1
|
Fares S, Krishna BA. Why Are Cytomegalovirus-Encoded G-Protein-Coupled Receptors Essential for Infection but Only Variably Conserved? Pathogens 2025; 14:245. [PMID: 40137730 PMCID: PMC11945030 DOI: 10.3390/pathogens14030245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/25/2025] [Accepted: 03/03/2025] [Indexed: 03/29/2025] Open
Abstract
Cytomegaloviruses (CMVs) encode viral G-protein-coupled receptors (vGPCRs) that have diverged from their cellular homologues to perform new functions. Human cytomegalovirus (HCMV) encodes four vGPCRs: UL33, UL78, US27, and US28, which contribute to viral pathogenesis, cellular signalling, and latency. While the role of US28 in chemokine signalling and viral latency is well characterised, the functions of other vGPCRs remain incompletely understood. Rodent cytomegaloviruses only have homologues to UL33 and UL78, while primates have two to five additional GPCRs which are homologues of US27 and US28. Different CMVs appear to have evolved vGPCRs with functions specific to infection of their respective host. As non-human CMVs are used as model organisms to understand clinical cytomegalovirus disease and develop vaccines and antivirals, understanding the differences between these vGPCRs helps researchers understand critical differences between their models. This review aims to address the differences between CMV vGPCRs, and how these differences may affect models of CMV disease to facilitate future research.
Collapse
Affiliation(s)
- Suzan Fares
- Occlutech Holding AG, Feldstrasse 22, 8200 Schaffhausen, Switzerland;
| | - Benjamin A. Krishna
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Cambridge CB2 0AW, UK
- Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| |
Collapse
|
2
|
Otero CE, Petkova S, Ebermann M, Taher H, John N, Hoffmann K, Davalos A, Moström MJ, Gilbride RM, Papen CR, Barber-Axthelm A, Scheef EA, Barfield R, Sprehe LM, Kendall S, Manuel TD, Beechwood T, Nguyen LK, Vande Burgt NH, Chan C, Denton M, Streblow ZJ, Streblow DN, Tarantal AF, Hansen SG, Kaur A, Permar S, Früh K, Hengel H, Malouli D, Kolb P. Rhesus Cytomegalovirus-encoded Fcγ-binding glycoproteins facilitate viral evasion from IgG-mediated humoral immunity. Nat Commun 2025; 16:1200. [PMID: 39885150 PMCID: PMC11782611 DOI: 10.1038/s41467-025-56419-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 01/16/2025] [Indexed: 02/01/2025] Open
Abstract
Human cytomegalovirus (HCMV) encodes four viral Fc-gamma receptors (vFcγRs) that counteract antibody-mediated activation in vitro, but their role in infection and pathogenesis is unknown. To examine their in vivo function in an animal model evolutionarily closely related to humans, we identified and characterized Rh05, Rh152/151 and Rh173 as the complete set of vFcγRs encoded by rhesus CMV (RhCMV). Each one of these proteins displays functional similarities to their prospective HCMV orthologs with respect to antagonizing host FcγR activation in vitro. When RhCMV-naïve male rhesus macaques were infected with vFcγR-deleted RhCMV, peak plasma DNAemia levels and anti-RhCMV antibody responses were comparable to wildtype infections of both male and female animals. However, the duration of plasma DNAemia was significantly shortened in immunocompetent, but not in CD4 + T cell-depleted animals. Since vFcγRs were not required for superinfection of rhesus macaques, we conclude that these proteins can prolong lytic replication during primary infection by evading virus-specific adaptive immune responses, particularly antibodies.
Collapse
Affiliation(s)
- Claire E Otero
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
- Department of Pathology, Duke University, Durham, North Carolina, USA
| | - Sophia Petkova
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Martin Ebermann
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Husam Taher
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Nessy John
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Katja Hoffmann
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Angel Davalos
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, USA
| | - Matilda J Moström
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Roxanne M Gilbride
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Courtney R Papen
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Aaron Barber-Axthelm
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Elizabeth A Scheef
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Richard Barfield
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, USA
| | - Lesli M Sprehe
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Savannah Kendall
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Tabitha D Manuel
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Teresa Beechwood
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Linh Khanh Nguyen
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Nathan H Vande Burgt
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Cliburn Chan
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, USA
| | - Michael Denton
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Zachary J Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Daniel N Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Alice F Tarantal
- Departments of Pediatrics and Cell Biology and Human Anatomy, School of Medicine, and California National Primate Research Center, University of California, Davis, CA, USA
| | - Scott G Hansen
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Amitinder Kaur
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Sallie Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Klaus Früh
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Hartmut Hengel
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Daniel Malouli
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA.
| | - Philipp Kolb
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
3
|
Bruton J, Hanke T. Exploitation of Unconventional CD8 T-Cell Responses Induced by Engineered Cytomegaloviruses for the Development of an HIV-1 Vaccine. Vaccines (Basel) 2025; 13:72. [PMID: 39852851 PMCID: PMC11769474 DOI: 10.3390/vaccines13010072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/08/2025] [Accepted: 01/12/2025] [Indexed: 01/26/2025] Open
Abstract
After four decades of intensive research, traditional vaccination strategies for HIV-1 remain ineffective due to HIV-1's extraordinary genetic diversity and complex immune evasion mechanisms. Cytomegaloviruses (CMV) have emerged as a novel type of vaccine vector with unique advantages due to CMV persistence and immunogenicity. Rhesus macaques vaccinated with molecular clone 68-1 of RhCMV (RhCMV68-1) engineered to express simian immunodeficiency virus (SIV) immunogens elicited an unconventional major histocompatibility complex class Ib allele E (MHC-E)-restricted CD8+ T-cell response, which consistently protected over half of the animals against a highly pathogenic SIV challenge. The RhCMV68-1.SIV-induced responses mediated a post-infection replication arrest of the challenge virus and eventually cleared it from the body. These observations in rhesus macaques opened a possibility that MHC-E-restricted CD8+ T-cells could achieve similar control of HIV-1 in humans. The potentially game-changing advantage of the human CMV (HCMV)-based vaccines is that they would induce protective CD8+ T-cells persisting at the sites of entry that would be insensitive to HIV-1 evasion. In the RhCMV68-1-protected rhesus macaques, MHC-E molecules and their peptide cargo utilise complex regulatory mechanisms and unique transport patterns, and researchers study these to guide human vaccine development. However, CMVs are highly species-adapted viruses and it is yet to be shown whether the success of RhCMV68-1 can be translated into an HCMV ortholog for humans. Despite some safety concerns regarding using HCMV as a vaccine vector in humans, there is a vision of immune programming of HCMV to induce pathogen-tailored CD8+ T-cells effective against HIV-1 and other life-threatening diseases.
Collapse
Affiliation(s)
- Joseph Bruton
- Hertford College, University of Oxford, Oxford OX1 3BW, UK;
| | - Tomáš Hanke
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK
| |
Collapse
|
4
|
Mocarski ES. Cytomegalovirus Biology Viewed Through a Cell Death Suppression Lens. Viruses 2024; 16:1820. [PMID: 39772130 PMCID: PMC11680106 DOI: 10.3390/v16121820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/22/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
Cytomegaloviruses, species-specific members of the betaherpesviruses, encode an impressive array of immune evasion strategies committed to the manipulation of the host immune system enabling these viruses to remain for life in a stand-off with host innate and adaptive immune mechanisms. Even though they are species-restricted, cytomegaloviruses are distributed across a wide range of different mammalian species in which they cause systemic infection involving many different cell types. Regulated, or programmed cell death has a recognized potential to eliminate infected cells prior to completion of viral replication and release of progeny. Cell death also naturally terminates replication during the final stages of replication. Over the past two decades, the host defense potential of known programmed cell death pathways (apoptosis, necroptosis, and pyroptosis), as well as a novel mitochondrial serine protease pathway have been defined through studies of cytomegalovirus-encoded cell death suppressors. Such virus-encoded inhibitors prevent virus-induced, cytokine-induced, and stress-induced death of infected cells while also moderating inflammation. By evading cell death and consequent inflammation as well as innate and adaptive immune clearance, cytomegaloviruses represent successful pathogens that become a critical disease threat when the host immune system is compromised. This review will discuss cell death programs acquired for mammalian host defense against cytomegaloviruses and enumerate the range of modulatory strategies this type of virus employs to balance host defense in favor of lifelong persistence.
Collapse
Affiliation(s)
- Edward S. Mocarski
- Department of Microbiology & Immunology, Stanford Medical School, Stanford University, Stanford, CA 94305, USA;
- Department of Microbiology & Immunology, Emory Medical School, Emory Vaccine Center, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
5
|
Malouli D, Tiwary M, Gilbride RM, Morrow DW, Hughes CM, Selseth A, Penney T, Castanha P, Wallace M, Yeung Y, Midgett M, Williams C, Reed J, Yu Y, Gao L, Yun G, Treaster L, Laughlin A, Lundy J, Tisoncik-Go J, Whitmore LS, Aye PP, Schiro F, Dufour JP, Papen CR, Taher H, Picker LJ, Früh K, Gale M, Maness NJ, Hansen SG, Barratt-Boyes S, Reed DS, Sacha JB. Cytomegalovirus vaccine vector-induced effector memory CD4 + T cells protect cynomolgus macaques from lethal aerosolized heterologous avian influenza challenge. Nat Commun 2024; 15:6007. [PMID: 39030218 PMCID: PMC11272155 DOI: 10.1038/s41467-024-50345-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 07/08/2024] [Indexed: 07/21/2024] Open
Abstract
An influenza vaccine approach that overcomes the problem of viral sequence diversity and provides long-lived heterosubtypic protection is urgently needed to protect against pandemic influenza viruses. Here, to determine if lung-resident effector memory T cells induced by cytomegalovirus (CMV)-vectored vaccines expressing conserved internal influenza antigens could protect against lethal influenza challenge, we immunize Mauritian cynomolgus macaques (MCM) with cynomolgus CMV (CyCMV) vaccines expressing H1N1 1918 influenza M1, NP, and PB1 antigens (CyCMV/Flu), and challenge with heterologous, aerosolized avian H5N1 influenza. All six unvaccinated MCM died by seven days post infection with acute respiratory distress, while 54.5% (6/11) CyCMV/Flu-vaccinated MCM survived. Survival correlates with the magnitude of lung-resident influenza-specific CD4 + T cells prior to challenge. These data demonstrate that CD4 + T cells targeting conserved internal influenza proteins can protect against highly pathogenic heterologous influenza challenge and support further exploration of effector memory T cell-based vaccines for universal influenza vaccine development.
Collapse
Affiliation(s)
- Daniel Malouli
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Meenakshi Tiwary
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Roxanne M Gilbride
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - David W Morrow
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Colette M Hughes
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Andrea Selseth
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Toni Penney
- Tulane National Primate Research Center, Tulane University, New Orleans, LA, USA
| | - Priscila Castanha
- Department of Infectious Diseases and Microbiology, Pittsburgh, PA, USA
| | - Megan Wallace
- Department of Infectious Diseases and Microbiology, Pittsburgh, PA, USA
| | - Yulia Yeung
- Department of Infectious Diseases and Microbiology, Pittsburgh, PA, USA
| | | | - Connor Williams
- Department of Infectious Diseases and Microbiology, Pittsburgh, PA, USA
| | - Jason Reed
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Yun Yu
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Lina Gao
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Gabin Yun
- Department of Diagnostic Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Luke Treaster
- Department of Diagnostic Radiology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | - Jennifer Tisoncik-Go
- Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA, USA
| | - Leanne S Whitmore
- Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA, USA
| | - Pyone P Aye
- Tulane National Primate Research Center, Tulane University, New Orleans, LA, USA
| | - Faith Schiro
- Tulane National Primate Research Center, Tulane University, New Orleans, LA, USA
| | - Jason P Dufour
- Tulane National Primate Research Center, Tulane University, New Orleans, LA, USA
| | - Courtney R Papen
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Husam Taher
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Louis J Picker
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Klaus Früh
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Michael Gale
- Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA, USA
- Washington National Primate Research Center, Seattle, WA, 98195, USA
| | - Nicholas J Maness
- Tulane National Primate Research Center, Tulane University, New Orleans, LA, USA
| | - Scott G Hansen
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | | | | | - Jonah B Sacha
- Oregon National Primate Research Center, Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA.
| |
Collapse
|
6
|
Riedl A, Bojková D, Tan J, Jeney Á, Larsen PK, Jeney C, Full F, Kalinke U, Ruzsics Z. Construction and Characterization of a High-Capacity Replication-Competent Murine Cytomegalovirus Vector for Gene Delivery. Vaccines (Basel) 2024; 12:791. [PMID: 39066429 PMCID: PMC11281640 DOI: 10.3390/vaccines12070791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
We investigated the basic characteristics of a new murine cytomegalovirus (MCMV) vector platform. Using BAC technology, we engineered replication-competent recombinant MCMVs with deletions of up to 26% of the wild-type genome. To this end, we targeted five gene blocks (m01-m17, m106-m109, m129-m141, m144-m158, and m159-m170). BACs featuring deletions from 18% to 26% of the wild-type genome exhibited delayed virus reconstitution, while smaller deletions (up to 16%) demonstrated reconstitution kinetics similar to those of the wild type. Utilizing an innovative methodology, we introduced large genomic DNA segments, up to 35 kbp, along with reporter genes into a newly designed vector with a potential cloning capacity of 46 kbp (Q4). Surprisingly, the insertion of diverse foreign DNAs alleviated the delayed plaque formation phenotype of Q4, and these large inserts remained stable through serial in vitro passages. With reporter-gene-expressing recombinant MCMVs, we successfully transduced not only mouse cell lines but also non-rodent mammalian cells, including those of human, monkey, bovine, and bat origin. Remarkably, even non-mammalian cell lines derived from chickens exhibited successful transduction.
Collapse
Affiliation(s)
- André Riedl
- Medical Center, Institute of Virology, University of Freiburg, 79104 Freiburg, Germany (F.F.)
- Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Denisa Bojková
- Medical Center, Institute of Virology, University of Freiburg, 79104 Freiburg, Germany (F.F.)
- Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
- Institute of Medical Virology, Goethe University Frankfurt, University Hospital, 60596 Frankfurt am Main, Germany
| | - Jiang Tan
- Medical Center, Institute of Virology, University of Freiburg, 79104 Freiburg, Germany (F.F.)
- Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Ábris Jeney
- Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Pia-Katharina Larsen
- TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture between the Hanover Medical School and the Helmholtz Centre for Infection Research, Institute for Experimental Infection Research, 30625 Hanover, Germany
| | - Csaba Jeney
- Department of Microsystems Engineering—IMTEK, University of Freiburg, 79110 Freiburg, Germany
| | - Florian Full
- Medical Center, Institute of Virology, University of Freiburg, 79104 Freiburg, Germany (F.F.)
- Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Ulrich Kalinke
- TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture between the Hanover Medical School and the Helmholtz Centre for Infection Research, Institute for Experimental Infection Research, 30625 Hanover, Germany
| | - Zsolt Ruzsics
- Medical Center, Institute of Virology, University of Freiburg, 79104 Freiburg, Germany (F.F.)
- Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| |
Collapse
|
7
|
Otero CE, Petkova S, Ebermann M, Taher H, John N, Hoffmann K, Davalos A, Moström MJ, Gilbride RM, Papen CR, Barber-Axthelm A, Scheef EA, Barfield R, Sprehe LM, Kendall S, Manuel TD, Vande Burgt NH, Chan C, Denton M, Streblow ZJ, Streblow DN, Hansen SG, Kaur A, Permar S, Früh K, Hengel H, Malouli D, Kolb P. Rhesus Cytomegalovirus-encoded Fcγ-binding glycoproteins facilitate viral evasion from IgG-mediated humoral immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.27.582371. [PMID: 38464092 PMCID: PMC10925275 DOI: 10.1101/2024.02.27.582371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Human cytomegalovirus (HCMV) encodes four viral Fc-gamma receptors (vFcγRs) that counteract antibody-mediated activation in vitro , but their role in infection and pathogenesis is unknown. To examine the in vivo function of vFcγRs in animal hosts closely related to humans, we identified and characterized vFcγRs encoded by rhesus CMV (RhCMV). We demonstrate that Rh05, Rh152/151 and Rh173 represent the complete set of RhCMV vFcγRs, each displaying functional similarities to their respective HCMV orthologs with respect to antagonizing host FcγR activation in vitro . When RhCMV-naïve rhesus macaques were infected with vFcγR-deleted RhCMV, peak plasma viremia levels and anti-RhCMV antibody responses were comparable to wildtype infections. However, the duration of plasma viremia was significantly shortened in immunocompetent, but not in CD4+ T cell-depleted animals. Since vFcγRs were not required for superinfection, we conclude that vFcγRs delay control by virus-specific adaptive immune responses, particularly antibodies, during primary infection.
Collapse
|
8
|
Picker LJ, Lifson JD, Gale M, Hansen SG, Früh K. Programming cytomegalovirus as an HIV vaccine. Trends Immunol 2023; 44:287-304. [PMID: 36894436 PMCID: PMC10089689 DOI: 10.1016/j.it.2023.02.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/10/2023] [Accepted: 02/11/2023] [Indexed: 03/09/2023]
Abstract
The initial development of cytomegalovirus (CMV) as a vaccine vector for HIV/simian immunodeficiency virus (SIV) was predicated on its potential to pre-position high-frequency, effector-differentiated, CD8+ T cells in tissues for immediate immune interception of nascent primary infection. This goal was achieved and also led to the unexpected discoveries that non-human primate (NHP) CMVs can be programmed to differentially elicit CD8+ T cell responses that recognize viral peptides via classical MHC-Ia, and/or MHC-II, and/or MHC-E, and that MHC-E-restricted CD8+ T cell responses can uniquely mediate stringent arrest and subsequent clearance of highly pathogenic SIV, an unprecedented type of vaccine-mediated protection. These discoveries delineate CMV vector-elicited MHC-E-restricted CD8+ T cells as a functionally distinct T cell response with the potential for superior efficacy against HIV-1, and possibly other infectious agents or cancers.
Collapse
Affiliation(s)
- Louis J Picker
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA.
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, Frederick National Laboratory, Frederick, MD, USA
| | - Michael Gale
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, WA, USA
| | - Scott G Hansen
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Klaus Früh
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| |
Collapse
|
9
|
Ohta E. Pathologic characteristics of infectious diseases in macaque monkeys used in biomedical and toxicologic studies. J Toxicol Pathol 2023; 36:95-122. [PMID: 37101957 PMCID: PMC10123295 DOI: 10.1293/tox.2022-0089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 01/16/2023] [Indexed: 04/28/2023] Open
Abstract
Nonhuman primates (NHPs), which have many advantages in scientific research and are often the only relevant animals to use in assessing the safety profiles and biological or pharmacological effects of drug candidates, including biologics. In scientific or developmental experiments, the immune systems of animals can be spontaneously compromised possibly due to background infection, experimental procedure-associated stress, poor physical condition, or intended or unintended mechanisms of action of test articles. Under these circumstances, background, incidental, or opportunistic infections can seriously can significantly complicate the interpretation of research results and findings and consequently affect experimental conclusions. Pathologists and toxicologists must understand the clinical manifestations and pathologic features of infectious diseases and the effects of these diseases on animal physiology and experimental results in addition to the spectrum of infectious diseases in healthy NHP colonies. This review provides an overview of the clinical and pathologic characteristics of common viral, bacterial, fungal, and parasitic infectious diseases in NHPs, especially macaque monkeys, as well as methods for definitive diagnosis of these diseases. Opportunistic infections that can occur in the laboratory setting have also been addressed in this review with examples of cases of infection disease manifestation that was observed or influenced during safety assessment studies or under experimental conditions.
Collapse
Affiliation(s)
- Etsuko Ohta
- Global Drug Safety, Eisai Co., Ltd., 5-1-3 Tokodai,
Tsukuba-shi, Ibaraki 300-2635, Japan
- *Corresponding author: E Ohta (e-mail: )
| |
Collapse
|
10
|
Recent Developments in NSG and NRG Humanized Mouse Models for Their Use in Viral and Immune Research. Viruses 2023; 15:v15020478. [PMID: 36851692 PMCID: PMC9962986 DOI: 10.3390/v15020478] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 02/04/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Humanized mouse models have been widely used in virology, immunology, and oncology in the last decade. With advances in the generation of knockout mouse strains, it is now possible to generate animals in which human immune cells or human tissue can be engrafted. These models have been used for the study of human infectious diseases, cancers, and autoimmune diseases. In recent years, there has been an increase in the use of humanized mice to model human-specific viral infections. A human immune system in these models is crucial to understand the pathogenesis observed in human patients, which allows for better treatment design and vaccine development. Recent advances in our knowledge about viral pathogenicity and immune response using NSG and NRG mice are reviewed in this paper.
Collapse
|
11
|
Yee JL, Strelow LI, White JA, Rosenthal AN, Barry PA. Horizontal transmission of endemic viruses among rhesus macaques (Macaca mulatta): Implications for human cytomegalovirus vaccine/challenge design. J Med Primatol 2023; 52:53-63. [PMID: 36151734 PMCID: PMC9825633 DOI: 10.1111/jmp.12621] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Rhesus macaques are natural hosts to multiple viruses including rhesus cytomegalovirus (RhCMV), rhesus rhadinovirus (RRV), and Simian Foamy Virus (SFV). While viral infections are ubiquitous, viral transmissions to uninfected animals are incompletely defined. Management procedures of macaque colonies include cohorts that are Specific Pathogen Free (SPF). Greater understanding of viral transmission would augment SPF protocols. Moreover, vaccine/challenge studies of human viruses would be enhanced by leveraging transmission of macaque viruses to recapitulate expected challenges of human vaccine trials. MATERIALS AND METHODS This study characterizes viral transmissions to uninfected animals following inadvertent introduction of RhCMV/RRV/SFV-infected adults to a cohort of uninfected juveniles. Following co-housing with virus-positive adults, juveniles were serially evaluated for viral infection. RESULTS Horizontal viral transmission was rapid and absolute, reaching 100% penetrance between 19 and 78 weeks. CONCLUSIONS This study provides insights into viral natural histories with implications for colony management and modeling vaccine-mediated immune protection studies.
Collapse
Affiliation(s)
- JoAnn L Yee
- California National Primate Research Center, Davis, California, USA
- University of California, Davis, Davis, California, USA
| | - Lisa I Strelow
- California National Primate Research Center, Davis, California, USA
- University of California, Davis, Davis, California, USA
- Center for Immunology and Infectious Diseases, Davis, California, USA
| | - Jessica A White
- California National Primate Research Center, Davis, California, USA
- University of California, Davis, Davis, California, USA
| | - Ann N Rosenthal
- California National Primate Research Center, Davis, California, USA
- University of California, Davis, Davis, California, USA
| | - Peter A Barry
- California National Primate Research Center, Davis, California, USA
- University of California, Davis, Davis, California, USA
- Center for Immunology and Infectious Diseases, Davis, California, USA
| |
Collapse
|
12
|
A Review on Zoonotic Pathogens Associated with Non-Human Primates: Understanding the Potential Threats to Humans. Microorganisms 2023; 11:microorganisms11020246. [PMID: 36838210 PMCID: PMC9964884 DOI: 10.3390/microorganisms11020246] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/07/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023] Open
Abstract
Non-human primates (NHP) share a close relationship with humans due to a genetic homology of 75-98.5%. NHP and humans have highly similar tissue structures, immunity, physiology, and metabolism and thus often can act as hosts to the same pathogens. Agriculture, meat consumption habits, tourism development, religious beliefs, and biological research have led to more extensive and frequent contact between NHPs and humans. Deadly viruses, such as rabies virus, herpes B virus, Marburg virus, Ebola virus, human immunodeficiency virus, and monkeypox virus can be transferred from NHP to humans. Similarly, herpes simplex virus, influenza virus, and yellow fever virus can be transmitted to NHP from humans. Infectious pathogens, including viruses, bacteria, and parasites, can affect the health of both primates and humans. A vast number of NHP-carrying pathogens exhibit a risk of transmission to humans. Therefore, zoonotic infectious diseases should be evaluated in future research. This article reviews the research evidence, diagnostic methods, prevention, and treatment measures that may be useful in limiting the spread of several common viral pathogens via NHP and providing ideas for preventing zoonotic diseases with epidemic potential.
Collapse
|
13
|
Malouli D, Gilbride RM, Wu HL, Hwang JM, Maier N, Hughes CM, Newhouse D, Morrow D, Ventura AB, Law L, Tisoncik-Go J, Whitmore L, Smith E, Golez I, Chang J, Reed JS, Waytashek C, Weber W, Taher H, Uebelhoer LS, Womack JL, McArdle MR, Gao J, Papen CR, Lifson JD, Burwitz BJ, Axthelm MK, Smedley J, Früh K, Gale M, Picker LJ, Hansen SG, Sacha JB. Cytomegalovirus-vaccine-induced unconventional T cell priming and control of SIV replication is conserved between primate species. Cell Host Microbe 2022; 30:1207-1218.e7. [PMID: 35981532 PMCID: PMC9927879 DOI: 10.1016/j.chom.2022.07.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 06/01/2022] [Accepted: 07/19/2022] [Indexed: 01/26/2023]
Abstract
Strain 68-1 rhesus cytomegalovirus expressing simian immunodeficiency virus (SIV) antigens (RhCMV/SIV) primes MHC-E-restricted CD8+ T cells that control SIV replication in 50%-60% of the vaccinated rhesus macaques. Whether this unconventional SIV-specific immunity and protection is unique to rhesus macaques or RhCMV or is intrinsic to CMV remains unknown. Here, using cynomolgus CMV vectors expressing SIV antigens (CyCMV/SIV) and Mauritian cynomolgus macaques, we demonstrate that the induction of MHC-E-restricted CD8+ T cells requires matching CMV to its host species. RhCMV does not elicit MHC-E-restricted CD8+ T cells in cynomolgus macaques. However, cynomolgus macaques vaccinated with species-matched 68-1-like CyCMV/SIV mounted MHC-E-restricted CD8+ T cells, and half of the vaccinees stringently controlled SIV post-challenge. Protected animals manifested a vaccine-induced IL-15 transcriptomic signature that is associated with efficacy in rhesus macaques. These findings demonstrate that the ability of species-matched CMV vectors to elicit MHC-E-restricted CD8+ T cells that are required for anti-SIV efficacy is conserved in nonhuman primates, and these data support the development of HCMV/HIV for a prophylactic HIV vaccine.
Collapse
Affiliation(s)
- Daniel Malouli
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Roxanne M Gilbride
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Helen L Wu
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Joseph M Hwang
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Nicholas Maier
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Colette M Hughes
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Daniel Newhouse
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - David Morrow
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Abigail B Ventura
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Lynn Law
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - Jennifer Tisoncik-Go
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - Leanne Whitmore
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - Elise Smith
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - Inah Golez
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - Jean Chang
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - Jason S Reed
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Courtney Waytashek
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Whitney Weber
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Husam Taher
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Luke S Uebelhoer
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Jennie L Womack
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Matthew R McArdle
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Junwei Gao
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Courtney R Papen
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - Benjamin J Burwitz
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Michael K Axthelm
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Jeremy Smedley
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Klaus Früh
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Michael Gale
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - Louis J Picker
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Scott G Hansen
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA.
| | - Jonah B Sacha
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA; Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97006, USA.
| |
Collapse
|
14
|
Phan QV, Bogdanow B, Wyler E, Landthaler M, Liu F, Hagemeier C, Wiebusch L. Engineering, decoding and systems-level characterization of chimpanzee cytomegalovirus. PLoS Pathog 2022; 18:e1010193. [PMID: 34982803 PMCID: PMC8759705 DOI: 10.1371/journal.ppat.1010193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 01/14/2022] [Accepted: 12/09/2021] [Indexed: 11/19/2022] Open
Abstract
The chimpanzee cytomegalovirus (CCMV) is the closest relative of human CMV (HCMV). Because of the high conservation between these two species and the ability of human cells to fully support CCMV replication, CCMV holds great potential as a model system for HCMV. To make the CCMV genome available for precise and rapid gene manipulation techniques, we captured the genomic DNA of CCMV strain Heberling as a bacterial artificial chromosome (BAC). Selected BAC clones were reconstituted to infectious viruses, growing to similar high titers as parental CCMV. DNA sequencing confirmed the integrity of our clones and led to the identification of two polymorphic loci and a deletion-prone region within the CCMV genome. To re-evaluate the CCMV coding potential, we analyzed the viral transcriptome and proteome and identified several novel ORFs, splice variants, and regulatory RNAs. We further characterized the dynamics of CCMV gene expression and found that viral proteins cluster into five distinct temporal classes. In addition, our datasets revealed that the host response to CCMV infection and the de-regulation of cellular pathways are in line with known hallmarks of HCMV infection. In a first functional experiment, we investigated a proposed frameshift mutation in UL128 that was suspected to restrict CCMV's cell tropism. In fact, repair of this frameshift re-established productive CCMV infection in endothelial and epithelial cells, expanding the options of CCMV as an infection model. Thus, BAC-cloned CCMV can serve as a powerful tool for systematic approaches in comparative functional genomics, exploiting the close phylogenetic relationship between CCMV and HCMV.
Collapse
Affiliation(s)
- Quang Vinh Phan
- Department of Pediatric Oncology/Hematology, Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Boris Bogdanow
- Department of Structural Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Emanuel Wyler
- Berlin Institute for Medical Systems Biology, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Markus Landthaler
- Berlin Institute for Medical Systems Biology, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Fan Liu
- Department of Structural Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Christian Hagemeier
- Department of Pediatric Oncology/Hematology, Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Lüder Wiebusch
- Department of Pediatric Oncology/Hematology, Charité—Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
15
|
Webster H, Valencia S, Kumar A, Chan C, Dennis M, Roark H, Woods A, John S, Carfi A, Permar SR. Pre-existing immunity to cytomegalovirus in macaques influences human CMV vaccine responses in preclinical models. Vaccine 2021; 39:5358-5367. [PMID: 34393017 PMCID: PMC11694353 DOI: 10.1016/j.vaccine.2021.08.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 07/31/2021] [Accepted: 08/04/2021] [Indexed: 02/05/2023]
Abstract
Development of a human cytomegalovirus (HCMV) vaccine is a Tier 1 priority by the National Institutes of Medicine, as HCMV is the most common congenital infection globally and most frequent infectious complication in transplant patients. Relevant preclinical non-human primate models used for testing HCMV vaccine immunogenicity are rhesus and cynomolgous monkeys. However, a complication in using these models is that species-specific CMV variants are endemic in non-human primate breeding colonies. We hypothesize that natural immunity to species-specific CMV in rhesus and cynomolgous monkeys impacts HCMV vaccine immunogenicity and may interfere with our ability to fully interpret vaccine immunogenicity. A modified mRNA vaccine encoding HCMV glycoprotein (gB) and the pentameric complex (PC) packaged in lipid nanoparticles (LNP) was delivered intramuscularly to groups of cynomolgous (n = 16, CyCMV-seropositive) and rhesus macaques (n = 24, RhCMV-seropositive). High pre-vaccination IgG binding responses to HCMV gB were present in both species, but pre-vaccination binding responses to PC were mostly present in rhesus macaques. Yet, at least a log increase in both PC and gB-specific plasma IgG levels was detected post-second HCMV mRNA vaccination in both species. Both species responded with high epithelial cell neutralizing antibody responses at 4 weeks post second HCMV mRNA vaccination, but limited fibroblast neutralizing antibodies. HCMV gB + PC mRNA/LNP vaccine also elicited IgG binding responses to cell-associated gB, an identified immune correlate of protection, in both species after the second vaccination, and there was a moderately strong direct correlation between this pre- and post-vaccination response in rhesus macaques. Based on the correlation between pre-existing and post-vaccine gB-specific binding responses in rhesus macaques, we conclude that species-specific CMV variant-specific antibody responses contribute to antibody responses to HCMV vaccination in primate models, indicating that pre-existing immunity must be taken into account in non-human primate preclinical models and will impact immunogenicity of HCMV vaccines seropositive human vaccinees.
Collapse
Affiliation(s)
- Helen Webster
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Sarah Valencia
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Amit Kumar
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Cliburn Chan
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Maria Dennis
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Hunter Roark
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | - Sallie R Permar
- Weill Cornell Department of Pediatrics, 525 East 68th St, M-622 Box 225, New York, NY 10065.
| |
Collapse
|
16
|
Immune Prophylaxis and Therapy for Human Cytomegalovirus Infection. Int J Mol Sci 2021; 22:ijms22168728. [PMID: 34445434 PMCID: PMC8395925 DOI: 10.3390/ijms22168728] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/29/2021] [Accepted: 07/30/2021] [Indexed: 02/06/2023] Open
Abstract
Human Cytomegalovirus (HCMV) infection is widespread and can result in severe sequelae in susceptible populations. Primary HCMV infection of naïve individuals results in life-long latency characterized by frequent and sporadic reactivations. HCMV infection elicits a robust antibody response, including neutralizing antibodies that can block the infection of susceptible cells in vitro and in vivo. Thus, antibody products and vaccines hold great promise for the prevention and treatment of HCMV, but to date, most attempts to demonstrate their safety and efficacy in clinical trials have been unsuccessful. In this review we summarize publicly available data on these products and highlight new developments and approaches that could assist in successful translation of HCMV immunotherapies.
Collapse
|
17
|
Méndez-Lagares G, Chin N, Chang WW, Lee J, Rosás-Umbert M, Kieu HT, Merriam D, Lu W, Kim S, Adamson L, Brander C, Luciw PA, Barry PA, Hartigan-O’Connor DJ. Cytomegalovirus mediates expansion of IL-15-responsive innate-memory cells with SIV killing function. J Clin Invest 2021; 131:148542. [PMID: 34153005 PMCID: PMC8321572 DOI: 10.1172/jci148542] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 06/03/2021] [Indexed: 12/13/2022] Open
Abstract
Interindividual immune variability is driven predominantly by environmental factors, including exposure to chronic infectious agents such as cytomegalovirus (CMV). We investigated the effects of rhesus CMV (RhCMV) on composition and function of the immune system in young macaques. Within months of infection, RhCMV was associated with impressive changes in antigen presenting cells, T cells, and NK cells-and marked expansion of innate-memory CD8+ T cells. These cells express high levels of NKG2A/C and the IL-2 and IL-15 receptor beta chain, CD122. IL-15 was sufficient to drive differentiation of the cells in vitro and in vivo. Expanded NKG2A/C+CD122+CD8+ T cells in RhCMV-infected macaques, but not their NKG2-negative counterparts, were endowed with cytotoxicity against class I-deficient K562 targets and prompt IFN-γ production in response to stimulation with IL-12 and IL-18. Because RhCMV clone 68-1 forms the viral backbone of RhCMV-vectored SIV vaccines, we also investigated immune changes following administration of RhCMV 68-1-vectored SIV vaccines. These vaccines led to impressive expansion of NKG2A/C+CD8+ T cells with capacity to inhibit SIV replication ex vivo. Thus, CMV infection and CMV-vectored vaccination drive expansion of functional innate-like CD8 cells via host IL-15 production, suggesting that innate-memory expansion could be achieved by other vaccine platforms expressing IL-15.
Collapse
Affiliation(s)
- Gema Méndez-Lagares
- California National Primate Research Center
- Department of Medical Microbiology and Immunology
| | - Ning Chin
- California National Primate Research Center
- Department of Medical Microbiology and Immunology
| | - W.L. William Chang
- California National Primate Research Center
- Department of Medical Microbiology and Immunology
| | - Jaewon Lee
- Graduate Group in Immunology, and
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, California, USA
| | | | - Hung T. Kieu
- California National Primate Research Center
- Department of Medical Microbiology and Immunology
| | - David Merriam
- California National Primate Research Center
- Department of Medical Microbiology and Immunology
| | - Wenze Lu
- California National Primate Research Center
- Department of Medical Microbiology and Immunology
| | - Sungjin Kim
- Department of Medical Microbiology and Immunology
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, California, USA
| | - Lourdes Adamson
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, California, USA
| | - Christian Brander
- IrsiCaixa - AIDS Research Institute, Badalona, Barcelona, Spain
- Universitat de Vic-Universitat Central de Catalunya (UVic-UCC), Vic, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Paul A. Luciw
- California National Primate Research Center
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, California, USA
- Department of Pathology and Laboratory Medicine, University of California, Davis, California, USA
| | - Peter A. Barry
- California National Primate Research Center
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, California, USA
- Department of Pathology and Laboratory Medicine, University of California, Davis, California, USA
| | - Dennis J. Hartigan-O’Connor
- California National Primate Research Center
- Department of Medical Microbiology and Immunology
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California, USA
| |
Collapse
|
18
|
Verweij MC, Hansen SG, Iyer R, John N, Malouli D, Morrow D, Scholz I, Womack J, Abdulhaqq S, Gilbride RM, Hughes CM, Ventura AB, Ford JC, Selseth AN, Oswald K, Shoemaker R, Berkemeier B, Bosche WJ, Hull M, Shao J, Sacha JB, Axthelm MK, Edlefsen PT, Lifson JD, Picker LJ, Früh K. Modulation of MHC-E transport by viral decoy ligands is required for RhCMV/SIV vaccine efficacy. Science 2021; 372:eabe9233. [PMID: 33766941 PMCID: PMC8354429 DOI: 10.1126/science.abe9233] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 03/15/2021] [Indexed: 12/15/2022]
Abstract
Strain 68-1 rhesus cytomegalovirus (RhCMV) vectors expressing simian immunodeficiency virus (SIV) antigens elicit CD8+ T cells recognizing epitopes presented by major histocompatibility complex II (MHC-II) and MHC-E but not MHC-Ia. These immune responses mediate replication arrest of SIV in 50 to 60% of monkeys. We show that the peptide VMAPRTLLL (VL9) embedded within the RhCMV protein Rh67 promotes intracellular MHC-E transport and recognition of RhCMV-infected fibroblasts by MHC-E-restricted CD8+ T cells. Deletion or mutation of viral VL9 abrogated MHC-E-restricted CD8+ T cell priming, resulting in CD8+ T cell responses exclusively targeting MHC-II-restricted epitopes. These responses were comparable in magnitude and differentiation to responses elicited by 68-1 vectors but did not protect against SIV. Thus, Rh67-enabled direct priming of MHC-E-restricted T cells is crucial for RhCMV/SIV vaccine efficacy.
Collapse
Affiliation(s)
- Marieke C Verweij
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Scott G Hansen
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Ravi Iyer
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Nessy John
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Daniel Malouli
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - David Morrow
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Isabel Scholz
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Jennie Womack
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Shaheed Abdulhaqq
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Roxanne M Gilbride
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Colette M Hughes
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Abigail B Ventura
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Julia C Ford
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Andrea N Selseth
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Kelli Oswald
- AIDS and Cancer Virus Program, Frederick National Laboratory, Frederick, MD 21702, USA
| | - Rebecca Shoemaker
- AIDS and Cancer Virus Program, Frederick National Laboratory, Frederick, MD 21702, USA
| | - Brian Berkemeier
- AIDS and Cancer Virus Program, Frederick National Laboratory, Frederick, MD 21702, USA
| | - William J Bosche
- AIDS and Cancer Virus Program, Frederick National Laboratory, Frederick, MD 21702, USA
| | - Michael Hull
- AIDS and Cancer Virus Program, Frederick National Laboratory, Frederick, MD 21702, USA
| | - Jason Shao
- Population Sciences and Computational Biology Programs, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Jonah B Sacha
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Michael K Axthelm
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Paul T Edlefsen
- Population Sciences and Computational Biology Programs, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, Frederick National Laboratory, Frederick, MD 21702, USA
| | - Louis J Picker
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA.
| | - Klaus Früh
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA.
| |
Collapse
|
19
|
Wu HL, Greene JM, Swanson T, Shriver-Munsch C, Armantrout K, Weber WC, Bateman KB, Maier NM, Northrup M, Legasse AW, Moats C, Axthelm MK, Smedley J, Maziarz RT, Martin LD, Hobbs T, Burwitz BJ, Sacha JB. Terumo spectra optia leukapheresis of cynomolgus macaques for hematopoietic stem cell and T cell collection. J Clin Apher 2021; 36:67-77. [PMID: 32941672 PMCID: PMC8670504 DOI: 10.1002/jca.21842] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/28/2020] [Accepted: 09/08/2020] [Indexed: 11/08/2022]
Abstract
Macaques are physiologically relevant animal models of human immunology and infectious disease that have provided key insights and advanced clinical treatment in transplantation, vaccinology, and HIV/AIDS. However, the small size of macaques is a stumbling block for studies requiring large numbers of cells, such as hematopoietic stem cells (HSCs) for transplantation, antigen-specific lymphocytes for in-depth immunological analysis, and latently-infected CD4+ T-cells for HIV cure studies. Here, we provide a detailed protocol for collection of large numbers of HSCs and T-cells from cynomolgus macaques as small as 3 kg using the Terumo Spectra Optia apheresis system, yielding an average of 5.0 × 109 total nucleated cells from mobilized animals and 1.2 × 109 total nucleated cells from nonmobilized animals per procedure. This report provides sufficient detail to adapt this apheresis technique at other institutions, which will facilitate more efficient and detailed analysis of HSCs and their progeny blood cells.
Collapse
Affiliation(s)
- Helen L. Wu
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, 505 NW 185 Avenue, Beaverton, OR 97006, USA
| | - Justin M. Greene
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, 505 NW 185 Avenue, Beaverton, OR 97006, USA
| | - Tonya Swanson
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185 Avenue, Beaverton, OR 97006, USA
| | - Christine Shriver-Munsch
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185 Avenue, Beaverton, OR 97006, USA
| | - Kimberly Armantrout
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185 Avenue, Beaverton, OR 97006, USA
| | - Whitney C. Weber
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, 505 NW 185 Avenue, Beaverton, OR 97006, USA
| | - Katherine B. Bateman
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, 505 NW 185 Avenue, Beaverton, OR 97006, USA
| | - Nicholas M. Maier
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, 505 NW 185 Avenue, Beaverton, OR 97006, USA
| | - Mina Northrup
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, 505 NW 185 Avenue, Beaverton, OR 97006, USA
| | - Alfred W. Legasse
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185 Avenue, Beaverton, OR 97006, USA
| | - Cassandra Moats
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185 Avenue, Beaverton, OR 97006, USA
| | - Michael K. Axthelm
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185 Avenue, Beaverton, OR 97006, USA
| | - Jeremy Smedley
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185 Avenue, Beaverton, OR 97006, USA
| | - Richard T. Maziarz
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Lauren Drew Martin
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185 Avenue, Beaverton, OR 97006, USA
| | - Theodore Hobbs
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185 Avenue, Beaverton, OR 97006, USA
| | - Benjamin J. Burwitz
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, 505 NW 185 Avenue, Beaverton, OR 97006, USA
| | - Jonah B. Sacha
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, 505 NW 185 Avenue, Beaverton, OR 97006, USA
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185 Avenue, Beaverton, OR 97006, USA
| |
Collapse
|
20
|
Taher H, Mahyari E, Kreklywich C, Uebelhoer LS, McArdle MR, Moström MJ, Bhusari A, Nekorchuk M, E X, Whitmer T, Scheef EA, Sprehe LM, Roberts DL, Hughes CM, Jackson KA, Selseth AN, Ventura AB, Cleveland-Rubeor HC, Yue Y, Schmidt KA, Shao J, Edlefsen PT, Smedley J, Kowalik TF, Stanton RJ, Axthelm MK, Estes JD, Hansen SG, Kaur A, Barry PA, Bimber BN, Picker LJ, Streblow DN, Früh K, Malouli D. In vitro and in vivo characterization of a recombinant rhesus cytomegalovirus containing a complete genome. PLoS Pathog 2020; 16:e1008666. [PMID: 33232376 PMCID: PMC7723282 DOI: 10.1371/journal.ppat.1008666] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 12/08/2020] [Accepted: 09/30/2020] [Indexed: 02/07/2023] Open
Abstract
Cytomegaloviruses (CMVs) are highly adapted to their host species resulting in strict species specificity. Hence, in vivo examination of all aspects of CMV biology employs animal models using host-specific CMVs. Infection of rhesus macaques (RM) with rhesus CMV (RhCMV) has been established as a representative model for infection of humans with HCMV due to the close evolutionary relationships of both host and virus. However, the only available RhCMV clone that permits genetic modifications is based on the 68-1 strain which has been passaged in fibroblasts for decades resulting in multiple genomic changes due to tissue culture adaptations. As a result, 68-1 displays reduced viremia in RhCMV-naïve animals and limited shedding compared to non-clonal, low passage isolates. To overcome this limitation, we used sequence information from primary RhCMV isolates to construct a full-length (FL) RhCMV by repairing all mutations affecting open reading frames (ORFs) in the 68-1 bacterial artificial chromosome (BAC). Inoculation of adult, immunocompetent, RhCMV-naïve RM with the reconstituted virus resulted in significant viremia in the blood similar to primary isolates of RhCMV and furthermore led to high viral genome copy numbers in many tissues at day 14 post infection. In contrast, viral dissemination was greatly reduced upon deletion of genes also lacking in 68-1. Transcriptome analysis of infected tissues further revealed that chemokine-like genes deleted in 68-1 are among the most highly expressed viral transcripts both in vitro and in vivo consistent with an important immunomodulatory function of the respective proteins. We conclude that FL-RhCMV displays in vitro and in vivo characteristics of a wildtype virus while being amenable to genetic modifications through BAC recombineering techniques.
Collapse
Affiliation(s)
- Husam Taher
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Eisa Mahyari
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Craig Kreklywich
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Luke S. Uebelhoer
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Matthew R. McArdle
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Matilda J. Moström
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, United States of America
| | - Amruta Bhusari
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Michael Nekorchuk
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Xiaofei E
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Travis Whitmer
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Elizabeth A. Scheef
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, United States of America
| | - Lesli M. Sprehe
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, United States of America
| | - Dawn L. Roberts
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Colette M. Hughes
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Kerianne A. Jackson
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Andrea N. Selseth
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Abigail B. Ventura
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Hillary C. Cleveland-Rubeor
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Yujuan Yue
- Center for Comparative Medicine and Department of Medical Pathology, University of California, Davis, California, United States of America
| | - Kimberli A. Schmidt
- Center for Comparative Medicine and Department of Medical Pathology, University of California, Davis, California, United States of America
| | - Jason Shao
- Statistical Center for HIV/AIDS Research and Prevention, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Paul T. Edlefsen
- Statistical Center for HIV/AIDS Research and Prevention, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Jeremy Smedley
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Timothy F. Kowalik
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Richard J. Stanton
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Michael K. Axthelm
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Jacob D. Estes
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Scott G. Hansen
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Amitinder Kaur
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, United States of America
| | - Peter A. Barry
- Center for Comparative Medicine and Department of Medical Pathology, University of California, Davis, California, United States of America
| | - Benjamin N. Bimber
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Louis J. Picker
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Daniel N. Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Klaus Früh
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Daniel Malouli
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| |
Collapse
|
21
|
Pomplun NL, Vosler L, Weisgrau KL, Furlott J, Weiler AM, Abdelaal HM, Evans DT, Watkins DI, Matano T, Skinner PJ, Friedrich TC, Rakasz EG. Immunophenotyping of Rhesus CMV-Specific CD8 T-Cell Populations. Cytometry A 2020; 99:278-288. [PMID: 32713108 PMCID: PMC7855655 DOI: 10.1002/cyto.a.24197] [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: 02/26/2020] [Revised: 07/12/2020] [Accepted: 07/20/2020] [Indexed: 01/06/2023]
Abstract
A vaccine to ameliorate cytomegalovirus (CMV)-related pathogenicity in transplantation patients is considered a top priority. A therapeutic vaccine must include components that elicit both neutralizing antibodies, and highly effective CD8 T-cell responses. The most important translational model of vaccine development is the captive-bred rhesus macaque (Macaca mulatta) of Indian origin. There is a dearth of information on rhesus cytomegalovirus (rhCMV)-specific CD8 T cells due to the absence of well-defined CD8 T-cell epitopes presented by classical MHC-I molecules. In the current study, we defined two CD8 T-cell epitopes restricted by high-frequency Mamu alleles: the Mamu-A1*002:01 restricted VY9 (VTTLGMALY aa291-299) epitope of protein IE-1, and the Mamu-A1*008:01 restricted NP8 (NPTDRPIP aa96-103) epitope of protein phosphoprotein 65-2. We developed tetramers and determined the level, phenotype, and functional capability of the two epitope-specific T-cell populations in circulation and various tissues. We demonstrated the value of these tetramers for in situ tetramer staining. Here, we first provided critical reagents and established a flow cytometric staining strategy to study rhCMV-specific T-cell responses in up to 40% of captive-bred rhesus macaques. © 2020 The Authors. Cytometry Part A published by Wiley Periodicals LLC on behalf of International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Nicholas L Pomplun
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Logan Vosler
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Kim L Weisgrau
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jessica Furlott
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Andrea M Weiler
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Hadia M Abdelaal
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - David T Evans
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - David I Watkins
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Tetsuro Matano
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Pamela J Skinner
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Thomas C Friedrich
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Eva G Rakasz
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
22
|
Wu HL, Weber WC, Shriver-Munsch C, Swanson T, Northrup M, Price H, Armantrout K, Robertson-LeVay M, Reed JS, Bateman KB, Mahyari E, Thomas A, Junell SL, Hobbs TR, Martin LD, MacAllister R, Bimber BN, Slifka MK, Legasse AW, Moats C, Axthelm MK, Smedley J, Lewis AD, Colgin L, Meyers G, Maziarz RT, Burwitz BJ, Stanton JJ, Sacha JB. Viral opportunistic infections in Mauritian cynomolgus macaques undergoing allogeneic stem cell transplantation mirror human transplant infectious disease complications. Xenotransplantation 2020; 27:e12578. [PMID: 31930750 PMCID: PMC7354885 DOI: 10.1111/xen.12578] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 12/11/2019] [Accepted: 12/18/2019] [Indexed: 12/11/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) and xenotransplantation are accompanied by viral reactivations and virus-associated complications resulting from immune deficiency. Here, in a Mauritian cynomolgus macaque model of fully MHC-matched allogeneic HSCT, we report reactivations of cynomolgus polyomavirus, lymphocryptovirus, and cytomegalovirus, macaque viruses analogous to HSCT-associated human counterparts BK virus, Epstein-Barr virus, and human cytomegalovirus. Viral replication in recipient macaques resulted in characteristic disease manifestations observed in HSCT patients, such as polyomavirus-associated hemorrhagic cystitis and tubulointerstitial nephritis or lymphocryptovirus-associated post-transplant lymphoproliferative disorder. However, in most cases, the reconstituted immune system, alone or in combination with short-term pharmacological intervention, exerted control over viral replication, suggesting engraftment of functional donor-derived immunity. Indeed, the donor-derived reconstituted immune systems of two long-term engrafted HSCT recipient macaques responded to live attenuated yellow fever 17D vaccine (YFV 17D) indistinguishably from untransplanted controls, mounting 17D-targeted neutralizing antibody responses and clearing YFV 17D within 14 days. Together, these data demonstrate that this macaque model of allogeneic HSCT recapitulates clinical situations of opportunistic viral infections in transplant patients and provides a pre-clinical model to test novel prophylactic and therapeutic modalities.
Collapse
Affiliation(s)
- Helen L. Wu
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Whitney C. Weber
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | | | - Tonya Swanson
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Mina Northrup
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Heidi Price
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Kimberly Armantrout
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | | | - Jason S. Reed
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Katherine B. Bateman
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Eisa Mahyari
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Archana Thomas
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Stephanie L. Junell
- Divison of Medical Physics, Department of Radiation Medicine, Oregon Health & Science University, Portland, OR Vaccine and Gene Therapy Institute, Oregon Health
| | - Theodore R. Hobbs
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Lauren D. Martin
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Rhonda MacAllister
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Benjamin N. Bimber
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Mark K. Slifka
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Alfred W. Legasse
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Cassandra Moats
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Michael K. Axthelm
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Jeremy Smedley
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Anne D. Lewis
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Lois Colgin
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Gabrielle Meyers
- Divison of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Richard T. Maziarz
- Divison of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Benjamin J. Burwitz
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Jeffrey J. Stanton
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| | - Jonah B. Sacha
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR
| |
Collapse
|
23
|
Cagliani R, Forni D, Mozzi A, Sironi M. Evolution and Genetic Diversity of Primate Cytomegaloviruses. Microorganisms 2020; 8:E624. [PMID: 32344906 PMCID: PMC7285053 DOI: 10.3390/microorganisms8050624] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/17/2020] [Accepted: 04/19/2020] [Indexed: 12/30/2022] Open
Abstract
Cytomegaloviruses (CMVs) infect many mammals, including humans and non-human primates (NHPs). Human cytomegalovirus (HCMV) is an important opportunistic pathogen among immunocompromised patients and represents the most common infectious cause of birth defects. HCMV possesses a large genome and very high genetic diversity. NHP-infecting CMVs share with HCMV a similar genomic organization and coding content, as well as the course of viral infection. Recent technological advances have allowed the sequencing of several HCMV strains from clinical samples and provided insight into the diversity of NHP-infecting CMVs. The emerging picture indicates that, with the exclusion of core genes (genes that have orthologs in all herpesviruses), CMV genomes are relatively plastic and diverse in terms of gene content, both at the inter- and at the intra-species level. Such variability most likely underlies the strict species-specificity of these viruses, as well as their ability to persist lifelong and with relatively little damage to their hosts. However, core genes, despite their strong conservation, also represented a target of adaptive evolution and subtle changes in their coding sequence contributed to CMV adaptation to different hosts. Indubitably, important knowledge gaps remain, the most relevant of which concerns the role of viral genetics in HCMV-associated human disease.
Collapse
Affiliation(s)
| | | | | | - Manuela Sironi
- Scientific Institute, IRCCS E. MEDEA, Bioinformatics, 23842 Bosisio Parini, Italy
| |
Collapse
|
24
|
Roark HK, Jenks JA, Permar SR, Schleiss MR. Animal Models of Congenital Cytomegalovirus Transmission: Implications for Vaccine Development. J Infect Dis 2020; 221:S60-S73. [PMID: 32134481 PMCID: PMC7057791 DOI: 10.1093/infdis/jiz484] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Although cytomegaloviruses (CMVs) are species-specific, the study of nonhuman CMVs in animal models can help to inform and direct research aimed at developing a human CMV (HCMV) vaccine. Because the driving force behind the development of HCMV vaccines is to prevent congenital infection, the animal model in question must be one in which vertical transmission of virus occurs to the fetus. Fortunately, two such animal models-the rhesus macaque CMV and guinea pig CMV-are characterized by congenital infection. Hence, each model can be evaluated in "proof-of-concept" studies of preconception vaccination aimed at blocking transplacental transmission. This review focuses on similarities and differences in the respective model systems, and it discusses key insights from each model germane to the study of HCMV vaccines.
Collapse
Affiliation(s)
- Hunter K Roark
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Jennifer A Jenks
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Sallie R Permar
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Mark R Schleiss
- Center for Infectious Diseases and Microbiology Translational Research, University of Minnesota Medical School, Department of Pediatrics, Division of Pediatric Infectious Diseases and Immunology, Minneapolis, Minnesota, USA
| |
Collapse
|
25
|
Abstract
: The use of cytomegalovirus (CMV) as a vaccine vector to express antigens against multiple infectious diseases, including simian immunodeficiency virus, Ebola virus, plasmodium, and mycobacterium tuberculosis, in rhesus macaques has generated extraordinary levels of protective immunity against subsequent pathogenic challenge. Moreover, the mechanisms of immune protection have altered paradigms about viral vector-mediated immunity against ectopically expressed vaccine antigens. Further optimization of CMV-vectored vaccines, particularly as this approach moves to human clinical trials will be augmented by a more complete understanding of how CMV engenders mechanisms of immune protection. This review summarizes the particulars of the specific CMV vaccine vector that has been used to date (rhesus CMV strain 68-1) in relation to CMV natural history.
Collapse
|
26
|
Caposio P, van den Worm S, Crawford L, Perez W, Kreklywich C, Gilbride RM, Hughes CM, Ventura AB, Ratts R, Marshall EE, Malouli D, Axthelm MK, Streblow D, Nelson JA, Picker LJ, Hansen SG, Früh K. Characterization of a live-attenuated HCMV-based vaccine platform. Sci Rep 2019; 9:19236. [PMID: 31848362 PMCID: PMC6917771 DOI: 10.1038/s41598-019-55508-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 11/29/2019] [Indexed: 02/07/2023] Open
Abstract
Vaccines based on cytomegalovirus (CMV) demonstrate protection in animal models of infectious disease and cancer. Vaccine efficacy is associated with the ability of CMV to elicit and indefinitely maintain high frequencies of circulating effector memory T cells (TEM) providing continuous, life-long anti-pathogen immune activity. To allow for the clinical testing of human CMV (HCMV)-based vaccines we constructed and characterized as a vector backbone the recombinant molecular clone TR3 representing a wildtype genome. We demonstrate that TR3 can be stably propagated in vitro and that, despite species incompatibility, recombinant TR3 vectors elicit high frequencies of TEM to inserted antigens in rhesus macaques (RM). Live-attenuated versions of TR3 were generated by deleting viral genes required to counteract intrinsic and innate immune responses. In addition, we eliminated subunits of a viral pentameric glycoprotein complex thus limiting cell tropism. We show in a humanized mouse model that such modified vectors were able to establish persistent infection but lost their ability to reactivate from latency. Nevertheless, attenuated TR3 vectors preserved the ability to elicit and maintain TEM to inserted antigens in RM. We further demonstrate that attenuated TR3 can be grown in approved cell lines upon elimination of an anti-viral host factor using small interfering RNA, thus obviating the need for a complementing cell line. In sum, we have established a versatile platform for the clinical development of live attenuated HCMV-vectored vaccines and immunotherapies.
Collapse
Affiliation(s)
- Patrizia Caposio
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, 97006, USA
| | - Sjoerd van den Worm
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, 97006, USA
- Batavia Biosciences B.V., Zernikedreef 16, 2333 CL, Leiden, Netherlands
| | - Lindsey Crawford
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, 97006, USA
| | - Wilma Perez
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, 97006, USA
| | - Craig Kreklywich
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, 97006, USA
| | - Roxanne M Gilbride
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, 97006, USA
| | - Colette M Hughes
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, 97006, USA
| | - Abigail B Ventura
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, 97006, USA
| | - Robert Ratts
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, 97006, USA
- Vir Biotechnology, 4640, SW Macadam Avenue, Portland, OR, 97239, USA
| | - Emily E Marshall
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, 97006, USA
- Vir Biotechnology, 4640, SW Macadam Avenue, Portland, OR, 97239, USA
| | - Daniel Malouli
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, 97006, USA
| | - Michael K Axthelm
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, 97006, USA
| | - Daniel Streblow
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, 97006, USA
| | - Jay A Nelson
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, 97006, USA
| | - Louis J Picker
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, 97006, USA
| | - Scott G Hansen
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, 97006, USA.
| | - Klaus Früh
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, 97006, USA.
| |
Collapse
|
27
|
Murthy S, O'Brien K, Agbor A, Angedakin S, Arandjelovic M, Ayimisin EA, Bailey E, Bergl RA, Brazzola G, Dieguez P, Eno-Nku M, Eshuis H, Fruth B, Gillespie TR, Ginath Y, Gray M, Herbinger I, Jones S, Kehoe L, Kühl H, Kujirakwinja D, Lee K, Madinda NF, Mitamba G, Muhindo E, Nishuli R, Ormsby LJ, Petrzelkova KJ, Plumptre AJ, Robbins MM, Sommer V, Ter Heegde M, Todd A, Tokunda R, Wessling E, Jarvis MA, Leendertz FH, Ehlers B, Calvignac-Spencer S. Cytomegalovirus distribution and evolution in hominines. Virus Evol 2019; 5:vez015. [PMID: 31384482 PMCID: PMC6671425 DOI: 10.1093/ve/vez015] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Herpesviruses are thought to have evolved in very close association with their hosts. This is notably the case for cytomegaloviruses (CMVs; genus Cytomegalovirus) infecting primates, which exhibit a strong signal of co-divergence with their hosts. Some herpesviruses are however known to have crossed species barriers. Based on a limited sampling of CMV diversity in the hominine (African great ape and human) lineage, we hypothesized that chimpanzees and gorillas might have mutually exchanged CMVs in the past. Here, we performed a comprehensive molecular screening of all 9 African great ape species/subspecies, using 675 fecal samples collected from wild animals. We identified CMVs in eight species/subspecies, notably generating the first CMV sequences from bonobos. We used this extended dataset to test competing hypotheses with various degrees of co-divergence/number of host switches while simultaneously estimating the dates of these events in a Bayesian framework. The model best supported by the data involved the transmission of a gorilla CMV to the panine (chimpanzee and bonobo) lineage and the transmission of a panine CMV to the gorilla lineage prior to the divergence of chimpanzees and bonobos, more than 800,000 years ago. Panine CMVs then co-diverged with their hosts. These results add to a growing body of evidence suggesting that viruses with a double-stranded DNA genome (including other herpesviruses, adenoviruses, and papillomaviruses) often jumped between hominine lineages over the last few million years.
Collapse
Affiliation(s)
- Sripriya Murthy
- Division 12 "Measles, Mumps, Rubella and Viruses Affecting Immune-Compromised Patients" Robert Koch Institute, Berlin, Germany
| | - Kathryn O'Brien
- School of Biomedical and Healthcare Sciences, University of Plymouth, Devon, UK
| | - Anthony Agbor
- Max Planck Institute for Evolutionary Anthropology (MPI EVA), Leipzig, Germany.,African Parks Network, Lonehill, Republic of South Africa
| | - Samuel Angedakin
- Max Planck Institute for Evolutionary Anthropology (MPI EVA), Leipzig, Germany
| | - Mimi Arandjelovic
- Max Planck Institute for Evolutionary Anthropology (MPI EVA), Leipzig, Germany
| | | | - Emma Bailey
- Max Planck Institute for Evolutionary Anthropology (MPI EVA), Leipzig, Germany
| | | | - Gregory Brazzola
- Max Planck Institute for Evolutionary Anthropology (MPI EVA), Leipzig, Germany
| | - Paula Dieguez
- Max Planck Institute for Evolutionary Anthropology (MPI EVA), Leipzig, Germany
| | | | - Henk Eshuis
- Max Planck Institute for Evolutionary Anthropology (MPI EVA), Leipzig, Germany
| | - Barbara Fruth
- Faculty of Science, School of Natural Sciences and hPsychology, Liverpool John Moores University, Liverpool, UK.,Centre for Research and Conservation, Royal Zoological Society of Antwerp, Antwerp, Belgium
| | - Thomas R Gillespie
- Department of Environmental Sciences and Program in Population Biology, Ecology, and Evolutionary Biology, Emory University, Atlanta, USA
| | - Yisa Ginath
- Max Planck Institute for Evolutionary Anthropology (MPI EVA), Leipzig, Germany
| | - Maryke Gray
- International Gorilla Conservation Programme, Kigali, Rwanda.,Batavia Coast Maritime Institute, Geraldton, WA, Australia
| | | | - Sorrel Jones
- Max Planck Institute for Evolutionary Anthropology (MPI EVA), Leipzig, Germany.,Royal Holloway, University of London, Egham, UK
| | - Laura Kehoe
- Wild Chimpanzee Foundation (WCF), Leipzig, Germany.,Department of Biology, University of Victoria, Victoria, Canada.,Department of Forest and Conservation Sciences, University of British Columbia, Vancouver, Canada
| | - Hjalmar Kühl
- Max Planck Institute for Evolutionary Anthropology (MPI EVA), Leipzig, Germany.,German Centre for Integrative Biodiversity Research (iDiv), Leipzig, Germany
| | | | - Kevin Lee
- Max Planck Institute for Evolutionary Anthropology (MPI EVA), Leipzig, Germany.,School of Human Evolution and Social Change, Arizona State University, Tempe, USA
| | - Nadège F Madinda
- Max Planck Institute for Evolutionary Anthropology (MPI EVA), Leipzig, Germany.,Epidemiology of highly pathogenic microorganisms, Robert Koch Institute, Berlin, Germany
| | | | | | - Radar Nishuli
- Réserve de Faune à Okapis, Institut Congolais pour la Conservation de la Nature, Kinshasa, Democratic Republic of the Congo
| | - Lucy J Ormsby
- Max Planck Institute for Evolutionary Anthropology (MPI EVA), Leipzig, Germany
| | - Klara J Petrzelkova
- Institute of Vertebrate Biology, Academy of Sciences, Brno, Czech Republic.,Department of Pathology and Parasitology, University of Veterinary and Pharmaceutical Sciences, Brno, Czech Republic.,Biology Centre, Institute of Parasitology, Academy of Sciences of the Czech Republic, Ceske Budejovice, Czech Republic.,Liberec Zoo, Liberec, Czech Republic
| | - Andrew J Plumptre
- Wildlife Conservation Society, NY, USA.,KBA Secretariat, c/o BirdLife International, Cambridge, UK.,Zoology Department, Conservation Science Group, University of Cambridge, Cambridge, UK
| | - Martha M Robbins
- Max Planck Institute for Evolutionary Anthropology (MPI EVA), Leipzig, Germany
| | - Volker Sommer
- Gashaka Primate Project, Nigeria c/o Department of Anthropology, University College London, London, UK
| | - Martijn Ter Heegde
- Epidemiology of highly pathogenic microorganisms, Robert Koch Institute, Berlin, Germany
| | - Angelique Todd
- Dzanga Sangha Protected Areas, WWF Central African Republic, Bangui, Central African Republic
| | - Raymond Tokunda
- Institute of Vertebrate Biology, Academy of Sciences, Brno, Czech Republic
| | - Erin Wessling
- Max Planck Institute for Evolutionary Anthropology (MPI EVA), Leipzig, Germany.,Dzanga Sangha Protected Areas, WWF Central African Republic, Bangui, Central African Republic
| | - Michael A Jarvis
- School of Biomedical and Healthcare Sciences, University of Plymouth, Devon, UK
| | - Fabian H Leendertz
- Epidemiology of highly pathogenic microorganisms, Robert Koch Institute, Berlin, Germany
| | - Bernhard Ehlers
- Division 12 "Measles, Mumps, Rubella and Viruses Affecting Immune-Compromised Patients" Robert Koch Institute, Berlin, Germany
| | - Sébastien Calvignac-Spencer
- Epidemiology of highly pathogenic microorganisms, Robert Koch Institute, Berlin, Germany.,Viral Evolution, Robert Koch Institute, Berlin, Germany
| |
Collapse
|
28
|
Marshall EE, Malouli D, Hansen SG, Gilbride RM, Hughes CM, Ventura AB, Ainslie E, Selseth AN, Ford JC, Burke D, Kreklywich CN, Womack J, Legasse AW, Axthelm MK, Kahl C, Streblow D, Edlefsen PT, Picker LJ, Früh K. Enhancing safety of cytomegalovirus-based vaccine vectors by engaging host intrinsic immunity. Sci Transl Med 2019; 11:eaaw2603. [PMID: 31316006 PMCID: PMC6830438 DOI: 10.1126/scitranslmed.aaw2603] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 03/14/2019] [Accepted: 06/13/2019] [Indexed: 12/29/2022]
Abstract
Rhesus cytomegalovirus (RhCMV)-based vaccines maintain effector memory T cell responses (TEM) that protect ~50% of rhesus monkeys (RMs) challenged with simian immunodeficiency virus (SIV). Because human CMV (HCMV) causes disease in immunodeficient subjects, clinical translation will depend upon attenuation strategies that reduce pathogenic potential without sacrificing CMV's unique immunological properties. We demonstrate that "intrinsic" immunity can be used to attenuate strain 68-1 RhCMV vectors without impairment of immunogenicity. The tegument proteins pp71 and UL35 encoded by UL82 and UL35 of HCMV counteract cell-intrinsic restriction via degradation of host transcriptional repressors. When the corresponding RhCMV genes, Rh110 and Rh59, were deleted from 68-1 RhCMV (ΔRh110 and ΔRh59), we observed only a modest growth defect in vitro, but in vivo, these modified vectors manifested little to no amplification at the injection site and dissemination to distant sites, in contrast to parental 68-1 RhCMV. ΔRh110 was not shed at any time after infection and was not transmitted to naïve hosts either by close contact (mother to infant) or by leukocyte transfusion. In contrast, ΔRh59 was both shed and transmitted by leukocyte transfusion, indicating less effective attenuation than pp71 deletion. The T cell immunogenicity of ΔRh110 was essentially identical to 68-1 RhCMV with respect to magnitude, TEM phenotype, epitope targeting, and durability. Thus, pp71 deletion preserves CMV vector immunogenicity while stringently limiting vector spread, making pp71 deletion an attractive attenuation strategy for HCMV vectors.
Collapse
Affiliation(s)
- Emily E Marshall
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Daniel Malouli
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Scott G Hansen
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Roxanne M Gilbride
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Colette M Hughes
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Abigail B Ventura
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Emily Ainslie
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Andrea N Selseth
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Julia C Ford
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - David Burke
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Craig N Kreklywich
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Jennie Womack
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Alfred W Legasse
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Michael K Axthelm
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Christoph Kahl
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Daniel Streblow
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Paul T Edlefsen
- Statistical Center for HIV/AIDS Research and Prevention, Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Louis J Picker
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA.
| | - Klaus Früh
- Vaccine and Gene Therapy Institute and Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA.
| |
Collapse
|
29
|
The Susceptibility of Primary Dermis Fibroblasts from the Chinese Tree Shrew to Human Cytomegalovirus Infection. Virol Sin 2019; 34:270-277. [PMID: 30989428 DOI: 10.1007/s12250-019-00106-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 02/18/2019] [Indexed: 10/27/2022] Open
Abstract
As a universal pathogen leading to neonatal defects and transplant failure, human cytomegalovirus (HCMV) has strict species specificity and this has prevented the development of a suitable animal model for the pathogenesis study. The mechanism of cross-species barrier remains elusive and there are so far no non-human cell culture models that support HCMV replication. The Chinese tree shrew (Tupaia belangeri chinensis) is a small laboratory animal and evolutionary closely related with primates. We investigated the susceptibility of primary tree shrew dermis fibroblasts (TSDF) to HCMV infection. Infection with a GFP-expressing HCMV virus resulted in green fluorescence in infected cells with the expression of IE1, UL44 and pp28. The titers of cell-free viruses reached 103 PFU/mL at 96 hpi, compared to titers of 104 PFU/mL observed in primary human foreskin fibroblasts. Our results suggested that TSDF was semi-permissive for HCMV infection. The TSDF model could be further used to investigate key factors influencing cross-species multiplication of HCMV.
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW CMV-vectored vaccines expressing SIV antigens have mediated unprecedented levels of virus control following SIV challenge in rhesus macaques. Remarkably, protection was dependent on nonclassically restricted CD8 T cells. Here, we review the latest research in CMV-vectored vaccines in both humans and nonhuman primates as well as recent advances in the understanding nonclassically restricted T cells, particularly MHC-E-restricted CD8 T cells. RECENT FINDINGS Recent studies have investigated human translation of CMV-vectored vaccines including studies to ensure vaccine vector safety. Other work has focused on testing of animal models to investigate the relative contribution of MHC diversity and CMV strain on T-cell induction. Lastly, several groups have investigated MHC-E peptide binding, including HLA-E, have found that MHC-E can accommodate different peptide motifs, consistent with the original observations in CMV-vaccinated macaques. SUMMARY CMV remains a promising vaccine vector with the potential to be protective against multiple diseases, including HIV. However, CMV is highly species-specific and in humans, congenital infection can lead to serious birth defects. To ensure safe translation to humans, further clinical and animal studies are needed to better understand CMV-vectored immunity as well as more basic immunological questions relating to the induction of classical vs. nonclassical T cells.
Collapse
Affiliation(s)
- Maria Abad-Fernandez
- UNC HIV Cure Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Nilu Goonetilleke
- UNC HIV Cure Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
31
|
Kolb P, Sijmons S, McArdle MR, Taher H, Womack J, Hughes C, Ventura A, Jarvis MA, Stahl-Hennig C, Hansen S, Picker LJ, Malouli D, Hengel H, Früh K. Identification and Functional Characterization of a Novel Fc Gamma-Binding Glycoprotein in Rhesus Cytomegalovirus. J Virol 2019; 93:e02077-18. [PMID: 30487278 PMCID: PMC6364020 DOI: 10.1128/jvi.02077-18] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 11/26/2018] [Indexed: 12/20/2022] Open
Abstract
Receptors recognizing the Fc part of immunoglobulin G (FcγRs) are key determinants in antibody-mediated immune responses. Members of the Herpesviridae interfere with this immune regulatory network by expressing viral FcγRs (vFcγRs). Human cytomegalovirus (HCMV) encodes four distinct vFcγRs that differ with respect to their IgG subtype specificity and their impact on antibody-mediated immune function in vitro The impact of vFcγRs on HCMV pathogenesis and immunomodulation in vivo is not known. The closest evolutionary animal model of HCMV is rhesus CMV (RhCMV) infection of rhesus macaques. To enable the characterization of vFcγR function in this model, we studied IgG binding by RhCMV. We show that lysates of RhCMV-infected cells contain an IgG-binding protein of 30 kDa encoded by the gene Rh05 that is a predicted type I glycoprotein belonging to the RL11 gene family. Upon deletion of Rh05, IgG-Fc binding by RhCMV strain 68-1 is lost, whereas ectopic expression of Rh05 results in IgG binding to transfected cells consistent with Rh05 being a vFcγR. Using a set of reporter cell lines stably expressing human and rhesus FcγRs, we further demonstrate that Rh05 antagonizes host FcγR activation. Compared to Rh05-intact RhCMV, RhCMVΔRh05 showed an increased activation of host FcγR upon exposure of infected cells to IgG from RhCMV-seropositive animals, suggesting that Rh05 protects infected cells from opsonization and IgG-dependent activation of host FcγRs. However, antagonizing host FcγR activation by Rh05 was not required for the establishment and maintenance of infection of RhCMV, even in a seropositive host, as shown by the induction of T cell responses to heterologous antigens expressed by RhCMV lacking the gene region encoding Rh05. In contrast to viral evasion of natural killer cells or T cell recognition, the evasion of antibody-mediated effects does not seem to be absolutely required for infection or reinfection. The identification of the first vFcγR that efficiently antagonizes host FcγR activation in the RhCMV genome will thus permit more detailed studies of this immunomodulatory mechanism in promoting viral dissemination in the presence of natural or vaccine-induced humoral immunity.IMPORTANCE Rhesus cytomegalovirus (RhCMV) offers a unique model for studying human cytomegalovirus (HCMV) pathogenesis and vaccine development. RhCMV infection of nonhuman primates greatly broadened the understanding of mechanisms by which CMVs evade or reprogram T cell and natural killer cell responses in vivo However, the role of humoral immunity and viral modulation of anti-CMV antibodies has not been studied in this model. There is evidence from in vitro studies that HCMVs can evade humoral immunity. By gene mapping and with the help of a novel cell-based reporter assay system we characterized the first RhCMV encoded IgG-Fcγ binding glycoprotein as a potent antagonist of rhesus FcγR activation. We further demonstrate that, unlike evasion of T cell immunity, this viral Fcγ receptor is not required to overcome anti-CMV immunity to establish secondary infections. These findings enable more detailed studies of the in vivo consequences of CMV evasion from IgG responses in nonhuman primate models.
Collapse
Affiliation(s)
- Philipp Kolb
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Steven Sijmons
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Matthew R McArdle
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Husam Taher
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Jennie Womack
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Colette Hughes
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Abigail Ventura
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Michael A Jarvis
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | | | - Scott Hansen
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Louis J Picker
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Daniel Malouli
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Hartmut Hengel
- Institute of Virology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Klaus Früh
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| |
Collapse
|
32
|
Lai JH, Luo SF, Ho LJ. Operation of mitochondrial machinery in viral infection-induced immune responses. Biochem Pharmacol 2018; 156:348-356. [PMID: 30172712 PMCID: PMC7092938 DOI: 10.1016/j.bcp.2018.08.044] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 08/29/2018] [Indexed: 01/05/2023]
Abstract
Mitochondria have been recognized as ancient bacteria that contain evolutionary endosymbionts. Metabolic pathways and inflammatory signals interact within mitochondria in response to different stresses, such as viral infections. In this commentary, we address several interesting questions, including (1) how do mitochondrial machineries participate in immune responses; (2) how do mitochondria mediate antiviral immunity; (3) what mechanisms involved in mitochondrial machinery, including the downregulation of mitochondrial DNA (mtDNA), disturbances of mitochondrial dynamics, and the induction of mitophagy and regulation of apoptosis, have been adopted by viruses to evade antiviral immunity; (4) what mechanisms involve the regulation of mitochondrial machineries in antiviral therapeutics; and (5) what are the potential challenges and perspectives in developing mitochondria-targeting antiviral treatments? This commentary provides a comprehensive review of the roles and mechanisms of mitochondrial machineries in immunity, viral infections and related antiviral therapeutics.
Collapse
Affiliation(s)
- Jenn-Haung Lai
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Tao-Yuan, Taiwan, ROC; Graduate Institute of Medical Science, National Defense Medical Center, Taipei, Taiwan, ROC.
| | - Shue-Fen Luo
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Tao-Yuan, Taiwan, ROC
| | - Ling-Jun Ho
- Institute of Cellular and System Medicine, National Health Research Institute, Zhunan, Taiwan, ROC.
| |
Collapse
|
33
|
WDR5 Facilitates Human Cytomegalovirus Replication by Promoting Capsid Nuclear Egress. J Virol 2018; 92:JVI.00207-18. [PMID: 29437978 DOI: 10.1128/jvi.00207-18] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Accepted: 02/05/2018] [Indexed: 01/27/2023] Open
Abstract
WD repeat-containing protein 5 (WDR5) is essential for assembling the VISA-associated complex to induce a type I interferon antiviral response to Sendai virus infection. However, the roles of WDR5 in DNA virus infections are not well described. Here, we report that human cytomegalovirus exploits WDR5 to facilitate capsid nuclear egress. Overexpression of WDR5 in fibroblasts slightly enhanced the infectious virus yield. However, WDR5 knockdown dramatically reduced infectious virus titers with only a small decrease in viral genome replication or gene expression. Further investigation of late steps of viral replication found that WDR5 knockdown significantly impaired formation of the viral nuclear egress complex and induced substantially fewer infoldings of the inner nuclear membrane. In addition, fewer capsids were associated with these infoldings, and there were fewer capsids in the cytoplasm. Restoration of WDR5 partially reversed these effects. These results suggest that WDR5 knockdown impairs the nuclear egress of capsids, which in turn decreases virus titers. These findings reveal an important role for a host factor whose function(s) is usurped by a viral pathogen to promote efficient replication. Thus, WDR5 represents an interesting regulatory mechanism and a potential antiviral target.IMPORTANCE Human cytomegalovirus (HCMV) has a large (∼235-kb) genome with over 170 open reading frames and exploits numerous cellular factors to facilitate its replication. HCMV infection increases protein levels of WD repeat-containing protein 5 (WDR5) during infection, overexpression of WDR5 enhances viral replication, and knockdown of WDR5 dramatically attenuates viral replication. Our results indicate that WDR5 promotes the nuclear egress of viral capsids, the depletion of WDR5 resulting in a significant decrease in production of infectious virions. This is the first report that WDR5 favors HCMV, a DNA virus, replication and highlights a novel target for antiviral therapy.
Collapse
|
34
|
Ke F, Gui JF, Chen ZY, Li T, Lei CK, Wang ZH, Zhang QY. Divergent transcriptomic responses underlying the ranaviruses-amphibian interaction processes on interspecies infection of Chinese giant salamander. BMC Genomics 2018; 19:211. [PMID: 29558886 PMCID: PMC5861657 DOI: 10.1186/s12864-018-4596-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 03/12/2018] [Indexed: 12/21/2022] Open
Abstract
Background Ranaviruses (family Iridoviridae, nucleocytoplasmic large DNA viruses) have been reported as promiscuous pathogens of cold-blooded vertebrates. Rana grylio virus (RGV, a ranavirus), from diseased frog Rana grylio with a genome of 105.79 kb and Andrias davidianus ranavirus (ADRV), from diseased Chinese giant salamander (CGS) with a genome of 106.73 kb, contains 99% homologous genes. Results To uncover the differences in virus replication and host responses under interspecies infection, we analyzed transcriptomes of CGS challenged with RGV and ADRV in different time points (1d, 7d) for the first time. A total of 128,533 unigenes were obtained from 820,858,128 clean reads. Transcriptome analysis revealed stronger gene expression of RGV than ADRV at 1 d post infection (dpi), which was supported by infection in vitro. RGV replicated faster and had higher titers than ADRV in cultured CGS cell line. RT-qPCR revealed the RGV genes including the immediate early gene (RGV-89R) had higher expression level than that of ADRV at 1 dpi. It further verified the acute infection of RGV in interspecies infection. The number of differentially expressed genes and enriched pathways from RGV were lower than that from ADRV, which reflected the variant host responses at transcriptional level. No obvious changes of key components in pathway “Antigen processing and presentation” were detected for RGV at 1 dpi. Contrarily, ADRV infection down-regulated the expression levels of MHC I and CD8. The divergent host immune responses revealed the differences between interspecies and natural infection, which may resulted in different fates of the two viruses. Altogether, these results revealed the differences in transcriptome responses among ranavirus interspecies infection of amphibian and new insights in DNA virus-host interactions in interspecies infection. Conclusion The DNA virus (RGV) not only expressed self-genes and replicated quickly after entry into host under interspecies infection, but also avoided the over-activation of host responses. The strategy could gain time for the survival of interspecies pathogen, and may provide opportunity for its adaptive evolution and interspecies transmission. Electronic supplementary material The online version of this article (10.1186/s12864-018-4596-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fei Ke
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Jian-Fang Gui
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Zhong-Yuan Chen
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Tao Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Cun-Ke Lei
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Zi-Hao Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Qi-Ya Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China.
| |
Collapse
|
35
|
Abstract
The development of a cytomegalovirus (CMV) vaccine has become a top priority due to its potential cost-effectiveness and associated public health benefits. However, there are a number of challenges facing vaccine development including the following: (1) CMV has many mechanisms for evading immune responses , and natural immunity is not perfect, (2) the immune correlates for protection are unclear, (3) a narrow range of CMV hosts limits the value of animal models, and (4) the placenta is a specialized organ formed transiently and its immunological status changes with time. In spite of these limitations, several types of CMV vaccine candidate, including live-attenuated, DISC , subunit, DNA, vectored, and peptide vaccines, have been developed or are currently under development. The recognition of the pentameric complex as the major neutralization target and identification of various strategies to block viral immune response evasion mechanisms have opened new avenues to CMV vaccine development. Here, we discuss the immune correlates for protection, the characteristics of the various vaccine candidates and their clinical trials, and the relevant animal models.
Collapse
|
36
|
Cytomegaloviruses in a Community of Wild Nonhuman Primates in Taï National Park, Côte D'Ivoire. Viruses 2017; 10:v10010011. [PMID: 29286318 PMCID: PMC5795424 DOI: 10.3390/v10010011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 12/19/2017] [Accepted: 12/20/2017] [Indexed: 12/01/2022] Open
Abstract
Cytomegaloviruses (CMVs) are known to infect many mammals, including a number of nonhuman primates (NHPs). However, most data available arose from studies led on captive individuals and little is known about CMV diversity in wild NHPs. Here, we analyzed a community of wild nonhuman primates (seven species) in Taï National Park (TNP), Côte d’Ivoire, with two PCR systems targeting betaherpesviruses. CMV DNA was detected in 17/87 primates (4/7 species). Six novel CMVs were identified in sooty mangabeys, Campbell’s monkeys and Diana monkeys, respectively. In 3/17 positive individuals (from three NHP species), different CMVs were co-detected. A major part of the glycoprotein B coding sequences of the novel viruses was amplified and sequenced, and phylogenetic analyses were performed that included three previously discovered CMVs of western red colobus from TNP and published CMVs from other NHP species and geographic locations. We find that, despite this locally intensified sampling, NHP CMVs from TNP are completely host-specific, pinpointing the absence or rarity of cross-species transmission. We also show that on longer timescales the evolution of CMVs is characterized by frequent co-divergence with their hosts, although other processes, including lineage duplication and host switching, also have to be invoked to fully explain their evolutionary relationships.
Collapse
|
37
|
Burwitz BJ, Wu HL, Abdulhaqq S, Shriver-Munsch C, Swanson T, Legasse AW, Hammond KB, Junell SL, Reed JS, Bimber BN, Greene JM, Webb GM, Northrup M, Laub W, Kievit P, MacAllister R, Axthelm MK, Ducore R, Lewis A, Colgin LMA, Hobbs T, Martin LD, Ferguson B, Thomas CR, Panoskaltsis-Mortari A, Meyers G, Stanton JJ, Maziarz RT, Sacha JB. Allogeneic stem cell transplantation in fully MHC-matched Mauritian cynomolgus macaques recapitulates diverse human clinical outcomes. Nat Commun 2017; 8:1418. [PMID: 29127275 PMCID: PMC5681693 DOI: 10.1038/s41467-017-01631-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/04/2017] [Indexed: 12/31/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is a critically important therapy for hematological malignancies, inborn errors of metabolism, and immunodeficiency disorders, yet complications such as graft-vs.-host disease (GvHD) limit survival. Development of anti-GvHD therapies that do not adversely affect susceptibility to infection or graft-vs.-tumor immunity are hampered by the lack of a physiologically relevant, preclinical model of allogeneic HSCT. Here we show a spectrum of diverse clinical HSCT outcomes including primary and secondary graft failure, lethal GvHD, and stable, disease-free full donor engraftment using reduced intensity conditioning and mobilized peripheral blood HSCT in unrelated, fully MHC-matched Mauritian-origin cynomolgus macaques. Anti-GvHD prophylaxis of tacrolimus, post-transplant cyclophosphamide, and CD28 blockade induces multi-lineage, full donor chimerism and recipient-specific tolerance while maintaining pathogen-specific immunity. These results establish a new preclinical allogeneic HSCT model for evaluation of GvHD prophylaxis and next-generation HSCT-mediated therapies for solid organ tolerance, cure of non-malignant hematological disease, and HIV reservoir clearance.
Collapse
Affiliation(s)
- Benjamin J Burwitz
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
| | - Helen L Wu
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
| | - Shaheed Abdulhaqq
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
| | - Christine Shriver-Munsch
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
| | - Tonya Swanson
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
| | - Alfred W Legasse
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
| | - Katherine B Hammond
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
| | - Stephanie L Junell
- Division of Medical Physics, Department of Radiation Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Jason S Reed
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
| | - Benjamin N Bimber
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
| | - Justin M Greene
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
| | - Gabriela M Webb
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
| | - Mina Northrup
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
| | - Wolfram Laub
- Division of Medical Physics, Department of Radiation Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Paul Kievit
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
| | - Rhonda MacAllister
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
| | - Michael K Axthelm
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
| | - Rebecca Ducore
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
| | - Anne Lewis
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
| | - Lois M A Colgin
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
| | - Theodore Hobbs
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
| | - Lauren D Martin
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
| | - Betsy Ferguson
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
| | - Charles R Thomas
- Division of Medical Physics, Department of Radiation Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Angela Panoskaltsis-Mortari
- Division of Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, 2450 Riverside Avenue, Minneapolis, MN, 55454, USA
| | - Gabrielle Meyers
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Jeffrey J Stanton
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA
| | - Richard T Maziarz
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Jonah B Sacha
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA.
- Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR, 97006, USA.
| |
Collapse
|
38
|
Bailer SM, Funk C, Riedl A, Ruzsics Z. Herpesviral vectors and their application in oncolytic therapy, vaccination, and gene transfer. Virus Genes 2017. [PMID: 28634751 DOI: 10.1007/s11262-017-1482-7] [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: 12/22/2022]
Abstract
Herpesviruses are enveloped DNA viruses that infect vertebrate cells. Their high potential cloning capacity and the lifelong persistence of their genomes in various host cells make them attractive platforms for vector-based therapy. In this review, we would like to highlight recent advances of three major areas of herpesvirus vector development and application: (i) oncolytic therapy, (ii) recombinant vaccines, and (iii) large capacity gene transfer vehicles.
Collapse
Affiliation(s)
- Susanne M Bailer
- Institute for Interfacial Engineering and Plasma Technology IGVP, University of Stuttgart, Nobelstrasse 12, 70569, Stuttgart, Germany. .,Fraunhofer-Institut für Grenzflächen- und Bioverfahrenstechnik IGB, Nobelstrasse 12, 70569, Stuttgart, Germany.
| | - Christina Funk
- Institute for Interfacial Engineering and Plasma Technology IGVP, University of Stuttgart, Nobelstrasse 12, 70569, Stuttgart, Germany.,Fraunhofer-Institut für Grenzflächen- und Bioverfahrenstechnik IGB, Nobelstrasse 12, 70569, Stuttgart, Germany
| | - André Riedl
- Department for Medical Microbiology and Hygiene, Institute of Virology, University Medical Center Freiburg, Hermann-Herder-Strasse 11, 79104, Freiburg, Germany.,German Center for Infection Research - DZIF, Freiburg, Germany
| | - Zsolt Ruzsics
- Department for Medical Microbiology and Hygiene, Institute of Virology, University Medical Center Freiburg, Hermann-Herder-Strasse 11, 79104, Freiburg, Germany. .,German Center for Infection Research - DZIF, Freiburg, Germany.
| |
Collapse
|