superfluous during early activation and proliferation of the antigen-activated CD8(+) T cells, but plays a critical role in the sustained expansion of the antigen-specific CD8(+) T-cell population during the primary T-cell response. Interestingly, a critical role for MyD88 was evident only under conditions of systemic infection with virus capable of causing prolonged infection, suggesting that MyD88 expression may function as an internal regulator of the threshold for antigen-driven, exhaustive differentiation.
Collapse
Affiliation(s)
- Christina Bartholdy
- Institute of International Health, Immunology and Microbiology, University of Copenhagen, The Panum Institute, 3C Blegdamsvej, Copenhagen, DK-2200, Denmark
| | - Jeanette E Christensen
- Institute of International Health, Immunology and Microbiology, University of Copenhagen, The Panum Institute, 3C Blegdamsvej, Copenhagen, DK-2200, Denmark
| | - Mirjana Grujic
- Institute of International Health, Immunology and Microbiology, University of Copenhagen, The Panum Institute, 3C Blegdamsvej, Copenhagen, DK-2200, Denmark
| | - Jan P Christensen
- Institute of International Health, Immunology and Microbiology, University of Copenhagen, The Panum Institute, 3C Blegdamsvej, Copenhagen, DK-2200, Denmark
| | - Allan R Thomsen
- Institute of International Health, Immunology and Microbiology, University of Copenhagen, The Panum Institute, 3C Blegdamsvej, Copenhagen, DK-2200, Denmark
| |
Collapse
|
28
|
Detje CN, Meyer T, Schmidt H, Kreuz D, Rose JK, Bechmann I, Prinz M, Kalinke U. Local type I IFN receptor signaling protects against virus spread within the central nervous system. THE JOURNAL OF IMMUNOLOGY 2009; 182:2297-304. [PMID: 19201884 DOI: 10.4049/jimmunol.0800596] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Several neurotropic viruses such as vesicular stomatitis virus (VSV) induce peripheral neutralizing Ab responses and still can infect cells within the CNS. To address whether local type I IFN receptor (IFNAR) triggering plays a role in controlling virus replication within the brain, we generated mice with a cell type-specific IFNAR deletion in neuroectodermal cells of the CNS (NesCre(+/-)IFNAR(flox/flox)). Intranasal VSV infection with 10(3) PFU was well tolerated by wild-type mice, whereas conventional IFNAR(-/-) mice died within 2-3 days. In contrast, brain-specific NesCre(+/-)IFNAR(flox/flox) mice survived until day 5-6 and then became hemiplegic and died. Terminally ill NesCre(+/-)IFNAR(flox/flox) mice showed 10- to 100-fold higher virus loads in the brain than IFNAR(-/-) mice, whereas little or no virus was found in other organs. In wild-type animals, virus could be reisolated only from the olfactory bulb until day 6 where also STAT1 activation as a measure of IFNAR triggering was detected. Virus infection was found exclusively in glomerular structures of the olfactory bulb, whereas surrounding cells that showed STAT1 phosphorylation as a measure of IFNAR trigging were free of virus. Our data indicate that upon intranasal VSV instillation, early and localized IFNAR triggering in the glomerular layer of the olfactory bulb is critically required to prevent viral spread over the entire CNS and thus confers survival.
Collapse
Affiliation(s)
- Claudia N Detje
- Division of Immunology, Paul-Ehrlich-Institut, Langen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Holst PJ, Sorensen MR, Mandrup Jensen CM, Orskov C, Thomsen AR, Christensen JP. MHC class II-associated invariant chain linkage of antigen dramatically improves cell-mediated immunity induced by adenovirus vaccines. THE JOURNAL OF IMMUNOLOGY 2008; 180:3339-46. [PMID: 18292559 DOI: 10.4049/jimmunol.180.5.3339] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The ideal vaccine induces a potent protective immune response, which should be rapidly induced, long-standing, and of broad specificity. Recombinant adenoviral vectors induce potent Ab and CD8+ T cell responses against transgenic Ags within weeks of administration, and they are among the most potent and versatile Ag delivery vehicles available. However, the impact of chronic infections like HIV and hepatitis C virus underscore the need for further improvements. In this study, we show that the protective immune response to an adenovirus-encoded vaccine Ag can be accelerated, enhanced, broadened, and prolonged by tethering of the rAg to the MHC class II-associated invariant chain (Ii). Thus, adenovirus-vectored vaccines expressing lymphocytic choriomeningitis virus (LCMV)-derived glycoprotein linked to Ii increased the CD4+ and CD8+ T cell stimulatory capacity in vitro and in vivo. Furthermore, mice vaccinated with a single dose of adenovirus-expressing LCMV-derived glycoprotein linked to Ii were protected against lethal virus-induced choriomeningitis, lethal challenge with strains mutated in immunodominant T cell epitopes, and systemic infection with a highly invasive strain. In therapeutic tumor vaccination, the vaccine was as efficient as live LCMV. In comparison, animals vaccinated with a conventional adenovirus vaccine expressing unmodified glycoprotein were protected against systemic infection, but only temporarily against lethal choriomeningitis, and this vaccine was less efficient in tumor therapy.
Collapse
Affiliation(s)
- Peter Johannes Holst
- Institute of International Health, Immunology and Microbiology, The Panum Institute, Copenhagen, Denmark
| | | | | | | | | | | |
Collapse
|
30
|
Grujic M, Christensen JP, Sørensen MR, Abrink M, Pejler G, Thomsen AR. Delayed contraction of the CD8+ T cell response toward lymphocytic choriomeningitis virus infection in mice lacking serglycin. THE JOURNAL OF IMMUNOLOGY 2008; 181:1043-51. [PMID: 18606656 DOI: 10.4049/jimmunol.181.2.1043] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
We previously reported that the lack of serglycin proteoglycan affects secretory granule morphology and granzyme B (GrB) storage in in vitro generated CTLs. In this study, the role of serglycin during viral infection was studied by infecting wild-type (wt) mice and serglycin-deficient (SG(-/-)) mice with lymphocytic choriomeningitis virus (LCMV). Wt and SG(-/-) mice cleared 10(3) PFU of highly invasive LCMV with the same kinetics, and the CD8(+) T lymphocytes from wt and SG(-/-) animals did not differ in GrB, perforin, IFN-gamma, or TNF-alpha content. However, when a less invasive LCMV strain was used, SG(-/-) GrB(+) CD8(+) T cells contained approximately 30% less GrB than wt GrB(+) CD8(+) T cells. Interestingly, the contraction of the antiviral CD8(+) T cell response to highly invasive LCMV was markedly delayed in SG(-/-) mice, and a delayed contraction of the virus-specific CD8(+) T cell response was also seen after infection with vesicular stomatitis virus. BrdU labeling of cells in vivo revealed that the delayed contraction was associated with sustained proliferation of Ag-specific CD8(+) T cells in SG(-/-) mice. Moreover, wt LCMV-specific CD8(+) T cells from TCR318 transgenic mice expanded much more extensively in virus-infected SG(-/-) mice than in matched wt mice, indicating that the delayed contraction represents a T cell extrinsic phenomenon. In summary, the present report points to a novel, previously unrecognized role for serglycin proteoglycan in regulating the kinetics of antiviral CD8(+) T cell responses.
Collapse
Affiliation(s)
- Mirjana Grujic
- University of Copenhagen, Institute of International Health, Immunology and Microbiology, Copenhagen, Denmark
| | | | | | | | | | | |
Collapse
|
31
|
Noser JA, Mael AA, Sakuma R, Ohmine S, Marcato P, Lee PW, Ikeda Y. The RAS/Raf1/MEK/ERK Signaling Pathway Facilitates VSV-mediated Oncolysis: Implication for the Defective Interferon Response in Cancer Cells. Mol Ther 2007; 15:1531-6. [PMID: 17505473 DOI: 10.1038/sj.mt.6300193] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Vesicular stomatitis virus (VSV) can replicate in malignant cells more efficiently than in normal cells. Although the selective replication appears to be caused by defects in the interferon (IFN) system in malignant cells, the mechanisms which render these cells less responsive to IFN remain poorly understood. Here we present evidence that an activated RAS/Raf1/MEK/ERK pathway plays a critical role in the defects. NIH 3T3 or human primary cells stably expressing active RAS or Raf1 were rapidly killed by VSV. Although IFNalpha treatment no longer protected the RAS- or Raf1-overexpressing cells from VSV infection, responsiveness to IFNalpha was restored following treatment with the mitogen-activated protein kinase kinase (MEK) inhibitor U0126. Similarly, human cancer-derived cell lines became more responsive to IFNalpha in conjunction with U0126 treatment. Intriguingly, dual treatment with both IFNalpha and U0126 severely reduced the levels of viral RNAs in the infected cells. Moreover, cancer cells showed defects in inducing an IFNalpha-responsive factor, MxA, which is known to block VSV RNA synthesis, and U0126 restored the MxA expression. Our observations suggest that activation of the extracellular signal-regulated protein kinase (ERK) signaling leads to the defect in IFNalpha-mediated upregulation of MxA protein, which facilitates VSV oncolysis. In view of the fact that 30% of all cancers have constitutive activation of the RAS/Raf1/MEK/ERK pathway, VSV would be an ideal oncolytic virus for targeting such cancers.
Collapse
Affiliation(s)
- Josh A Noser
- Molecular Medicine Program, Mayo Clinic College of Medicine, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Ostrowski M, Vermeulen M, Zabal O, Zamorano PI, Sadir AM, Geffner JR, Lopez OJ. The early protective thymus-independent antibody response to foot-and-mouth disease virus is mediated by splenic CD9+ B lymphocytes. J Virol 2007; 81:9357-67. [PMID: 17567692 PMCID: PMC1951431 DOI: 10.1128/jvi.00677-07] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Infection of mice with cytopathic foot-and-mouth disease virus (FMDV) induces a rapid and specific thymus-independent (TI) neutralizing antibody response that promptly clears the virus. Herein, it is shown that FMDV-infected dendritic cells (DCs) directly stimulate splenic innate-like CD9(+) B lymphocytes to rapidly (3 days) produce neutralizing anti-FMDV immunoglobulin M antibodies without T-lymphocyte collaboration. In contrast, neither follicular (CD9(-)) B lymphocytes from the spleen nor B lymphocytes from lymph nodes efficiently respond to stimulation with FMDV-infected DCs. The production of these protective neutralizing antibodies is dependent on DC-derived interleukin-6 (IL-6) and on CD9(+) cell-derived IL-10 secretion. In comparison, DCs loaded with UV-inactivated FMDV are significantly less efficient in directly stimulating B lymphocytes to secrete TI antibodies. A critical role of the spleen in the early production of anti-FMDV antibodies in infected mice was also demonstrated in vivo. Indeed, either splenectomy or functional disruption of the marginal zone of the spleen delays and reduces the magnitude of the TI anti-FMDV antibody response in infected mice. Together, these results indicate that in addition to virus localization, the FMDV-mediated modulation of DC functionality is a key parameter that collaborates in the induction of a rapid and protective TI antibody response against this virus.
Collapse
Affiliation(s)
- Matias Ostrowski
- Instituto de Virologia, Centro de Investigaciones en Ciencias Veterinarias, INTA-Castelar, Buenos Aires, Argentina
| | | | | | | | | | | | | |
Collapse
|
33
|
Bach P, Kamphuis E, Odermatt B, Sutter G, Buchholz CJ, Kalinke U. Vesicular Stomatitis Virus Glycoprotein Displaying Retrovirus-Like Particles Induce a Type I IFN Receptor-Dependent Switch to Neutralizing IgG Antibodies. THE JOURNAL OF IMMUNOLOGY 2007; 178:5839-47. [PMID: 17442968 DOI: 10.4049/jimmunol.178.9.5839] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Vesicular stomatitis virus (VSV) infection rapidly induces IFN-alphabeta that confers initial survival, whereas long-term protection is mediated by neutralizing IgG responses. Because coadministration of IFN-alphabeta can enhance Ab responses against soluble Ags, we addressed whether virus-induced IFN-alphabeta also had an impact on the induction of neutralizing Ab responses. To this end, we generated apathogenic retrovirus-like particles (VLP) displaying the VSV gp (VLP-VSV). Reminiscent of live VSV, VLP-VSV induced VSV-neutralizing IgM responses that switched to IgG in a T help-dependent manner. In type I IFN receptor-deficient (IFNAR(-/-)) mice, VLP-VSV injection elicited neutralizing IgM, whereas the IgG switch was absent. The lack of subclass switch was associated with a reduced germinal center reaction. Conditional knockout mice with a lymphocyte-specific IFNAR ablation showed normal Ab responses against VLP-VSV, as well as against live VSV. Thus, IFNAR triggering critically promoted the T help-dependent subclass switch of virus-neutralizing Ab responses against VLP-VSV. Interestingly, in the context of VLP-VSV as well as VSV immunization, IFNAR triggering of B lymphocytes did not play a critical role.
Collapse
Affiliation(s)
- Patricia Bach
- Division of Immunology, Paul-Ehrlich-Institut, Langen, Germany
| | | | | | | | | | | |
Collapse
|
34
|
Bartholdy C, Kauffmann SØ, Christensen JP, Thomsen AR. Agonistic anti-CD40 antibody profoundly suppresses the immune response to infection with lymphocytic choriomeningitis virus. THE JOURNAL OF IMMUNOLOGY 2007; 178:1662-70. [PMID: 17237416 DOI: 10.4049/jimmunol.178.3.1662] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Previous work has shown that agonistic Abs to CD40 (anti-CD40) can boost weak CD8 T cell responses as well as substitute for CD4 T cell function during chronic gammaherpes virus infection. Agonistic anti-CD40 treatment has, therefore, been suggested as a potential therapeutic strategy in immunocompromised patients. In this study, we investigated whether agonistic anti-CD40 could substitute for CD4 T cell help in generating a sustained CD8 T cell response and prevent viral recrudescence following infection with lymphocytic choriomeningitis virus (LCMV). Contrary to expectations, we found that anti-CD40 treatment of MHC class II-deficient mice infected with a moderate dose of LCMV resulted in severe suppression of the antiviral CD8 T cell response and uncontrolled virus spread, rather than improved CD8 T cell immune surveillance. In Ab-treated wild-type mice, the antiviral CD8 T cell response also collapsed prematurely, and virus clearance was delayed. Additional analysis revealed that, following anti-CD40 treatment, the virus-specific CD8 T cells initially proliferated normally, but an increased cell loss compared with that in untreated mice was observed. The anti-CD40-induced abortion of virus-specific CD8 T cells during LCMV infection was IL-12 independent, but depended partly on Fas expression. Notably, similar anti-CD40 treatment of vesicular stomatitis virus-infected mice resulted in an improved antiviral CD8 T cell response, demonstrating that the effect of anti-CD40 treatment varies with the virus infection studied. For this reason, we recommend further evaluation of the safety of this regimen before being applied to human patients.
Collapse
Affiliation(s)
- Christina Bartholdy
- Institute of Medical Microbiology, University of Copenhagen, Copenhagen, Denmark
| | | | | | | |
Collapse
|
35
|
Vähä-Koskela MJ, Heikkilä JE, Hinkkanen AE. Oncolytic viruses in cancer therapy. Cancer Lett 2007; 254:178-216. [PMID: 17383089 PMCID: PMC7126325 DOI: 10.1016/j.canlet.2007.02.002] [Citation(s) in RCA: 220] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2006] [Revised: 02/01/2007] [Accepted: 02/05/2007] [Indexed: 12/26/2022]
Abstract
Oncolytic virotherapy is a promising form of gene therapy for cancer, employing nature’s own agents to find and destroy malignant cells. The purpose of this review is to provide an introduction to this very topical field of research and to point out some of the current observations, insights and ideas circulating in the literature. We have strived to acknowledge as many different oncolytic viruses as possible to give a broader picture of targeting cancer using viruses. Some of the newest additions to the panel of oncolytic viruses include the avian adenovirus, foamy virus, myxoma virus, yaba-like disease virus, echovirus type 1, bovine herpesvirus 4, Saimiri virus, feline panleukopenia virus, Sendai virus and the non-human coronaviruses. Although promising, virotherapy still faces many obstacles that need to be addressed, including the emergence of virus-resistant tumor cells.
Collapse
Affiliation(s)
- Markus J.V. Vähä-Koskela
- Åbo Akademi University, Department of Biochemistry and Pharmacy and Turku Immunology Centre, Turku, Finland
- Turku Graduate School of Biomedical Sciences, Turku, Finland
- Corresponding author. Address: Åbo Akademi University, Department of Biochemistry and Pharmacy and Turku Immunology Centre, Turku, Finland. Tel.: +358 2 215 4018; fax: +358 2 215 4745.
| | - Jari E. Heikkilä
- Åbo Akademi University, Department of Biochemistry and Pharmacy and Turku Immunology Centre, Turku, Finland
| | - Ari E. Hinkkanen
- Åbo Akademi University, Department of Biochemistry and Pharmacy and Turku Immunology Centre, Turku, Finland
| |
Collapse
|
36
|
Labuda T, Christensen JP, Rasmussen S, Bonnesen B, Karin M, Thomsen AR, Odum N. MEK kinase 1 is a negative regulator of virus-specific CD8(+) T cells. Eur J Immunol 2006; 36:2076-84. [PMID: 16761309 DOI: 10.1002/eji.200535163] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
MEK kinase 1 (MEKK1) is a potent JNK-activating kinase, a regulator of T helper cell differentiation, cytokine production and proliferation in vitro. Using mice deficient for MEKK1 activity (Mekk1(DeltaKD)) exclusively in their hematopoietic system, we show that MEKK1 has a negative regulatory role in the generation of a virus-specific immune response. Mekk1(DeltaKD) mice challenged with vesicular stomatitis virus (VSV) showed a fourfold increase in splenic CD8(+) T cell numbers. In contrast, the number of splenic T cells in infected WT mice was only marginally increased. The CD8(+) T cell expansion in Mekk1(DeltaKD) mice following VSV infection is virus-specific and the frequency of virus-specific T cells is significantly higher (more than threefold) in Mekk1(DeltaKD) as compared to WT animals. Moreover, the hyper-expansion of T cells seen in Mekk1(DeltaKD) mice after VSV infection is a result of increased proliferation, since a significantly higher percentage of virus-specific Mekk1(DeltaKD) CD8(+) T cells incorporated BrdU as compared to virus-specific WT CD8(+) T cells. In contrast, similar levels of apoptosis were detected in Mekk1(DeltaKD) and WT T cells following VSV infection. These results strongly suggest that MEKK1 plays a negative regulatory role in the expansion of virus-specific CD8(+) T cells in vivo.
Collapse
Affiliation(s)
- Tord Labuda
- Department of Medical Microbiology and Immunology, University of Copenhagen, Copenhagen, Denmark.
| | | | | | | | | | | | | |
Collapse
|
37
|
Ciavarra RP, Stephens A, Nagy S, Sekellick M, Steel C. Evaluation of immunological paradigms in a virus model: are dendritic cells critical for antiviral immunity and viral clearance? THE JOURNAL OF IMMUNOLOGY 2006; 177:492-500. [PMID: 16785546 DOI: 10.4049/jimmunol.177.1.492] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We have examined the role of dendritic cells (DCs) in the antiviral immune response and viral clearance using a transgenic mouse model (CD11c-diphtheria toxin (DT) receptor GFP) that allows for their conditional ablation in vivo. DT administration systemically ablated conventional and IFN-producing plasmacytoid DCs (pDCs) in transgenic, but not nontransgenic littermates, without elimination of splenic macrophages. Unexpectedly, early (12 and 48 h postinfection) viral clearance of vesicular stomatitis virus was normal in DC-depleted mice despite markedly reduced serum titers of type I IFN. DC-depleted mice remained virus-free with the exception of a subset (approximately 30%) that developed overwhelming and fatal brain infections 6 days postinfection. However, DT treatment profoundly inhibited clonal expansion of naive CD8+ vesicular stomatitis virus-specific T cells without altering the primary Th1 and Th2 cytokine response. Optimal clonal expansion required pDCs because selective elimination of these cells in vivo with a depleting Ab also suppressed expansion of tetramer+ cells, although Th1/Th2 cytokine production remained unaltered. Collectively, these data indicate that conventional DCs and to a lesser extent pDCs are critical for proliferation of naive antiviral T cells. However, other components of the primary adaptive immune response (Th1/Th2 cytokines) are essentially normal in the absence of DCs, which may account for the efficient viral clearance seen in DC-depleted mice. Thus, sufficient redundancy exists in the immune system to sustain efficient viral clearance despite loss of an APC considered essential for induction of a primary antiviral immune response.
Collapse
Affiliation(s)
- Richard P Ciavarra
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, 700 Olney Road, Norfolk, VA 23501, USA.
| | | | | | | | | |
Collapse
|
38
|
Hangartner L, Zinkernagel RM, Hengartner H. Antiviral antibody responses: the two extremes of a wide spectrum. Nat Rev Immunol 2006; 6:231-43. [PMID: 16498452 DOI: 10.1038/nri1783] [Citation(s) in RCA: 254] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Viruses elicit a diverse spectrum of antiviral antibody responses. In this review, we discuss two widely used experimental model systems for viral infections - non-cytopathic lymphocytic choriomeningitis virus (LCMV) and acutely cytopathic vesicular stomatitis virus (VSV) - to analyse two fundamentally different types of antiviral antibody response. The basic principles found in these model infections are discussed in the context of other viral infections, and with regard to protective neutralizing versus non-protective enzyme-linked immunosorbent assay (ELISA)-detected antibody responses. Issues of antibody specificity, affinity and avidity, maturation and escape are discussed in the context of co-evolution of the host and viruses.
Collapse
Affiliation(s)
- Lars Hangartner
- Institute of Experimental Immunology, University Hospital Zurich, Schmelzbergstrasse 12, 8091 Zürich, Switzerland
| | | | | |
Collapse
|
39
|
Yager E, Bitsaktsis C, Nandi B, McBride JW, Winslow G. Essential role for humoral immunity during Ehrlichia infection in immunocompetent mice. Infect Immun 2006; 73:8009-16. [PMID: 16299294 PMCID: PMC1307037 DOI: 10.1128/iai.73.12.8009-8016.2005] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Although cellular immunity is essential for host defense during intracellular bacterial infections, humoral immunity can also play a significant role in host defense during infection by some intracellular bacteria, including the ehrlichiae. Antibodies can protect susceptible SCID mice from fatal Ehrlichia chaffeensis infection, an observation that has been hypothesized to involve the opsonization of bacteria released from host cells. To determine whether humoral immunity plays an essential role during ehrlichia infection in immunocompetent mice, we utilized a murine model of fatal monocytotropic ehrlichiosis caused by Ixodes ovatus ehrlichia. Mice lacking either B cells or FcgammaRI were unable to resolve a low-dose (sublethal) I. ovatus ehrlichia infection, which suggested that humoral immunity is essential for resistance. Polyclonal sera generated in I. ovatus ehrlichia-infected mice recognized a conserved ehrlichia outer membrane protein and, when administered to infected mice, caused a significant decrease in bacterial infection. Mice experimentally depleted of complement, or deficient for complement receptors 1 and 2, were also susceptible to sublethal I. ovatus ehrlichia infection, as were mice that lacked the phox91 subunit of NADPH oxidase. The data are consistent with a mechanism whereby bacteria released from infected cells are lysed directly by complement or undergo antibody-mediated FcgammaR-dependent phagocytosis and subsequent exposure to reactive oxygen intermediates. The findings suggest mechanisms whereby antibodies contribute to immunity against intracellular bacteria in immunocompetent mice.
Collapse
Affiliation(s)
- Eric Yager
- The Department of Biomedical Sciences, School of Public Health, University at Albany, Albany, New York 12201-0509, USA
| | | | | | | | | |
Collapse
|
40
|
Abstract
The emergence of vesicular stomatatis virus (VSV) as a potent antitumor agent has made a dissection of the molecular determinants of host-cell permissiveness to this virus an important objective. Such insight would not only enable the intelligent design of future generations of recombinant VSV vectors to combat disease, but may also resolve general features of cellular transformation that may be exploited by this virus, and perhaps other oncolytic viruses. The defective pathways underlining the oncolytic activity of VSV remain to be fully determined but recent data indicates that flaws in innate immune responses, involving the interferon (IFN) system, may commonly occur in tumor cells and thus play a large role in facilitating oncolysis. Aside from the IFN system, however, it is almost certain that other key cellular pathways may be similarly defective and therefore cooperatively contribute towards mediating rapid oncolytic virus activity. Recent data have indicated that defects in cancer cell translational regulation could be one area that may be exploited by VSV. Certainly, all viruses require cellular protein synthesis pathways to facilitate their replication and many have devised numerous mechanisms to ensure that viral mRNAs become translated at the expense of the host. Using VSV as a model, this review will discuss some of the recent developments in the fields of innate immunity and translational regulation that may help explain mechanisms of viral oncolysis.
Collapse
Affiliation(s)
- Glen N Barber
- Department of Microbiology and Immunology, Sylvester Comprehensive Cancer Center, University of Miami School of Medicine, FL 33136, USA.
| |
Collapse
|
41
|
Rivera J, Zaragoza O, Casadevall A. Antibody-mediated protection against Cryptococcus neoformans pulmonary infection is dependent on B cells. Infect Immun 2005; 73:1141-50. [PMID: 15664957 PMCID: PMC546959 DOI: 10.1128/iai.73.2.1141-1150.2005] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The pathogenesis of pulmonary Cryptococcus neoformans infection and the efficacy of passive immunoglobulin G1 (IgG1) administration were investigated in B-cell-deficient and C57BL/6J mice. C57BL/6J mice lived longer than B-cell-deficient mice after both intratracheal and intravenous infections. Administration of IgG1 prior to infection prolonged the survival of C57BL/6J mice but had no effect on the survival or numbers of CFU in the lungs of B-cell-deficient mice. C. neoformans infection in B-cell-deficient mice resulted in significantly higher levels of gamma interferon (IFN-gamma), monocyte chemoattractant protein-1 (MCP-1), and macrophage inflammatory protein-1alpha (MIP-1alpha) than in C57BL/6J mice. IgG1 administration reduced IFN-gamma and MCP-1 levels in C57BL/6J mice but not in B-cell-deficient mice. In addition, compared to its effect in C57BL/6J mice, C. neoformans infection in FcRgammaIII-deficient, athymic, and SCID mice significantly increased IFN-gamma and MCP-1 levels. IgG1 administration was associated with reduced IFN-gamma levels in C57BL/6J mice but not in FcRgammaIII-deficient, athymic, and SCID mice. These observations suggest that IgG1-mediated protection in this system is a consequence of alterations in the inflammatory response that translate into less damage to the host without directly reducing the fungal burden. For hosts with impaired immunities, the ineffectiveness of passive antibody (Ab) may reflect an inability to down-regulate inflammation and avoid self-damage. The results indicate an important role for B cells in host defense against C. neoformans infection and demonstrate a surprising dependence of Ab-mediated protection on B cells in this system.
Collapse
Affiliation(s)
- Johanna Rivera
- Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | | | | |
Collapse
|
42
|
Abstract
Recent data has shown that viruses such as vesicular stomatitis virus (VSV), a relatively non-pathogenic, negative-stranded RNA virus, can preferentially replicate in malignant cells and less so in normal cells. VSV appears able to carry out this function in transformed cells since these hosts exhibit the hallmarks of flawed host defense, probably involving the interferon system, which is essential for preventing virus replication. The simple genetic constitution of VSV, lack of any known transforming, integrating or reassortment properties, extensive knowledge relating to its interaction with the immune system and the ability to genetically manipulate this agent affords an ideal opportunity to exploit the oncolytic and gene targeting potential of this innocuous virus. Thus, aside from preferentially targeting malignant cells VSV recombinants could be generated that could increase a tumor's susceptibility to chemotherapeutic agents and/ or importantly, the host immune response. Collectively, our data and others demonstrate that VSV as well as other RNA viruses could provide a promising and exciting approach to cancer therapy.
Collapse
Affiliation(s)
- Glen N Barber
- Department of Microbiology and Immunology, Sylvester Comprehensive Cancer Center, University of Miami School of Medicine, Miami, Florida 33136, USA.
| |
Collapse
|
43
|
Recher M, Hunziker L, Ciurea A, Harris N, Lang KS. Public, private and non-specific antibodies induced by non-cytopathic viral infections. Curr Opin Microbiol 2004; 7:426-33. [PMID: 15358263 DOI: 10.1016/j.mib.2004.06.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Lymphocytic choriomeningitis virus (LCMV) represents a useful experimental model of murine infection with a non-cytopathic virus, bearing resemblance to HIV and hepatitis C virus (HCV) infections in humans. Recent data from the LCMV model indicate that the humoral immune response that is induced by non-cytopathic viruses is far more complex than previously appreciated. LCMV-induced IgG production is largely polyclonal, with more than 90% of the antibody repertoire constituting non-relevant specificities. A delayed virus-neutralizing antibody response is induced, including specificities directed not only against the parental LCMV-strain present in the host but also cross-specifically against LCMV-variants isolated from other hosts. These findings provide novel insights to aid our understanding of clinically relevant observations that are recorded following human infection with HIV, HCV and dengue viruses.
Collapse
Affiliation(s)
- Mike Recher
- Institute for Experimental Immunology, University Hospital, Schmelzbergstrasse 12, CH-8091 Zürich, Switzerland.
| | | | | | | | | |
Collapse
|
44
|
Recher M, Lang KS, Hunziker L, Freigang S, Eschli B, Harris NL, Navarini A, Senn BM, Fink K, Lötscher M, Hangartner L, Zellweger R, Hersberger M, Theocharides A, Hengartner H, Zinkernagel RM. Deliberate removal of T cell help improves virus-neutralizing antibody production. Nat Immunol 2004; 5:934-42. [PMID: 15300247 DOI: 10.1038/ni1102] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2004] [Accepted: 07/06/2004] [Indexed: 12/15/2022]
Abstract
The B cell response to lymphocytic choriomeningitis virus is characterized by a CD4(+) T cell-dependent polyclonal hypergammaglobulinemia and delayed formation of virus-specific neutralizing antibodies. Here we provide evidence that, paradoxically, because of polyclonal B cell activation, virus-specific T cell help impairs the induction of neutralizing antibody responses. Experimental reduction in CD4(+) T cell help in vivo resulted in potent neutralizing antibody responses without impairment of CD8(+) T cell activity. These unexpected consequences of polyclonal B cell activation may affect vaccine strategies and the treatment of clinically relevant chronic bacterial, parasitic and viral infections in which hypergammaglobulinemia is regularly found.
Collapse
Affiliation(s)
- Mike Recher
- Institute for Experimental Immunology, University Hospital Zürich, Schmelzbergstrasse 12, CH-8091 Zürich, Switzerland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Kristensen NN, Madsen AN, Thomsen AR, Christensen JP. Cytokine production by virus-specific CD8(+) T cells varies with activation state and localization, but not with TCR avidity. J Gen Virol 2004; 85:1703-1712. [PMID: 15166455 DOI: 10.1099/vir.0.79903-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The ability of virus-specific CD8(+) T cells to produce cytokines was studied in mice infected with lymphocytic choriomeningitis virus and vesicular stomatitis virus. Intracellular staining was used to visualize cytokine-producing CD8(+) and CD4(+) T cells. Overall, virus-specific CD8(+) T cells produce a similar range of cytokines (IFN-gamma, TNF-alpha, IL-2, GM-CSF, RANTES, MIP-1alpha and MIP-1beta) as CD4(+) T cells, but the relative distribution of cytokine-producing subsets is different. Moreover, cytokine-producing CD8(+) T cells were found to dominate numerically at all time-points tested. Co-staining for more than one cytokine revealed that while all cytokine-producing CD8(+) T cells synthesized IFN-gamma, additional cytokines were produced by partly overlapping subsets of this population. The frequency of cells producing more than one cytokine was higher in a tertiary site (peritoneum) and generally increased with transition into the memory phase; however, GM-CSF producing cells were only present transiently. Concerning factors predicted to influence the distribution of cytokine-producing subsets, IFN-gamma and IL-12 did not play a role, nor was extensive virus replication essential. Notably, regarding the heterogeneity in cytokine production by individual cells with similar epitope specificity, variation in TCR avidity was not the cause, since in vivo-activated TCR transgene-expressing cells were as heterogeneous in cytokine expression as polyclonal cells specific for the same epitope.
Collapse
Affiliation(s)
- Nanna Ny Kristensen
- Institute of Medical Microbiology and Immunology, University of Copenhagen, Copenhagen, Denmark
| | - Andreas Nygaard Madsen
- Institute of Medical Microbiology and Immunology, University of Copenhagen, Copenhagen, Denmark
| | - Allan Randrup Thomsen
- Institute of Medical Microbiology and Immunology, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
46
|
Gupta M, Mahanty S, Greer P, Towner JS, Shieh WJ, Zaki SR, Ahmed R, Rollin PE. Persistent infection with ebola virus under conditions of partial immunity. J Virol 2004; 78:958-67. [PMID: 14694127 PMCID: PMC368745 DOI: 10.1128/jvi.78.2.958-967.2004] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Ebola hemorrhagic fever in humans is associated with high mortality; however, some infected hosts clear the virus and recover. The mechanisms by which this occurs and the correlates of protective immunity are not well defined. Using a mouse model, we determined the role of the immune system in clearance of and protection against Ebola virus. All CD8 T-cell-deficient mice succumbed to subcutaneous infection and had high viral antigen titers in tissues, whereas mice deficient in B cells or CD4 T cells cleared infection and survived, suggesting that CD8 T cells, independent of CD4 T cells and antibodies, are critical to protection against subcutaneous Ebola virus infection. B-cell-deficient mice that survived the primary subcutaneous infection (vaccinated mice) transiently depleted or not depleted of CD4 T cells also survived lethal intraperitoneal rechallenge for >/==" BORDER="0">25 days. However, all vaccinated B-cell-deficient mice depleted of CD8 T cells had high viral antigen titers in tissues following intraperitoneal rechallenge and died within 6 days, suggesting that memory CD8 T cells by themselves can protect mice from early death. Surprisingly, vaccinated B-cell-deficient mice, after initially clearing the infection, were found to have viral antigens in tissues later (day 120 to 150 post-intraperitoneal infection). Furthermore, following intraperitoneal rechallenge, vaccinated B-cell-deficient mice that were transiently depleted of CD4 T cells had high levels of viral antigen in tissues earlier (days 50 to 70) than vaccinated undepleted mice. This demonstrates that under certain immunodeficiency conditions, Ebola virus can persist and that loss of primed CD4 T cells accelerates the course of persistent infections. These data show that CD8 T cells play an important role in protection against acute disease, while both CD4 T cells and antibodies are required for long-term protection, and they provide evidence of persistent infection by Ebola virus suggesting that under certain conditions of immunodeficiency a host can harbor virus for prolonged periods, potentially acting as a reservoir.
Collapse
Affiliation(s)
- Manisha Gupta
- Special Pathogens Branch, Division of Viral and Rickettsial Diseases, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia 30333, USA.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Andreasen SØ, Thomsen AR, Koteliansky VE, Novobrantseva TI, Sprague AG, de Fougerolles AR, Christensen JP. Expression and functional importance of collagen-binding integrins, alpha 1 beta 1 and alpha 2 beta 1, on virus-activated T cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 171:2804-11. [PMID: 12960301 DOI: 10.4049/jimmunol.171.6.2804] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Adhesive interactions are crucial to cell migration into inflammatory sites. Using murine lymphocytic choriomeningitis virus as an Ag model system, we have investigated expression and function of collagen-binding integrins, alpha(1)beta(1) and alpha(2)beta(1), on activated and memory T cells. Using this system and MHC tetramers to define Ag-specific T cells, we demonstrate that contrary to being VLAs, expression of alpha(1)beta(1) and alpha(2)beta(1) can be rapidly induced on acutely activated T cells, that expression of alpha(1)beta(1) remains elevated on memory T cells, and that expression of alpha(1)beta(1) parallels that of viral-specific effector CD8(+) T cells (defined by tetramer and IFN-gamma staining). In an adoptive transfer model, mAb-mediated blockade of these integrins on activated effector and memory T cells inhibited Ag-specific delayed-type hypersensitivity responses; similar decreased responses were seen upon transfer of alpha(1)-deficient activated/memory T cells. Thus, expression of alpha(1)beta(1) and alpha(2)beta(1) integrins on activated T cells is directly functionally important for generation of inflammatory responses within tissues. Finally, the inhibitory effect of alpha(1)beta(1) blockade on the delayed-type hypersensitivity response could be bypassed by direct injection of Ag-specific T cells to inflammatory sites, demonstrating for the first time in vivo that collagen-binding integrins are involved in leukocyte migration into tissues.
Collapse
Affiliation(s)
- Susanne Ø Andreasen
- Institute of Medical Microbiology and Immunology, Panum Institute, Copenhagen, Denmark. Biogen, Cambridge, MA 02142, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Obuchi M, Fernandez M, Barber GN. Development of recombinant vesicular stomatitis viruses that exploit defects in host defense to augment specific oncolytic activity. J Virol 2003; 77:8843-56. [PMID: 12885903 PMCID: PMC167243 DOI: 10.1128/jvi.77.16.8843-8856.2003] [Citation(s) in RCA: 164] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2003] [Accepted: 05/28/2003] [Indexed: 12/11/2022] Open
Abstract
Vesicular stomatitis virus (VSV) is a negative-stranded RNA virus normally sensitive to the antiviral actions of alpha/beta interferon (IFN-alpha/beta). Recently, we reported that VSV replicates to high levels in many transformed cells due, in part, to susceptible cells harboring defects in the IFN system. These observations were exploited to demonstrate that VSV can be used as a viral oncolytic agent to eradicate malignant cells in vivo while leaving normal tissue relatively unaffected. To attempt to improve the specificity and efficacy of this system as a potential tool in gene therapy and against malignant disease, we have genetically engineered VSV that expresses the murine IFN-beta gene. The resultant virus (VSV-IFNbeta) was successfully propagated in cells not receptive to murine IFN-alpha/beta and expressed high levels of functional heterologous IFN-beta. In normal murine embryonic fibroblasts (MEFs), the growth of VSV-IFNbeta was greatly reduced and diminished cytopathic effect was observed due to the production of recombinant IFN-beta, which by functioning in a manner involving autocrine and paracrine mechanisms induced an antiviral effect, preventing virus growth. However, VSV-IFNbeta grew to high levels and induced the rapid apoptosis of transformed cells due to defective IFN pathways being prevalent and thus unable to initiate proficient IFN-mediated host defense. Importantly, VSV expressing the human IFN-beta gene (VSV-hIFNbeta) behaved comparably and, while nonlytic to normal human cells, readily killed their malignant counterparts. Similar to our in vitro observations, following intravenous and intranasal inoculation in mice, recombinant VSV (rVSV)-IFNbeta was also significantly attenuated compared to wild-type VSV or rVSV expressing green fluorescent protein. However, VSV-IFNbeta retained propitious oncolytic activity against metastatic lung disease in immunocompetent animals and was able to generate robust antitumor T-cell responses. Our data indicate that rVSV designed to exploit defects in mechanisms of host defense can provide the basis for new generations of effective, specific, and safer viral vectors for the treatment of malignant and other disease.
Collapse
Affiliation(s)
- Masatsugu Obuchi
- Department of Microbiology and Immunology and Sylvester Comprehensive Cancer Center, University of Miami School of Medicine, Miami, Florida 33136, USA
| | | | | |
Collapse
|
49
|
Henderson DK. Managing occupational risks for hepatitis C transmission in the health care setting. Clin Microbiol Rev 2003; 16:546-68. [PMID: 12857782 PMCID: PMC164218 DOI: 10.1128/cmr.16.3.546-568.2003] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Hepatitis C virus (HCV) infection is a significant contemporary health problem in the United States and elsewhere. Because it is primarily transmitted via blood, hepatitis C infection presents risks for both nosocomial transmission to patients and occupational spread to health care workers. Recent insights into the pathogenesis, immunopathogenesis, natural history, and treatment of infection caused by this unique flavivirus provide a rationale for the use of new strategies for managing occupational hepatitis C infections when they occur. This article reviews this developing information. Recently published data demonstrate success rates in the treatment of "acute hepatitis C syndrome" that approach 100\%, and although these studies are not directly applicable to all occupational infections, they may provide important clues to optimal management strategies. In addition, the article delineates approaches to the prevention of occupational exposures and also addresses the difficult issue of managing HCV-infected health care providers. The article summarizes currently available data about the nosocomial epidemiology of HCV infection and the magnitude of risk and discusses several alternatives for managing exposure and infection. No evidence supports the use of immediate postexposure prophylaxis with immunoglobulin, immunomodulators, or antiviral agents. Based on the very limited data available, the watchful waiting and preemptive therapy strategies described in detail in this article represent reasonable interim approaches to the complex problem of managing occupational HCV infections, at least until more definitive data are obtained.
Collapse
Affiliation(s)
- David K Henderson
- Warren G. Magnuson Clinical Center, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland 20892, USA.
| |
Collapse
|
50
|
Oehen S, Odermatt B, Karrer U, Hengartner H, Zinkernagel R, López-Macías C. Marginal zone macrophages and immune responses against viruses. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:1453-8. [PMID: 12133971 DOI: 10.4049/jimmunol.169.3.1453] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The effective establishment of antiviral protection requires a coordinated interplay between the innate and adaptive immune system. Using osteopetrotic (op(-/-)) mice, this study investigated the influence of marginal zone macrophages in controlling and initiating a protective immune response against a cytopathic vs a non- or low-cytopathic virus. Despite the generation of potent adaptive immune responses, antiviral protection against cytopathic vesicular stomatitis virus critically depended on the presence of marginal zone macrophages. Infection with low doses (100 PFU) of non- or low-cytopathic lymphocytic choriomeningitis virus was rarely cleared and usually resulted in a carrier state in the majority of mice. This shows that the early innate immune system provides an important preparatory phase to the adaptive immune system and is particularly important for antiviral protection.
Collapse
Affiliation(s)
- Stephan Oehen
- Institute for Experimental Immunology, University Hospital, Zurich, Switzerland.
| | | | | | | | | | | |
Collapse
|