1
|
Eletreby M, Thiessen L, Prager A, Brizic I, Materljan J, Kubic L, Jäger K, Jurinović K, Jerak J, Krey K, Adler B. Dissecting the cytomegalovirus CC chemokine: Chemokine activity and gHgLchemokine-dependent cell tropism are independent players in CMV infection. PLoS Pathog 2023; 19:e1011793. [PMID: 38064525 PMCID: PMC10732436 DOI: 10.1371/journal.ppat.1011793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 12/20/2023] [Accepted: 11/01/2023] [Indexed: 12/21/2023] Open
Abstract
Like all herpesviruses, cytomegaloviruses (CMVs) code for many immunomodulatory proteins including chemokines. The human cytomegalovirus (HCMV) CC chemokine pUL128 has a dual role in the infection cycle. On one hand, it forms the pentameric receptor-binding complex gHgLpUL(128,130,131A), which is crucial for the broad cell tropism of HCMV. On the other hand, it is an active chemokine that attracts leukocytes and shapes their activation. All animal CMVs studied so far have functionally homologous CC chemokines. In murine cytomegalovirus (MCMV), the CC chemokine is encoded by the m131/m129 reading frames. The MCMV CC chemokine is called MCK2 and forms a trimeric gHgLMCK2 entry complex. Here, we have generated MCK2 mutant viruses either unable to form gHgLMCK2 complexes, lacking the chemokine function or lacking both functions. By using these viruses, we could demonstrate that gHgLMCK2-dependent entry and MCK2 chemokine activity are independent functions of MCK2 in vitro and in vivo. The gHgLMCK2 complex promotes the tropism for leukocytes like macrophages and dendritic cells and secures high titers in salivary glands in MCMV-infected mice independent of the chemokine activity of MCK2. In contrast, reduced early antiviral T cell responses in MCMV-infected mice are dependent on MCK2 being an active chemokine and do not require the formation of gHgLMCK2 complexes. High levels of CCL2 and IFN-γ in spleens of infected mice and MCMV virulence depend on both, the formation of gHgLMCK2 complexes and the MCK2 chemokine activity. Thus, independent and concerted functions of MCK2 serving as chemokine and part of a gHgL entry complex shape antiviral immunity and virus dissemination.
Collapse
Affiliation(s)
- Marwa Eletreby
- Max von Pettenkofer Institute & Gene Center, Virology, Faculty of Medicine, Ludwig- Maximilians-University Munich, Munich, Germany
| | - Lena Thiessen
- Max von Pettenkofer Institute & Gene Center, Virology, Faculty of Medicine, Ludwig- Maximilians-University Munich, Munich, Germany
| | - Adrian Prager
- Max von Pettenkofer Institute & Gene Center, Virology, Faculty of Medicine, Ludwig- Maximilians-University Munich, Munich, Germany
| | - Ilija Brizic
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Jelena Materljan
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Lucie Kubic
- Max von Pettenkofer Institute & Gene Center, Virology, Faculty of Medicine, Ludwig- Maximilians-University Munich, Munich, Germany
| | - Katharina Jäger
- Max von Pettenkofer Institute & Gene Center, Virology, Faculty of Medicine, Ludwig- Maximilians-University Munich, Munich, Germany
| | - Križan Jurinović
- Max von Pettenkofer Institute & Gene Center, Virology, Faculty of Medicine, Ludwig- Maximilians-University Munich, Munich, Germany
| | - Josipa Jerak
- Max von Pettenkofer Institute & Gene Center, Virology, Faculty of Medicine, Ludwig- Maximilians-University Munich, Munich, Germany
| | - Karsten Krey
- Max von Pettenkofer Institute & Gene Center, Virology, Faculty of Medicine, Ludwig- Maximilians-University Munich, Munich, Germany
| | - Barbara Adler
- Max von Pettenkofer Institute & Gene Center, Virology, Faculty of Medicine, Ludwig- Maximilians-University Munich, Munich, Germany
| |
Collapse
|
2
|
Bošnjak B, Henze E, Lueder Y, Do KTH, Rezalotfi A, Čuvalo B, Ritter C, Schimrock A, Willenzon S, Georgiev H, Fritz L, Galla M, Wagner K, Messerle M, Förster R. MCK2-mediated MCMV infection of macrophages and virus dissemination to the salivary gland depends on MHC class I molecules. Cell Rep 2023; 42:112597. [PMID: 37289588 DOI: 10.1016/j.celrep.2023.112597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 03/14/2023] [Accepted: 05/18/2023] [Indexed: 06/10/2023] Open
Abstract
Murine cytomegalovirus (MCMV) infection of macrophages relies on MCMV-encoded chemokine 2 (MCK2), while infection of fibroblasts occurs independently of MCK2. Recently, MCMV infection of both cell types was found to be dependent on cell-expressed neuropilin 1. Using a CRISPR screen, we now identify that MCK2-dependent infection requires MHC class Ia/β-2-microglobulin (B2m) expression. Further analyses reveal that macrophages expressing MHC class Ia haplotypes H-2b and H-2d, but not H-2k, are susceptible to MCK2-dependent infection with MCMV. The importance of MHC class I expression for MCK2-dependent primary infection and viral dissemination is highlighted by experiments with B2m-deficient mice, which lack surface expression of MHC class I molecules. In those mice, intranasally administered MCK2-proficient MCMV mimics infection patterns of MCK2-deficient MCMV in wild-type mice: it does not infect alveolar macrophages and subsequently fails to disseminate into the salivary glands. Together, these data provide essential knowledge for understanding MCMV-induced pathogenesis, tissue targeting, and virus dissemination.
Collapse
Affiliation(s)
- Berislav Bošnjak
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany.
| | - Elisa Henze
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Yvonne Lueder
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Kim Thi Hoang Do
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Alaleh Rezalotfi
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Berislav Čuvalo
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Christiane Ritter
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Anja Schimrock
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Stefanie Willenzon
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Hristo Georgiev
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Lea Fritz
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Melanie Galla
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Karen Wagner
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Martin Messerle
- Institute of Virology, Hannover Medical School, Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625 Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625 Hannover, Germany; German Centre for Infection Research (DZIF), Partner site Hannover, Hannover, Germany.
| |
Collapse
|
3
|
Bruce K, Ma J, Lawler C, Xie W, Stevenson PG, Farrell HE. Recent Advancements in Understanding Primary Cytomegalovirus Infection in a Mouse Model. Viruses 2022; 14:v14091934. [PMID: 36146741 PMCID: PMC9505653 DOI: 10.3390/v14091934] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/17/2022] Open
Abstract
Animal models that mimic human infections provide insights in virus–host interplay; knowledge that in vitro approaches cannot readily predict, nor easily reproduce. Human cytomegalovirus (HCMV) infections are acquired asymptomatically, and primary infections are difficult to capture. The gap in our knowledge of the early events of HCMV colonization and spread limits rational design of HCMV antivirals and vaccines. Studies of natural infection with mouse cytomegalovirus (MCMV) have demonstrated the olfactory epithelium as the site of natural colonization. Systemic spread from the olfactory epithelium is facilitated by infected dendritic cells (DC); tracking dissemination uncovered previously unappreciated DC trafficking pathways. The olfactory epithelium also provides a unique niche that supports efficient MCMV superinfection and virus recombination. In this review, we summarize recent advances to our understanding of MCMV infection and spread and the tissue-specific mechanisms utilized by MCMV to modulate DC trafficking. As these mechanisms are likely conserved with HCMV, they may inform new approaches for preventing HCMV infections in humans.
Collapse
|
4
|
Hancock TJ, Hetzel ML, Ramirez A, Sparer TE. MCMV Centrifugal Enhancement: A New Spin on an Old Topic. Pathogens 2021; 10:1577. [PMID: 34959531 PMCID: PMC8705575 DOI: 10.3390/pathogens10121577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/02/2021] [Accepted: 12/02/2021] [Indexed: 11/17/2022] Open
Abstract
Human cytomegalovirus (HCMV) is a ubiquitous pathogen infecting a majority of people worldwide, with diseases ranging from mild to life-threatening. Its clinical relevance in immunocompromised people and congenital infections have made treatment and vaccine development a top priority. Because of cytomegaloviruses' species specificity, murine cytomegalovirus (MCMV) models have historically informed and advanced translational CMV therapies. Using the phenomenon of centrifugal enhancement, we explored differences between MCMVs derived in vitro and in vivo. We found centrifugal enhancement on tissue culture-derived virus (TCV) was ~3× greater compared with salivary gland derived virus (SGV). Using novel "flow virometry", we found that TCV contained a distinct submicron particle composition compared to SGV. Using an inhibitor of exosome production, we show these submicron particles are not extracellular vesicles that contribute to centrifugal enhancement. We examined how these differences in submicron particles potentially contribute to differing centrifugal enhancement phenotypes, as well as broader in vivo vs. in vitro MCMV differences.
Collapse
Affiliation(s)
| | | | | | - Tim E. Sparer
- Department of Microbiology, University of Tennessee, Knoxville, TN 37996, USA; (T.J.H.); (M.L.H.); (A.R.)
| |
Collapse
|
5
|
Mandal P, Nagrani LN, Hernandez L, McCormick AL, Dillon CP, Koehler HS, Roback L, Alnemri ES, Green DR, Mocarski ES. Multiple Autonomous Cell Death Suppression Strategies Ensure Cytomegalovirus Fitness. Viruses 2021; 13:v13091707. [PMID: 34578288 PMCID: PMC8473406 DOI: 10.3390/v13091707] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/13/2021] [Accepted: 08/20/2021] [Indexed: 12/31/2022] Open
Abstract
Programmed cell death pathways eliminate infected cells and regulate infection-associated inflammation during pathogen invasion. Cytomegaloviruses encode several distinct suppressors that block intrinsic apoptosis, extrinsic apoptosis, and necroptosis, pathways that impact pathogenesis of this ubiquitous herpesvirus. Here, we expanded the understanding of three cell autonomous suppression mechanisms on which murine cytomegalovirus relies: (i) M38.5-encoded viral mitochondrial inhibitor of apoptosis (vMIA), a BAX suppressor that functions in concert with M41.1-encoded viral inhibitor of BAK oligomerization (vIBO), (ii) M36-encoded viral inhibitor of caspase-8 activation (vICA), and (iii) M45-encoded viral inhibitor of RIP/RHIM activation (vIRA). Following infection of bone marrow-derived macrophages, the virus initially deflected receptor-interacting protein kinase (RIPK)3-dependent necroptosis, the most potent of the three cell death pathways. This process remained independent of caspase-8, although suppression of this apoptotic protease enhances necroptosis in most cell types. Second, the virus deflected TNF-mediated extrinsic apoptosis, a pathway dependent on autocrine TNF production by macrophages that proceeds independently of mitochondrial death machinery or RIPK3. Third, cytomegalovirus deflected BCL-2 family protein-dependent mitochondrial cell death through combined TNF-dependent and -independent signaling even in the absence of RIPK1, RIPK3, and caspase-8. Furthermore, each of these cell death pathways dictated a distinct pattern of cytokine and chemokine activation. Therefore, cytomegalovirus employs sequential, non-redundant suppression strategies to specifically modulate the timing and execution of necroptosis, extrinsic apoptosis, and intrinsic apoptosis within infected cells to orchestrate virus control and infection-dependent inflammation. Virus-encoded death suppressors together hold control over an intricate network that upends host defense and supports pathogenesis in the intact mammalian host.
Collapse
Affiliation(s)
- Pratyusha Mandal
- Emory Vaccine Center, Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA; (L.H.); (H.S.K.); (L.R.)
- Correspondence: (P.M.); (E.S.M.); Tel.: +404-727-0563 (P.M.); +404-727-4273 (E.S.M.)
| | | | - Liliana Hernandez
- Emory Vaccine Center, Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA; (L.H.); (H.S.K.); (L.R.)
| | | | | | - Heather S. Koehler
- Emory Vaccine Center, Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA; (L.H.); (H.S.K.); (L.R.)
| | - Linda Roback
- Emory Vaccine Center, Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA; (L.H.); (H.S.K.); (L.R.)
| | - Emad S. Alnemri
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| | - Douglas R. Green
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA;
| | - Edward S. Mocarski
- Emory Vaccine Center, Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA; (L.H.); (H.S.K.); (L.R.)
- Correspondence: (P.M.); (E.S.M.); Tel.: +404-727-0563 (P.M.); +404-727-4273 (E.S.M.)
| |
Collapse
|
6
|
Zhang S, Springer LE, Rao HZ, Espinosa Trethewy RG, Bishop LM, Hancock MH, Grey F, Snyder CM. Hematopoietic cell-mediated dissemination of murine cytomegalovirus is regulated by NK cells and immune evasion. PLoS Pathog 2021; 17:e1009255. [PMID: 33508041 PMCID: PMC7872266 DOI: 10.1371/journal.ppat.1009255] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 02/09/2021] [Accepted: 12/21/2020] [Indexed: 02/06/2023] Open
Abstract
Cytomegalovirus (CMV) causes clinically important diseases in immune compromised and immune immature individuals. Based largely on work in the mouse model of murine (M)CMV, there is a consensus that myeloid cells are important for disseminating CMV from the site of infection. In theory, such dissemination should expose CMV to cell-mediated immunity and thus necessitate evasion of T cells and NK cells. However, this hypothesis remains untested. We constructed a recombinant MCMV encoding target sites for the hematopoietic specific miRNA miR-142-3p in the essential viral gene IE3. This virus disseminated poorly to the salivary gland following intranasal or footpad infections but not following intraperitoneal infection in C57BL/6 mice, demonstrating that dissemination by hematopoietic cells is essential for specific routes of infection. Remarkably, depletion of NK cells or T cells restored dissemination of this virus in C57BL/6 mice after intranasal infection, while dissemination occurred normally in BALB/c mice, which lack strong NK cell control of MCMV. These data show that cell-mediated immunity is responsible for restricting MCMV to hematopoietic cell-mediated dissemination. Infected hematopoietic cells avoided cell-mediated immunity via three immune evasion genes that modulate class I MHC and NKG2D ligands (m04, m06 and m152). MCMV lacking these 3 genes spread poorly to the salivary gland unless NK cells were depleted, but also failed to replicate persistently in either the nasal mucosa or salivary gland unless CD8+ T cells were depleted. Surprisingly, CD8+ T cells primed after intranasal infection required CD4+ T cell help to expand and become functional. Together, our data suggest that MCMV can use both hematopoietic cell-dependent and -independent means of dissemination after intranasal infection and that cell mediated immune responses restrict dissemination to infected hematopoietic cells, which are protected from NK cells during dissemination by viral immune evasion. In contrast, viral replication within mucosal tissues depends on evasion of T cells. Cytomegalovirus (CMV) is a common cause of disease in immune compromised individuals as well as a common cause of congenital infections leading to disease in newborns. The virus is thought to enter primarily via mucosal barrier tissues, such as the oral and nasal mucosa. However, it is not clear how the virus escapes these barrier tissues to reach distant sites. In this study, we used a mouse model of CMV infection. Our data illustrate a complex balance between the immune system and viral infection of “myeloid cells”, which are most commonly thought to carry the virus around the body after infection. In particular, our data suggest that robust immune responses at the site of infection force the virus to rely on myeloid cells to escape the site of infection. Moreover, viral genes designed to evade these immune responses were needed to protect the virus during and after its spread to distant sites. Together, this work sheds light on the mechanisms of immune control and viral survival during CMV infection of mucosal tissues and spread to distant sites of the body.
Collapse
Affiliation(s)
- Shunchuan Zhang
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Lauren E. Springer
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Han-Zhi Rao
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Renee G. Espinosa Trethewy
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Lindsey M. Bishop
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Meaghan H. Hancock
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Finn Grey
- Division of Infection and Immunity, The Roslin Institute, University of Edinburgh, Easter Bush, Midlothian, United Kingdom
- * E-mail: (FG); (CMS)
| | - Christopher M. Snyder
- Department of Microbiology and Immunology, Sidney Kimmel Medical College, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
- * E-mail: (FG); (CMS)
| |
Collapse
|
7
|
Jones IKA, Haese NN, Gatault P, Streblow ZJ, Andoh TF, Denton M, Streblow CE, Bonin K, Kreklywich CN, Burg JM, Orloff SL, Streblow DN. Rat Cytomegalovirus Virion-Associated Proteins R131 and R129 Are Necessary for Infection of Macrophages and Dendritic Cells. Pathogens 2020; 9:E963. [PMID: 33228102 PMCID: PMC7699341 DOI: 10.3390/pathogens9110963] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/05/2020] [Accepted: 11/17/2020] [Indexed: 12/15/2022] Open
Abstract
Cytomegalovirus (CMV) establishes persistent, latent infection in hosts, causing diseases in immunocompromised patients, transplant recipients, and neonates. CMV infection modifies the host chemokine axis by modulating chemokine and chemokine receptor expression and by encoding putative chemokine and chemokine receptor homologues. The viral proteins have roles in cellular signaling, migration, and transformation, as well as viral dissemination, tropism, latency and reactivation. Herein, we review the contribution of CMV-encoded chemokines and chemokine receptors to these processes, and further elucidate the viral tropism role of rat CMV (RCMV) R129 and R131. These homologues of the human CMV (HCMV)-encoded chemokines UL128 and UL130 are of particular interest because of their dual role as chemokines and members of the pentameric entry complex, which is required for entry into cell types that are essential for viral transmission and dissemination. The contributions of UL128 and UL130 to acceleration of solid organ transplant chronic rejection are poorly understood, and are in need of an effective in vivo model system to elucidate the phenomenon. We demonstrated similar molecular entry requirements for R129 and R131 in the rat cells, as observed for HCMV, and provided evidence that R129 and R131 are part of the viral entry complex required for entry into macrophages, dendritic cells, and bone marrow cells.
Collapse
Affiliation(s)
- Iris K. A. Jones
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Portland, OR 97239, USA; (I.K.A.J.); (N.N.H.); (Z.J.S.); (T.F.A.); (M.D.); (C.E.S.); (K.B.); (C.N.K.)
| | - Nicole N. Haese
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Portland, OR 97239, USA; (I.K.A.J.); (N.N.H.); (Z.J.S.); (T.F.A.); (M.D.); (C.E.S.); (K.B.); (C.N.K.)
| | - Philippe Gatault
- Renal Transplant Unit, 10 Boulevard Tonnellé, University Hospital of Tours, 37032 Tours, France;
| | - Zachary J. Streblow
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Portland, OR 97239, USA; (I.K.A.J.); (N.N.H.); (Z.J.S.); (T.F.A.); (M.D.); (C.E.S.); (K.B.); (C.N.K.)
| | - Takeshi F. Andoh
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Portland, OR 97239, USA; (I.K.A.J.); (N.N.H.); (Z.J.S.); (T.F.A.); (M.D.); (C.E.S.); (K.B.); (C.N.K.)
- Department of Surgery, Oregon Health & Science University, Portland, OR 97239, USA; (J.M.B.); (S.L.O.)
| | - Michael Denton
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Portland, OR 97239, USA; (I.K.A.J.); (N.N.H.); (Z.J.S.); (T.F.A.); (M.D.); (C.E.S.); (K.B.); (C.N.K.)
| | - Cassilyn E. Streblow
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Portland, OR 97239, USA; (I.K.A.J.); (N.N.H.); (Z.J.S.); (T.F.A.); (M.D.); (C.E.S.); (K.B.); (C.N.K.)
| | - Kiley Bonin
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Portland, OR 97239, USA; (I.K.A.J.); (N.N.H.); (Z.J.S.); (T.F.A.); (M.D.); (C.E.S.); (K.B.); (C.N.K.)
| | - Craig N. Kreklywich
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Portland, OR 97239, USA; (I.K.A.J.); (N.N.H.); (Z.J.S.); (T.F.A.); (M.D.); (C.E.S.); (K.B.); (C.N.K.)
| | - Jennifer M. Burg
- Department of Surgery, Oregon Health & Science University, Portland, OR 97239, USA; (J.M.B.); (S.L.O.)
| | - Susan L. Orloff
- Department of Surgery, Oregon Health & Science University, Portland, OR 97239, USA; (J.M.B.); (S.L.O.)
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Daniel N. Streblow
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Portland, OR 97239, USA; (I.K.A.J.); (N.N.H.); (Z.J.S.); (T.F.A.); (M.D.); (C.E.S.); (K.B.); (C.N.K.)
| |
Collapse
|
8
|
Repair of an Attenuated Low-Passage Murine Cytomegalovirus Bacterial Artificial Chromosome Identifies a Novel Spliced Gene Essential for Salivary Gland Tropism. J Virol 2020; 94:JVI.01456-20. [PMID: 32847854 DOI: 10.1128/jvi.01456-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 08/23/2020] [Indexed: 01/22/2023] Open
Abstract
The cloning of herpesviruses as bacterial artificial chromosomes (BACs) has revolutionized the study of herpesvirus biology, allowing rapid and precise manipulation of viral genomes. Several clinical strains of human cytomegalovirus (HCMV) have been cloned as BACs; however, no low-passage strains of murine CMV (MCMV), which provide a model mimicking these isolates, have been cloned. Here, the low-passage G4 strain of was BAC cloned. G4 carries an m157 gene that does not ligate the natural killer (NK) cell-activating receptor, Ly49H, meaning that unlike laboratory strains of MCMV, this virus replicates well in C57BL/6 mice. This BAC clone exhibited normal replication during acute infection in the spleen and liver but was attenuated for salivary gland tropism. Next-generation sequencing revealed a C-to-A mutation at nucleotide position 188422, located in the 3' untranslated region of sgg1, a spliced gene critical for salivary gland tropism. Repair of this mutation restored tropism for the salivary glands. Transcriptional analysis revealed a novel spliced gene within the sgg1 locus. This small open reading frame (ORF), sgg1.1, starts at the 3' end of the first exon of sgg1 and extends exon 2 of sgg1. This shorter spliced gene is prematurely terminated by the nonsense mutation at nt 188422. Sequence analysis of tissue culture-passaged virus demonstrated that sgg1.1 was stable, although other mutational hot spots were identified. The G4 BAC will allow in vivo studies in a broader range of mice, avoiding the strong NK cell responses seen in B6 mice with other MCMV BAC-derived MCMVs.IMPORTANCE Murine cytomegalovirus (MCMV) is widely used as a model of human CMV (HCMV) infection. However, this model relies on strains of MCMV that have been serially passaged in the laboratory for over four decades. These laboratory strains have been cloned as bacterial artificial chromosomes (BACs), which permits rapid and precise manipulation. Low-passage strains of MCMV add to the utility of the mouse model of HCMV infection but do not exist as cloned BACs. This study describes the first such low-passage MCMV BAC. This BAC-derived G4 was initially attenuated in vivo, with subsequent full genomic sequencing revealing a novel spliced transcript required for salivary gland tropism. These data suggest that MCMV, like HCMV, undergoes tissue culture adaptation that can limit in vivo growth and supports the use of BAC clones as a way of standardizing viral strains and minimizing interlaboratory strain variation.
Collapse
|
9
|
Milovanovic J, Arsenijevic A, Stojanovic B, Kanjevac T, Arsenijevic D, Radosavljevic G, Milovanovic M, Arsenijevic N. Interleukin-17 in Chronic Inflammatory Neurological Diseases. Front Immunol 2020; 11:947. [PMID: 32582147 PMCID: PMC7283538 DOI: 10.3389/fimmu.2020.00947] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/22/2020] [Indexed: 12/15/2022] Open
Abstract
A critical role for IL-17, a cytokine produced by T helper 17 (Th17) cells, has been indicated in the pathogenesis of chronic inflammatory and autoimmune diseases. A positive effect of blockade of IL-17 secreted by autoreactive T cells has been shown in various inflammatory diseases. Several cytokines, whose production is affected by environmental factors, control Th17 differentiation and its maintenance in tissues during chronic inflammation. The roles of IL-17 in the pathogenesis of chronic neuroinflammatory conditions, multiple sclerosis (MS), experimental autoimmune encephalomyelitis (EAE), Alzheimer's disease, and ischemic brain injury are reviewed here. The role of environmental stimuli in Th17 differentiation is also summarized, highlighting the role of viral infection in the regulation of pathogenic T helper cells in EAE.
Collapse
Affiliation(s)
- Jelena Milovanovic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
- Department of Histology and Embriology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Aleksandar Arsenijevic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
| | - Bojana Stojanovic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
- Department of Pathophysiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Tatjana Kanjevac
- Department of Dentistry, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Dragana Arsenijevic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Gordana Radosavljevic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
| | - Marija Milovanovic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
| | - Nebojsa Arsenijevic
- Faculty of Medical Sciences, Center for Molecular Medicine and Stem Cell Research, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
10
|
Where do we Stand after Decades of Studying Human Cytomegalovirus? Microorganisms 2020; 8:microorganisms8050685. [PMID: 32397070 PMCID: PMC7284540 DOI: 10.3390/microorganisms8050685] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 04/27/2020] [Accepted: 05/05/2020] [Indexed: 12/26/2022] Open
Abstract
Human cytomegalovirus (HCMV), a linear double-stranded DNA betaherpesvirus belonging to the family of Herpesviridae, is characterized by widespread seroprevalence, ranging between 56% and 94%, strictly dependent on the socioeconomic background of the country being considered. Typically, HCMV causes asymptomatic infection in the immunocompetent population, while in immunocompromised individuals or when transmitted vertically from the mother to the fetus it leads to systemic disease with severe complications and high mortality rate. Following primary infection, HCMV establishes a state of latency primarily in myeloid cells, from which it can be reactivated by various inflammatory stimuli. Several studies have shown that HCMV, despite being a DNA virus, is highly prone to genetic variability that strongly influences its replication and dissemination rates as well as cellular tropism. In this scenario, the few currently available drugs for the treatment of HCMV infections are characterized by high toxicity, poor oral bioavailability, and emerging resistance. Here, we review past and current literature that has greatly advanced our understanding of the biology and genetics of HCMV, stressing the urgent need for innovative and safe anti-HCMV therapies and effective vaccines to treat and prevent HCMV infections, particularly in vulnerable populations.
Collapse
|
11
|
Wilski NA, Stotesbury C, Del Casale C, Montoya B, Wong E, Sigal LJ, Snyder CM. STING Sensing of Murine Cytomegalovirus Alters the Tumor Microenvironment to Promote Antitumor Immunity. THE JOURNAL OF IMMUNOLOGY 2020; 204:2961-2972. [PMID: 32284333 DOI: 10.4049/jimmunol.1901136] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 03/18/2020] [Indexed: 01/04/2023]
Abstract
CMV has been proposed to play a role in cancer progression and invasiveness. However, CMV has been increasingly studied as a cancer vaccine vector, and multiple groups, including ours, have reported that the virus can drive antitumor immunity in certain models. Our previous work revealed that intratumoral injections of wild-type murine CMV (MCMV) into B16-F0 melanomas caused tumor growth delay in part by using a viral chemokine to recruit macrophages that were subsequently infected. We now show that MCMV acts as a STING agonist in the tumor. MCMV infection of tumors in STING-deficient mice resulted in normal recruitment of macrophages to the tumor, but poor recruitment of CD8+ T cells, reduced production of inflammatory cytokines and chemokines, and no delay in tumor growth. In vitro, expression of type I IFN was dependent on both STING and the type I IFNR. Moreover, type I IFN alone was sufficient to induce cytokine and chemokine production by macrophages and B16 tumor cells, suggesting that the major role for STING activation was to produce type I IFN. Critically, viral infection of wild-type macrophages alone was sufficient to restore tumor growth delay in STING-deficient animals. Overall, these data show that MCMV infection and sensing in tumor-associated macrophages through STING signaling is sufficient to promote antitumor immune responses in the B16-F0 melanoma model.
Collapse
Affiliation(s)
- Nicole A Wilski
- Department of Microbiology and Immunology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Colby Stotesbury
- Department of Microbiology and Immunology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Christina Del Casale
- Department of Microbiology and Immunology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Brian Montoya
- Department of Microbiology and Immunology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Eric Wong
- Department of Microbiology and Immunology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Luis J Sigal
- Department of Microbiology and Immunology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Christopher M Snyder
- Department of Microbiology and Immunology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| |
Collapse
|
12
|
Murine Cytomegalovirus Infection of Melanoma Lesions Delays Tumor Growth by Recruiting and Repolarizing Monocytic Phagocytes in the Tumor. J Virol 2019; 93:JVI.00533-19. [PMID: 31375579 DOI: 10.1128/jvi.00533-19] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 07/18/2019] [Indexed: 02/06/2023] Open
Abstract
Cytomegalovirus (CMV) is a ubiquitous betaherpesvirus that infects many different cell types. Human CMV (HCMV) has been found in several solid tumors, and it has been hypothesized that it may promote cellular transformation or exacerbate tumor growth. Paradoxically, in some experimental situations, murine CMV (MCMV) infection delays tumor growth. We previously showed that wild-type MCMV delayed the growth of poorly immunogenic B16 melanomas via an undefined mechanism. Here, we show that MCMV delayed the growth of these immunologically "cold" tumors by recruiting and modulating tumor-associated macrophages. Depletion of monocytic phagocytes with clodronate completely prevented MCMV from delaying tumor growth. Mechanistically, our data suggest that MCMV recruits new macrophages to the tumor via the virus-encoded chemokine MCK2, and viruses lacking this chemokine were unable to delay tumor growth. Moreover, MCMV infection of macrophages drove them toward a proinflammatory (M1)-like state. Importantly, adaptive immune responses were also necessary for MCMV to delay tumor growth as the effect was substantially blunted in Rag-deficient animals. However, viral spread was not needed and a spread-defective MCMV strain was equally effective. In most mice, the antitumor effect of MCMV was transient. Although the recruited macrophages persisted, tumor regrowth correlated with a loss of viral activity in the tumor. However, an additional round of MCMV infection further delayed tumor growth, suggesting that tumor growth delay was dependent on active viral infection. Together, our results suggest that MCMV infection delayed the growth of an immunologically cold tumor by recruiting and modulating macrophages in order to promote anti-tumor immune responses.IMPORTANCE Cytomegalovirus (CMV) is an exciting new platform for vaccines and cancer therapy. Although CMV may delay tumor growth in some settings, there is also evidence that CMV may promote cancer development and progression. Thus, defining the impact of CMV on tumors is critical. Using a mouse model of melanoma, we previously found that murine CMV (MCMV) delayed tumor growth and activated tumor-specific immunity although the mechanism was unclear. We now show that MCMV delayed tumor growth through a mechanism that required monocytic phagocytes and a viral chemokine that recruited macrophages to the tumor. Furthermore, MCMV infection altered the functional state of macrophages. Although the effects of MCMV on tumor growth were transient, we found that repeated MCMV injections sustained the antitumor effect, suggesting that active viral infection was needed. Thus, MCMV altered tumor growth by actively recruiting macrophages to the tumor, where they were modulated to promote antitumor immunity.
Collapse
|
13
|
Murine cytomegalovirus infection in mice results in an acute inflammatory reaction in peripheral nerves. J Neuroimmunol 2019; 335:577017. [PMID: 31430710 DOI: 10.1016/j.jneuroim.2019.577017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 07/31/2019] [Accepted: 07/31/2019] [Indexed: 02/08/2023]
Abstract
Human cytomegalovirus (CMV) infection is asymptomatic in immunocompetent individuals. However, it can lead to disease in immunodeficient population. Little is known of the mechanisms underlying the pathogenicity of the virus. We investigated the impact of CMV infection on mouse nervous system. Peripheral nerves but not spinal cord was permissive to MCMV during acute infection. Activated CD8+ T cells, monocytes/macrophages and cytokine expression were increased in the blood and sciatic nerves of infected mice, which exhibited transient sensory dysfunction. This study indicates that systemic MCMV infection leads to a dissemination of MCMV into peripheral nerves, which is associated with a local inflammation but not nerve tissue damage in the acute phase.
Collapse
|
14
|
Wilski NA, Snyder CM. From Vaccine Vector to Oncomodulation: Understanding the Complex Interplay between CMV and Cancer. Vaccines (Basel) 2019; 7:E62. [PMID: 31323930 PMCID: PMC6789822 DOI: 10.3390/vaccines7030062] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/02/2019] [Accepted: 07/04/2019] [Indexed: 12/14/2022] Open
Abstract
Cytomegalovirus (CMV) is a herpesvirus that establishes a persistent, but generally asymptomatic, infection in most people in the world. However, CMV drives and sustains extremely large numbers of antigen-specific T cells and is, therefore, emerging as an exciting platform for vaccines against infectious diseases and cancer. Indeed, pre-clinical data strongly suggest that CMV-based vaccines can sustain protective CD8+ T cell and antibody responses. In the context of vaccines for infectious diseases, substantial pre-clinical studies have elucidated the efficacy and protective mechanisms of CMV-based vaccines, including in non-human primate models of various infections. In the context of cancer vaccines, however, much less is known and only very early studies in mice have been conducted. To develop CMV-based cancer vaccines further, it will be critical to better understand the complex interaction of CMV and cancer. An array of evidence suggests that naturally-acquired human (H)CMV can be detected in cancers, and it has been proposed that HCMV may promote tumor growth. This would obviously be a concern for any therapeutic cancer vaccines. In experimental models, CMV has been shown to play both positive and negative roles in tumor progression, depending on the model studied. However, the mechanisms are still largely unknown. Thus, more studies assessing the interaction of CMV with the tumor microenvironment are needed. This review will summarize the existing literature and major open questions about CMV-based vaccines for cancer, and discuss our hypothesis that the balance between pro-tumor and anti-tumor effects driven by CMV depends on the location and the activity of the virus in the lesion.
Collapse
Affiliation(s)
- Nicole A Wilski
- Department of Microbiology and Immunology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Christopher M Snyder
- Department of Microbiology and Immunology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
15
|
The Human Cytomegalovirus Chemokine vCXCL-1 Modulates Normal Dissemination Kinetics of Murine Cytomegalovirus In Vivo. mBio 2019; 10:mBio.01289-19. [PMID: 31239384 PMCID: PMC6593410 DOI: 10.1128/mbio.01289-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
An adequate in vivo analysis of HCMV’s viral chemokine vCXCL-1 has been lacking. Here we generate recombinant MCMVs expressing vCXCL-1 to study vCXCL-1 function in vivo using MCMV as a surrogate. We demonstrate that vCXCL-1 increases MCMV dissemination kinetics for both primary and secondary dissemination. Additionally, we provide evidence, that the murine neutrophil is largely a bystander in the mouse’s response to vCXCL-1. We confirm the hypothesis that vCXCL-1 is a HCMV virulence factor. Infection of severely immunocompromised mice with MCMVs expressing vCXCL-1 was lethal in more than 50% of infected animals, while all animals infected with parental virus survived during a 12-day period. This work provides needed insights into vCXCL-1 function in vivo. Human cytomegalovirus (HCMV) is a betaherpesvirus that is a significant pathogen within newborn and immunocompromised populations. Morbidity associated with HCMV infection is the consequence of viral dissemination. HCMV has evolved to manipulate the host immune system to enhance viral dissemination and ensure long-term survival within the host. The immunomodulatory protein vCXCL-1, a viral chemokine functioning primarily through the CXCR2 chemokine receptor, is hypothesized to attract CXCR2+ neutrophils to infection sites, aiding viral dissemination. Neutrophils harbor HCMV in vivo; however, the interaction between vCXCL-1 and the neutrophil has not been evaluated in vivo. Using the mouse model and mouse cytomegalovirus (MCMV) infection, we show that murine neutrophils harbor and transfer infectious MCMV and that virus replication initiates within this cell type. Utilizing recombinant MCMVs expressing vCXCL-1 from the HCMV strain (Toledo), we demonstrated that vCXCL-1 significantly enhances MCMV dissemination kinetics. Through cellular depletion experiments, we observe that neutrophils impact dissemination but that overall dissemination is largely neutrophil independent. This work adds neutrophils to the list of innate cells (i.e., dendritic and macrophages/monocytes) that contribute to MCMV dissemination but refutes the hypothesis that neutrophils are the primary cell responding to vCXCL-1.
Collapse
|
16
|
Zhang S, Caldeira-Dantas S, Smith CJ, Snyder CM. Persistent viral replication and the development of T-cell responses after intranasal infection by MCMV. Med Microbiol Immunol 2019; 208:457-468. [PMID: 30848361 DOI: 10.1007/s00430-019-00589-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 03/01/2019] [Indexed: 12/15/2022]
Abstract
Natural transmission of cytomegalovirus (CMV) has been difficult to observe. However, recent work using the mouse model of murine (M)CMV demonstrated that MCMV initially infects the nasal mucosa after transmission from mothers to pups. We found that intranasal (i.n.) inoculation of C57BL/6J mice resulted in reliable recovery of replicating virus from the nasal mucosa as assessed by plaque assay. After i.n. inoculation, CD8+ T-cell priming occurred in the mandibular, deep-cervical, and mediastinal lymph nodes within 3 days of infection. Although i.n. infection induced "memory inflation" of T cells specific for the M38316-323 epitope, there were no detectable CD8+ T-cell responses against the late-appearing IE3416-423 epitope, which contrasts with intraperitoneal (i.p.) infection. MCMV-specific T cells migrated into the nasal mucosa where they developed a tissue-resident memory (TRM) phenotype and this could occur independently of local virus infection or antigen. Strikingly however, virus replication was poorly controlled in the nasal mucosa and MCMV was detectable by plaque assay for at least 4 months after primary infection, making the nasal mucosa a second site for MCMV persistence. Unlike in the salivary glands, the persistence of MCMV in the nasal mucosa was not modulated by IL-10. Taken together, our data characterize the development of local and systemic T-cell responses after intranasal infection by MCMV and define the nasal mucosa, a natural site of viral entry, as a novel site of viral persistence.
Collapse
Affiliation(s)
- Shunchuan Zhang
- Department of Microbiology and Immunology, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College, Thomas Jefferson University, 19107, Philadelphia, PA, USA
| | - Sofia Caldeira-Dantas
- Department of Microbiology and Immunology, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College, Thomas Jefferson University, 19107, Philadelphia, PA, USA.,Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,PT Government Associate Laboratory, ICVS/3B's, Braga/Guimarães, Portugal
| | - Corinne J Smith
- Department of Microbiology and Immunology, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College, Thomas Jefferson University, 19107, Philadelphia, PA, USA
| | - Christopher M Snyder
- Department of Microbiology and Immunology, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College, Thomas Jefferson University, 19107, Philadelphia, PA, USA.
| |
Collapse
|
17
|
Murine Cytomegalovirus Glycoprotein O Promotes Epithelial Cell Infection In Vivo. J Virol 2019; 93:JVI.01378-18. [PMID: 30404805 DOI: 10.1128/jvi.01378-18] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 10/29/2018] [Indexed: 02/07/2023] Open
Abstract
Cytomegaloviruses (CMVs) establish systemic infections across diverse cell types. Glycoproteins that alter tropism can potentially guide their spread. Glycoprotein O (gO) is a nonessential fusion complex component of both human CMV (HCMV) and murine CMV (MCMV). We tested its contribution to MCMV spread from the respiratory tract. In vitro, MCMV lacking gO poorly infected fibroblasts and epithelial cells. Cell binding was intact, but penetration was delayed. In contrast, myeloid infection was preserved, and in the lungs, where myeloid and type 2 alveolar epithelial cells are the main viral targets, MCMV lacking gO showed a marked preference for myeloid infection. Its poor epithelial cell infection was associated with poor primary virus production and reduced virulence. Systemic spread, which proceeds via infected CD11c+ myeloid cells, was initially intact but then diminished, because less epithelial infection led ultimately to less myeloid infection. Thus, the tight linkage between peripheral and systemic MCMV infections gave gO-dependent infection a central role in host colonization.IMPORTANCE Human cytomegalovirus is a leading cause of congenital disease. This reflects its capacity for systemic spread. A vaccine is needed, but the best viral targets are unclear. Attention has focused on the virion membrane fusion complex. It has 2 forms, so we need to know what each contributes to host colonization. One includes the virion glycoprotein O. We used murine cytomegalovirus, which has equivalent fusion complexes, to determine the importance of glycoprotein O after mucosal infection. We show that it drives local virus replication in epithelial cells. It was not required to infect myeloid cells, which establish systemic infection, but poor local replication reduced systemic spread as a secondary effect. Therefore, targeting glycoprotein O of human cytomegalovirus has the potential to reduce both local and systemic infections.
Collapse
|
18
|
Reddehase MJ, Lemmermann NAW. Mouse Model of Cytomegalovirus Disease and Immunotherapy in the Immunocompromised Host: Predictions for Medical Translation that Survived the "Test of Time". Viruses 2018; 10:v10120693. [PMID: 30563202 PMCID: PMC6315540 DOI: 10.3390/v10120693] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 11/26/2018] [Accepted: 12/03/2018] [Indexed: 12/14/2022] Open
Abstract
Human Cytomegalovirus (hCMV), which is the prototype member of the β-subfamily of the herpesvirus family, is a pathogen of high clinical relevance in recipients of hematopoietic cell transplantation (HCT). hCMV causes multiple-organ disease and interstitial pneumonia in particular upon infection during the immunocompromised period before hematopoietic reconstitution restores antiviral immunity. Clinical investigation of pathomechanisms and of strategies for an immune intervention aimed at restoring antiviral immunity earlier than by hematopoietic reconstitution are limited in patients to observational studies mainly because of ethical issues including the imperative medical indication for chemotherapy with antivirals. Aimed experimental studies into mechanisms, thus, require animal models that match the human disease as close as possible. Any model for hCMV disease is, however, constrained by the strict host-species specificity of CMVs that prevents the study of hCMV in any animal model including non-human primates. During eons of co-speciation, CMVs each have evolved a set of "private genes" in adaptation to their specific mammalian host including genes that have no homolog in the CMV virus species of any other host species. With a focus on the mouse model of CD8 T cell-based immunotherapy of CMV disease after experimental HCT and infection with murine CMV (mCMV), we review data in support of the phenomenon of "biological convergence" in virus-host adaptation. This includes shared fundamental principles of immune control and immune evasion, which allows us to at least make reasoned predictions from the animal model as an experimental "proof of concept." The aim of a model primarily is to define questions to be addressed by clinical investigation for verification, falsification, or modification and the results can then give feedback to refine the experimental model for research from "bedside to bench".
Collapse
Affiliation(s)
- Matthias J Reddehase
- Institute for Virology, University Medical Center and Center for Immunotherapy of the Johannes Gutenberg-University Mainz, Obere Zahlbacher Str. 67, D-55131 Mainz, Germany.
| | - Niels A W Lemmermann
- Institute for Virology, University Medical Center and Center for Immunotherapy of the Johannes Gutenberg-University Mainz, Obere Zahlbacher Str. 67, D-55131 Mainz, Germany.
| |
Collapse
|
19
|
Jackson JW, Sparer T. There Is Always Another Way! Cytomegalovirus' Multifaceted Dissemination Schemes. Viruses 2018; 10:v10070383. [PMID: 30037007 PMCID: PMC6071125 DOI: 10.3390/v10070383] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 07/17/2018] [Accepted: 07/18/2018] [Indexed: 12/12/2022] Open
Abstract
Human cytomegalovirus (HCMV) is a β-herpes virus that is a significant pathogen within immune compromised populations. HCMV morbidity is induced through viral dissemination and inflammation. Typically, viral dissemination is thought to follow Fenner's hypothesis where virus replicates at the site of infection, followed by replication in the draining lymph nodes, and eventually replicating within blood filtering organs. Although CMVs somewhat follow Fenner's hypothesis, they deviate from it by spreading primarily through innate immune cells as opposed to cell-free virus. Also, in vivo CMVs infect new cells via cell-to-cell spread and disseminate directly to secondary organs through novel mechanisms. We review the historic and recent literature pointing to CMV's direct dissemination to secondary organs and the genes that it has evolved for increasing its ability to disseminate. We also highlight aspects of CMV infection for studying viral dissemination when using in vivo animal models.
Collapse
Affiliation(s)
- Joseph W Jackson
- Department of Microbiology, University of Tennessee Knoxville, Knoxville, TN 37996, USA.
| | - Tim Sparer
- Department of Microbiology, University of Tennessee Knoxville, Knoxville, TN 37996, USA.
| |
Collapse
|
20
|
Pontejo SM, Murphy PM. Chemokines encoded by herpesviruses. J Leukoc Biol 2017; 102:1199-1217. [PMID: 28848041 DOI: 10.1189/jlb.4ru0417-145rr] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 07/25/2017] [Accepted: 07/26/2017] [Indexed: 12/15/2022] Open
Abstract
Viruses use diverse strategies to elude the immune system, including copying and repurposing host cytokine and cytokine receptor genes. For herpesviruses, the chemokine system of chemotactic cytokines and receptors is a common source of copied genes. Here, we review the current state of knowledge about herpesvirus-encoded chemokines and discuss their possible roles in viral pathogenesis, as well as their clinical potential as novel anti-inflammatory agents or targets for new antiviral strategies.
Collapse
Affiliation(s)
- Sergio M Pontejo
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Philip M Murphy
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
21
|
Coleman S, Choi KY, McGregor A. Cytomegalovirus UL128 homolog mutants that form a pentameric complex produce virus with impaired epithelial and trophoblast cell tropism and altered pathogenicity in the guinea pig. Virology 2017. [PMID: 28651121 DOI: 10.1016/j.virol.2017.06.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Guinea pig cytomegalovirus (GPCMV) encodes a homolog pentameric complex (PC) for specific cell tropism and congenital infection. In human cytomegalovirus, the PC is an important antibody neutralizing target and GPCMV studies will aid in the development of intervention strategies. Deletion mutants of the C-terminal domains of unique PC proteins (UL128, UL130 and UL131 homologs) were unable to form a PC in separate transient expression assays. Minor modifications to the UL128 homolog (GP129) C-terminal domain enabled PC formation but viruses encoding these mutants had altered tropism to renal and placental trophoblast cells. Mutation of the presumptive CC chemokine motif encoded by GP129 was investigated by alanine substitution of the CC motif (codons 26-27) and cysteines (codons 47 and 62). GP129 chemokine mutants formed PC but GP129 chemokine mutant viruses had reduced epitropism. A GP129 chemokine mutant virus pathogenicity study demonstrated reduced viral load to target organs but highly extended viremia.
Collapse
Affiliation(s)
- Stewart Coleman
- Department of Microbial Pathogenesis & Immunology, Texas A&M University, Health Science Center, College of Medicine, College Station, TX, United States
| | - K Yeon Choi
- Department of Microbial Pathogenesis & Immunology, Texas A&M University, Health Science Center, College of Medicine, College Station, TX, United States
| | - Alistair McGregor
- Department of Microbial Pathogenesis & Immunology, Texas A&M University, Health Science Center, College of Medicine, College Station, TX, United States.
| |
Collapse
|
22
|
Pontejo SM, Murphy PM. Two glycosaminoglycan-binding domains of the mouse cytomegalovirus-encoded chemokine MCK-2 are critical for oligomerization of the full-length protein. J Biol Chem 2017; 292:9613-9626. [PMID: 28432120 DOI: 10.1074/jbc.m117.785121] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 04/20/2017] [Indexed: 01/22/2023] Open
Abstract
Chemokines are essential for antimicrobial host defenses and tissue repair. Herpesviruses and poxviruses also encode chemokines, copied from their hosts and repurposed for multiple functions, including immune evasion. The CC chemokine MCK-2 encoded by mouse CMV (MCMV) has an atypical structure consisting of a classic chemokine domain N-terminal to a second unique domain, resulting from the splicing of MCMV ORFs m131 and m129 MCK-2 is essential for full MCMV infectivity in macrophages and for persistent infection in the salivary gland. However, information about its mechanism of action and specific biochemical roles for the two domains has been lacking. Here, using genetic, chemical, and enzymatic analyses of multiple mouse cell lines as well as primary mouse fibroblasts from salivary gland and lung, we demonstrate that MCK-2 binds glycosaminoglycans (GAGs) with affinities in the following order: heparin > heparan sulfate > chondroitin sulfate = dermatan sulfate. Both MCK-2 domains bound these GAGs independently, and computational analysis together with site-directed mutagenesis identified five basic residues distributed across the N terminus and the 30s and 50s loops of the chemokine domain that are important GAG binding determinants. Both domains were required for GAG-dependent oligomerization of full-length MCK-2. Thus, MCK-2 is an atypical viral chemokine consisting of a CC chemokine domain and a unique non-chemokine domain, both of which bind GAGs and are critical for GAG-dependent oligomerization of the full-length protein.
Collapse
Affiliation(s)
- Sergio M Pontejo
- From the Laboratory of Molecular Immunology, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| | - Philip M Murphy
- From the Laboratory of Molecular Immunology, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
23
|
Milovanovic J, Popovic B, Milovanovic M, Kvestak D, Arsenijevic A, Stojanovic B, Tanaskovic I, Krmpotic A, Arsenijevic N, Jonjic S, Lukic ML. Murine Cytomegalovirus Infection Induces Susceptibility to EAE in Resistant BALB/c Mice. Front Immunol 2017; 8:192. [PMID: 28289417 PMCID: PMC5326788 DOI: 10.3389/fimmu.2017.00192] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Accepted: 02/09/2017] [Indexed: 12/24/2022] Open
Abstract
In contrast to C57BL/6 mice, BALB/c mice are relatively resistant to the induction of experimental autoimmune encephalomyelitis (EAE) after challenge with MOG35–55 peptide. Here, we provide the first evidence that infection with murine cytomegalovirus (MCMV) in adulthood abrogates this resistance. Infected BALB/c mice developed clinical and histological signs similar to those seen in susceptible C57BL/6 mice. In addition to CD4+ cells, large proportion of cells in the infiltrate of diseased BALB/c mice was CD8+, similar with findings in multiple sclerosis. CD8+ cells that responded to ex vivo restimulation with MOG35–55 were not specific for viral epitopes pp89 and m164. MCMV infection favors proinflammatory type of dendritic cells (CD86+CD40+CD11c+) in the peripheral lymph organs, M1 type of microglia in central nervous system, and increases development of Th1/Th17 encephalitogenic cells. This study indicates that MCMV may enhance autoimmune neuropathology and abrogate inherent resistance to EAE in mouse strain by enhancing proinflammatory phenotype of antigen-presenting cells, Th1/Th17, and CD8 response to MOG35–55.
Collapse
Affiliation(s)
- Jelena Milovanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia; Faculty of Medical Sciences, Institute of Histology, University of Kragujevac, Kragujevac, Serbia
| | - Branka Popovic
- Center for Proteomics, Faculty of Medicine, Department for Histology and Embryology, University of Rijeka , Rijeka , Croatia
| | - Marija Milovanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac , Kragujevac , Serbia
| | - Daria Kvestak
- Center for Proteomics, Faculty of Medicine, Department for Histology and Embryology, University of Rijeka , Rijeka , Croatia
| | - Aleksandar Arsenijevic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac , Kragujevac , Serbia
| | - Bojana Stojanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia; Faculty of Medical Sciences, Institute of Pathophysiology, University of Kragujevac, Kragujevac, Serbia
| | - Irena Tanaskovic
- Faculty of Medical Sciences, Institute of Histology, University of Kragujevac , Kragujevac , Serbia
| | - Astrid Krmpotic
- Center for Proteomics, Faculty of Medicine, Department for Histology and Embryology, University of Rijeka , Rijeka , Croatia
| | - Nebojsa Arsenijevic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac , Kragujevac , Serbia
| | - Stipan Jonjic
- Center for Proteomics, Faculty of Medicine, Department for Histology and Embryology, University of Rijeka , Rijeka , Croatia
| | - Miodrag L Lukic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac , Kragujevac , Serbia
| |
Collapse
|
24
|
Alston CI, Dix RD. Murine cytomegalovirus infection of mouse macrophages stimulates early expression of suppressor of cytokine signaling (SOCS)1 and SOCS3. PLoS One 2017; 12:e0171812. [PMID: 28182772 PMCID: PMC5300177 DOI: 10.1371/journal.pone.0171812] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 01/26/2017] [Indexed: 12/13/2022] Open
Abstract
Human cytomegalovirus (HCMV) is a species-specific β-herpesvirus that infects for life up to 80% of the world’s population and causes severe morbidity in at-risk immunocompromised populations. Suppressors of cytokine signaling (SOCS)1 and SOCS3 are host proteins that act as inducible negative feedback regulators of cytokine signaling and have been implicated in several ocular diseases and viral infections. We recently found in our mouse model of experimental cytomegalovirus retinitis that subretinally-injected murine cytomegalovirus (MCMV) stimulates ocular SOCS1 and SOCS3 during retrovirus-induced immune suppression of murine AIDS (MAIDS), and that infiltrating macrophages are prominent cellular sources of retinal SOCS1 and SOCS3 expression. Herein we investigate possible virologic mechanisms whereby MCMV infection may stimulate SOCS1 and/or SOCS3 expression in cell culture. We report that infection of IC-21 mouse macrophages with MCMV propagated through the salivary glands of BALB/c mice, but not from tissue culture in C57BL/6 fibroblasts, transiently stimulates SOCS1 and SOCS3 mRNA transcripts, but not SOCS5 mRNA. Viral tegument proteins are insufficient for this stimulation, as replication-deficient UV-inactivated MCMV fails to stimulate SOCS1 or SOCS3 in IC-21 macrophages. By contrast, infection of murine embryonic fibroblasts (MEFs) with either productive MCMV or UV-inactivated MCMV significantly stimulates SOCS1 and SOCS3 mRNA expression early after infection. Treatment of MCMV-infected IC-21 mouse macrophages with the antiviral drug ganciclovir significantly decreases MCMV-stimulated SOCS3 expression at 3 days post-infection. These data suggest cell type-specific, different roles for viral immediate early or early gene expression and/or viral tegument proteins in the early stimulation of SOCS1 and SOCS3 during MCMV infection. Furthermore, putative biphasic stimulation of SOCS3 during late MCMV infection of IC-21 mouse macrophages may occur by divergent virologic mechanisms.
Collapse
Affiliation(s)
- Christine I. Alston
- Viral Immunology Center, Department of Biology, Georgia State University, Atlanta, Georgia, United States of America
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Richard D. Dix
- Viral Immunology Center, Department of Biology, Georgia State University, Atlanta, Georgia, United States of America
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
25
|
Murine Cytomegalovirus Deubiquitinase Regulates Viral Chemokine Levels To Control Inflammation and Pathogenesis. mBio 2017; 8:mBio.01864-16. [PMID: 28096485 PMCID: PMC5241396 DOI: 10.1128/mbio.01864-16] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Maintaining control over inflammatory processes represents a paradox for viral pathogens. Although many viruses induce host inflammatory responses to facilitate infection, control is necessary to avoid overactivation. One way is through the manipulation of proinflammatory chemokine levels, both host and viral. Murine cytomegalovirus (MCMV), a model betaherpesvirus, encodes a viral C-C chemokine, MCK2, which promotes host inflammatory responses and incorporates into virions to facilitate viral dissemination. Here, we show that the activity of M48, the conserved MCMV deubiquitinating enzyme (DUB), regulates MCK2 levels during infection. Inactivation of M48 DUB activity results in viral attenuation and exacerbates virally induced, MCK2-dependent inflammatory responses. M48 DUB activity also influences MCK2 incorporation into virions. Importantly, attenuation of DUB-mutant virus acute replication in vitro and in vivo is largely ameliorated by targeted deletion of MCK2. Thus, uncontrolled MCK2 levels appear to mediate DUB-mutant virus attenuation in specific tissues or cell types. This demonstrates that MCMV M48 DUB activity plays a previously unappreciated role in controlling MCK2 levels, thereby managing MCK2-dependent processes. These findings reveal a novel intrinsic control mechanism of virally induced inflammation and support the identification of betaherpesvirus DUBs as possible new targets for antiviral therapies. Human cytomegalovirus infections represent a tremendous burden not only to those afflicted but also to health care systems worldwide. As cytomegalovirus infections are a leading cause of nongenetic sensory loss and neurodevelopmental delay, it is imperative that valuable model systems exist in order that we might understand what viral factors contribute to replication and pathogenesis. Currently, the only approved drug treatments against CMV infection are nucleoside analogues, to which some strains have become resistant. Understanding unique viral enzymatic contributions to infections will allow the development of novel pharmacological therapies. Here, we show that M48, the conserved MCMV deubiquitinase, is critical for MCMV replication in mice and demonstrate that attenuation is due to deregulated production of a viral proinflammatory chemokine. The deubiquitinases of both human and murine CMV represent structurally unique DUBs and are therefore attractive targets for pharmacological intervention. Continued research into the substrates of these DUBs will lend additional insight into their potential as targets.
Collapse
|
26
|
Dogra P, Miller-Kittrell M, Pitt E, Jackson JW, Masi T, Copeland C, Wu S, Miller WE, Sparer T. A little cooperation helps murine cytomegalovirus (MCMV) go a long way: MCMV co-infection rescues a chemokine salivary gland defect. J Gen Virol 2016; 97:2957-2972. [PMID: 27638684 DOI: 10.1099/jgv.0.000603] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Cytomegaloviruses (CMVs) produce chemokines (vCXCLs) that have both sequence and functional homology to host chemokines. Assessment of vCXCL-1's role in CMV infection is limited to in vitro and in silico analysis due to CMVs species specificity. In this study, we used the murine CMV (MCMV) mouse model to evaluate the function of vCXCL-1 in vivo. Recombinant MCMVs expressing chimpanzee CMV vCXCL-1 (vCXCL-1CCMV) or host chemokine, mCXCL1, underwent primary dissemination to the popliteal lymph node, spleen and lung similar to the parental MCMV. However, neither of the recombinants expressing chemokines was recovered from the salivary gland (SG) at any time post-infection although viral DNA was detected. This implies that the virus does not grow in the SG or the overexpressed chemokine induces an immune response that leads to suppressed growth. Pointing to immune suppression of virus replication, recombinant viruses were isolated from the SG following infection of immune-ablated mice [i.e. SCID (severe combined immunodeficiency), NSG (non-obese diabetic SCID gamma) or cyclophosphamide treated]. Depletion of neutrophils or NK cells does not rescue the recovery of chemokine-expressing recombinants in the SG. Surprisingly we found that co-infection of parental virus and chemokine-expressing virus leads to the recovery of the recombinants in the SG. We suggest that parental virus reduces the levels of chemokine expression leading to a decrease in inflammatory monocytes and subsequent SG growth. Therefore, aberrant expression of the chemokines induces cells of the innate and adaptive immune system that curtail the growth and dissemination of the recombinants in the SG.
Collapse
Affiliation(s)
- Pranay Dogra
- Department of Microbiology, University of Tennessee, 1414 Cumberland Avenue, Knoxville, TN 37996, USA
| | - Mindy Miller-Kittrell
- Department of Microbiology, University of Tennessee, 1414 Cumberland Avenue, Knoxville, TN 37996, USA
| | - Elisabeth Pitt
- Department of Microbiology, University of Tennessee, 1414 Cumberland Avenue, Knoxville, TN 37996, USA
| | - Joseph W Jackson
- Department of Microbiology, University of Tennessee, 1414 Cumberland Avenue, Knoxville, TN 37996, USA
| | - Tom Masi
- Department of Microbiology, University of Tennessee, 1414 Cumberland Avenue, Knoxville, TN 37996, USA
| | - Courtney Copeland
- Department of Microbiology, University of Tennessee, 1414 Cumberland Avenue, Knoxville, TN 37996, USA
| | - Shuen Wu
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH 45267-0524, USA
| | - William E Miller
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH 45267-0524, USA
| | - Tim Sparer
- Department of Microbiology, University of Tennessee, 1414 Cumberland Avenue, Knoxville, TN 37996, USA
| |
Collapse
|
27
|
Campadelli-Fiume G, Collins-McMillen D, Gianni T, Yurochko AD. Integrins as Herpesvirus Receptors and Mediators of the Host Signalosome. Annu Rev Virol 2016; 3:215-236. [PMID: 27501260 DOI: 10.1146/annurev-virology-110615-035618] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The repertoire of herpesvirus receptors consists of nonintegrin and integrin molecules. Integrins interact with the conserved glycoproteins gH/gL or gB. This interaction is a conserved biology across the Herpesviridae family, likely directed to promote virus entry and endocytosis. Herpesviruses exploit this interaction to execute a range of critical functions that include (a) relocation of nonintegrin receptors (e.g., herpes simplex virus nectin1 and Kaposi's sarcoma-associated herpesvirus EphA2), or association with nonintegrin receptors (i.e., human cytomegalovirus EGFR), to dictate species-specific entry pathways; (b) activation of multiple signaling pathways (e.g., Ca2+ release, c-Src, FAK, MAPK, and PI3K); and (c) association with Rho GTPases, tyrosine kinase receptors, Toll-like receptors, which result in cytoskeletal remodeling, differential cell type targeting, and innate responses. In turn, integrins can be modulated by viral proteins (e.g., Epstein-Barr virus LMPs) to favor spread of transformed cells. We propose that herpesviruses evolved a multipartite entry system to allow interaction with multiple receptors, including integrins, required for their sophisticated life cycle.
Collapse
Affiliation(s)
- Gabriella Campadelli-Fiume
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40126 Bologna, Italy;
| | - Donna Collins-McMillen
- Department of Microbiology and Immunology, Center for Molecular and Tumor Virology, and Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71130;
| | - Tatiana Gianni
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40126 Bologna, Italy;
| | - Andrew D Yurochko
- Department of Microbiology and Immunology, Center for Molecular and Tumor Virology, and Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71130; .,Feist-Weiller Cancer Center and Center for Excellence in Arthritis and Rheumatology, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71130
| |
Collapse
|
28
|
Oduro JD, Redeker A, Lemmermann NAW, Ebermann L, Marandu TF, Dekhtiarenko I, Holzki JK, Busch DH, Arens R, Čičin-Šain L. Murine cytomegalovirus (CMV) infection via the intranasal route offers a robust model of immunity upon mucosal CMV infection. J Gen Virol 2015; 97:185-195. [PMID: 26555192 DOI: 10.1099/jgv.0.000339] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Cytomegalovirus (CMV) is a ubiquitous virus, causing the most common congenital infection in humans, yet a vaccine against this virus is not available. Experimental studies of immunity against CMV in animal models of infection, such as the infection of mice with mouse CMV (MCMV), have relied mainly on parenteral infection protocols, although the virus naturally transmits by mucosal routes via body fluids. To characterize the biology of infections by mucosal routes, we compared the kinetics of virus replication, latent viral load and CD8 T-cell responses in lymphoid organs upon experimental intranasal (targeting the respiratory tract) and intragastric (targeting the digestive tract) infection with systemic intraperitoneal infection of two unrelated mouse strains. We observed that intranasal infection induced robust and long-term virus replication in the lungs and salivary glands but limited replication in the spleen. CD8 T-cell responses were somewhat weaker than upon intraperitoneal infection but showed similar kinetic profiles and phenotypes of antigen-specific cells. In contrast, intragastric infection resulted in abortive or poor virus replication in all tested organs and poor T-cell responses to the virus, especially at late times after infection. Consistent with the T-cell kinetics, the MCMV latent load was high in the lungs but low in the spleen of intranasally infected mice and lowest in all tested organs upon intragastric infection. In conclusion, we showed that intranasal but not intragastric infection of mice with MCMV represents a robust model to study the short- and long-term biology of CMV infection by a mucosal route.
Collapse
Affiliation(s)
- Jennifer D Oduro
- Department of Vaccinology/Immune Aging and Chronic Infections, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Anke Redeker
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Niels A W Lemmermann
- Institute for Virology and Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Linda Ebermann
- Department of Vaccinology/Immune Aging and Chronic Infections, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Thomas F Marandu
- Department of Vaccinology/Immune Aging and Chronic Infections, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Dar es Salaam University College of Education, Tanzania
| | - Iryna Dekhtiarenko
- Department of Vaccinology/Immune Aging and Chronic Infections, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Julia K Holzki
- Department of Vaccinology/Immune Aging and Chronic Infections, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Dirk H Busch
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München, Munich, Germany.,Focus Group 'Clinical Cell Processing and Purification', Institute for Advanced Study, Technische Universität München, Munich, Germany.,German Center for Infection Research (DZIF), Braunschweig, Germany
| | - Ramon Arens
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Luka Čičin-Šain
- German Center for Infection Research (DZIF), Braunschweig, Germany.,Department of Vaccinology/Immune Aging and Chronic Infections, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Department for Virology, Medical School Hannover, Hannover, Germany
| |
Collapse
|
29
|
Dogra P, Martin EB, Williams A, Richardson RL, Foster JS, Hackenback N, Kennel SJ, Sparer TE, Wall JS. Novel heparan sulfate-binding peptides for blocking herpesvirus entry. PLoS One 2015; 10:e0126239. [PMID: 25992785 PMCID: PMC4436313 DOI: 10.1371/journal.pone.0126239] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 03/31/2015] [Indexed: 11/18/2022] Open
Abstract
Human cytomegalovirus (HCMV) infection can lead to congenital hearing loss and mental retardation. Upon immune suppression, reactivation of latent HCMV or primary infection increases morbidity in cancer, transplantation, and late stage AIDS patients. Current treatments include nucleoside analogues, which have significant toxicities limiting their usefulness. In this study we screened a panel of synthetic heparin-binding peptides for their ability to prevent CMV infection in vitro. A peptide designated, p5+14 exhibited ~ 90% reduction in murine CMV (MCMV) infection. Because negatively charged, cell-surface heparan sulfate proteoglycans (HSPGs), serve as the attachment receptor during the adsorption phase of the CMV infection cycle, we hypothesized that p5+14 effectively competes for CMV adsorption to the cell surface resulting in the reduction in infection. Positively charged Lys residues were required for peptide binding to cell-surface HSPGs and reducing viral infection. We show that this inhibition was not due to a direct neutralizing effect on the virus itself and that the peptide blocked adsorption of the virus. The peptide also inhibited infection of other herpesviruses: HCMV and herpes simplex virus 1 and 2 in vitro, demonstrating it has broad-spectrum antiviral activity. Therefore, this peptide may offer an adjunct therapy for the treatment of herpes viral infections and other viruses that use HSPGs for entry.
Collapse
Affiliation(s)
- Pranay Dogra
- Department of Microbiology, The University of Tennessee, Knoxville, Tennessee, United States of America
| | - Emily B. Martin
- Department of Medicine, The University of Tennessee Graduate School of Medicine, Knoxville, Tennessee, United States of America
| | - Angela Williams
- Department of Medicine, The University of Tennessee Graduate School of Medicine, Knoxville, Tennessee, United States of America
| | - Raphael L. Richardson
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, The University of Tennessee, Knoxville, Tennessee, United States of America
| | - James S. Foster
- Department of Medicine, The University of Tennessee Graduate School of Medicine, Knoxville, Tennessee, United States of America
| | - Nicole Hackenback
- Department of Medicine, The University of Tennessee Graduate School of Medicine, Knoxville, Tennessee, United States of America
| | - Stephen J. Kennel
- Department of Medicine, The University of Tennessee Graduate School of Medicine, Knoxville, Tennessee, United States of America
- Department of Radiology, The University of Tennessee Graduate School of Medicine, Knoxville, Tennessee, United States of America
| | - Tim E. Sparer
- Department of Microbiology, The University of Tennessee, Knoxville, Tennessee, United States of America
| | - Jonathan S. Wall
- Department of Medicine, The University of Tennessee Graduate School of Medicine, Knoxville, Tennessee, United States of America
- Department of Radiology, The University of Tennessee Graduate School of Medicine, Knoxville, Tennessee, United States of America
| |
Collapse
|
30
|
Podlech J, Reddehase MJ, Adler B, Lemmermann NAW. Principles for studying in vivo attenuation of virus mutants: defining the role of the cytomegalovirus gH/gL/gO complex as a paradigm. Med Microbiol Immunol 2015; 204:295-305. [DOI: 10.1007/s00430-015-0405-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 03/04/2015] [Indexed: 10/23/2022]
|
31
|
Non-redundant and redundant roles of cytomegalovirus gH/gL complexes in host organ entry and intra-tissue spread. PLoS Pathog 2015; 11:e1004640. [PMID: 25659098 PMCID: PMC4450070 DOI: 10.1371/journal.ppat.1004640] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 12/22/2014] [Indexed: 01/05/2023] Open
Abstract
Herpesviruses form different gH/gL virion envelope glycoprotein complexes that serve as entry complexes for mediating viral cell-type tropism in vitro; their roles in vivo, however, remained speculative and can be addressed experimentally only in animal models. For murine cytomegalovirus two alternative gH/gL complexes, gH/gL/gO and gH/gL/MCK-2, have been identified. A limitation of studies on viral tropism in vivo has been the difficulty in distinguishing between infection initiation by viral entry into first-hit target cells and subsequent cell-to-cell spread within tissues. As a new strategy to dissect these two events, we used a gO-transcomplemented ΔgO mutant for providing the gH/gL/gO complex selectively for the initial entry step, while progeny virions lack gO in subsequent rounds of infection. Whereas gH/gL/gO proved to be critical for establishing infection by efficient entry into diverse cell types, including liver macrophages, endothelial cells, and hepatocytes, it was dispensable for intra-tissue spread. Notably, the salivary glands, the source of virus for host-to-host transmission, represent an exception in that entry into virus-producing cells did not strictly depend on either the gH/gL/gO or the gH/gL/MCK-2 complex. Only if both complexes were absent in gO and MCK-2 double-knockout virus, in vivo infection was abolished at all sites.
Collapse
|
32
|
McWhorter AR, Smith LM, Shellam GR, Redwood AJ. Murine cytomegalovirus strains co-replicate at multiple tissue sites and establish co-persistence in salivary glands in the absence of Ly49H-mediated competition. J Gen Virol 2015; 96:1127-1137. [PMID: 25575707 DOI: 10.1099/vir.0.000047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 01/06/2015] [Indexed: 11/18/2022] Open
Abstract
Infection with multiple genetically distinct strains of pathogen is common and can lead to positive (complementation) or negative (competitive) within-host interactions. These interactions can alter aspects of the disease process and help shape pathogen evolution. Infection of the host with multiple strains of cytomegalovirus (CMV) occurs frequently in humans and mice. Profound, NK-cell-mediated (apparent) competition has been identified in C57BL/6 mice, and prevented the replication and shedding of certain co-infecting CMV strains. However, the frequency of such strong competition has not been established. Other within-host interactions such as complementation or alternative forms of competition remain possible. Moreover, high rates of recombination in both human CMV and murine CMV (MCMV) suggest prolonged periods of viral co-replication, rather than strong competitive suppression. An established model was employed to investigate the different possible outcomes of multi-strain infection in other mouse strains. In this study, co-replication of up to four strains of MCMV in the spleen, liver and salivary glands was observed in both MCMV-susceptible and MCMV-resistant mice. In the absence of apparent competition, no other forms of competition were unmasked. In addition, no evidence of complementation between viral strains was observed. Importantly, co-replication of MCMV strains was apparent for up to 90 days in the salivary glands. These data indicated that competition was not the default outcome of multi-strain CMV infection. Prolonged, essentially neutral, co-replication may be the norm, allowing for multi-strain transmission and prolonged opportunities for recombination.
Collapse
Affiliation(s)
- Andrea R McWhorter
- School of Pathology and Laboratory Medicine, M504, The University of Western Australia, 35 Stirling Highway, Crawley, Western Australia 6009, Australia
| | - Lee M Smith
- School of Pathology and Laboratory Medicine, M504, The University of Western Australia, 35 Stirling Highway, Crawley, Western Australia 6009, Australia
| | - Geoffrey R Shellam
- School of Pathology and Laboratory Medicine, M504, The University of Western Australia, 35 Stirling Highway, Crawley, Western Australia 6009, Australia
| | - Alec J Redwood
- The Institute for Immunology and Infectious Diseases, Murdoch University, Building 390, Discovery Way, Murdoch, Western Australia 6150, Australia.,School of Pathology and Laboratory Medicine, M504, The University of Western Australia, 35 Stirling Highway, Crawley, Western Australia 6009, Australia
| |
Collapse
|
33
|
Stahl FR, Keyser KA, Heller K, Bischoff Y, Halle S, Wagner K, Messerle M, Förster R. Mck2-dependent infection of alveolar macrophages promotes replication of MCMV in nodular inflammatory foci of the neonatal lung. Mucosal Immunol 2015; 8:57-67. [PMID: 24894498 DOI: 10.1038/mi.2014.42] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 04/15/2014] [Indexed: 02/08/2023]
Abstract
Infection with cytomegalovirus (CMV) shows a worldwide high prevalence with only immunocompromised individuals or newborns to become symptomatic. The host's constitution and the pathogen's virulence determine whether disease occurs after infection. Mouse CMV (MCMV) is an appreciated pathogen for in vivo investigation of host-pathogen interactions. It has recently been reported that a single base pair deletion can spontaneously occur in the open reading frame of MCMV-encoded chemokine 2 (MCK2), preventing the expression of the full-length gene product. To study the consequences of this mutation, we compared the Mck2-defective reporter virus MCMV-3D with the newly generated repaired Mck2(+) mutant MCMV-3DR. Compared with MCMV-3D, neonatal mice infected with MCMV-3DR showed severe viral disease after lung infection. Viral disease coincided with high viral activity in multiple organs and increased virus replication in previously described nodular inflammatory foci (NIF) in the lung. Notably, MCMV-3DR showed tropism for alveolar macrophages in vitro and in vivo, whereas MCMV-3D did not infect this cell type. Moreover, in vivo depletion of alveolar macrophages reduced MCMV-3DR replication in the lung. We proposed an Mck2-mediated mechanism by which MCMV exploits alveolar macrophages to increase replication upon first encounter with the host's lung mucosa.
Collapse
Affiliation(s)
- F R Stahl
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - K A Keyser
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - K Heller
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Y Bischoff
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - S Halle
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - K Wagner
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - M Messerle
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - R Förster
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
34
|
Alexandre YO, Cocita CD, Ghilas S, Dalod M. Deciphering the role of DC subsets in MCMV infection to better understand immune protection against viral infections. Front Microbiol 2014; 5:378. [PMID: 25120535 PMCID: PMC4114203 DOI: 10.3389/fmicb.2014.00378] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 07/04/2014] [Indexed: 12/22/2022] Open
Abstract
Infection of mice with murine cytomegalovirus (MCMV) recapitulates many physiopathological characteristics of human CMV infection and enables studying the interactions between a virus and its natural host. Dendritic cells (DC) are mononuclear phagocytes linking innate and adaptive immunity which are both necessary for MCMV control. DC are critical for the induction of cellular immunity because they are uniquely efficient for the activation of naïve T cells during their first encounter with a pathogen. DC are equipped with a variety of innate immune recognition receptors (I2R2) allowing them to detect pathogens or infections and to engulf molecules, microorganisms or cellular debris. The combinatorial engagement of I2R2 during infections controls DC maturation and shapes their response in terms of cytokine production, activation of natural killer (NK) cells and functional polarization of T cells. Several DC subsets exist which express different arrays of I2R2 and are specialized in distinct functions. The study of MCMV infection helped deciphering the physiological roles of DC subsets and their molecular regulation. It allowed the identification and first in vivo studies of mouse plasmacytoid DC which produce high level of interferons-α/β early after infection. Despite its ability to infect DC and dampen their functions, MCMV induces very robust, efficient and long-lasting CD8 T cell responses. Their priming may rely on the unique ability of uninfected XCR1+ DC to cross-present engulfed viral antigens and thus to counter MCMV interference with antigen presentation. A balance appears to have been reached during co-evolution, allowing controlled replication of the virus for horizontal spread without pathological consequences for the immunocompetent host. We will discuss the role of the interplay between the virus and DC in setting this balance, and how advancing this knowledge further could help develop better vaccines against other intracellular infectious agents.
Collapse
Affiliation(s)
- Yannick O Alexandre
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille University, UM2 Marseille, France ; Institut National de la Santé et de la Recherche Médicale, U1104 Marseille, France ; Centre National de la Recherche Scientifique, UMR7280 Marseille, France
| | - Clément D Cocita
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille University, UM2 Marseille, France ; Institut National de la Santé et de la Recherche Médicale, U1104 Marseille, France ; Centre National de la Recherche Scientifique, UMR7280 Marseille, France
| | - Sonia Ghilas
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille University, UM2 Marseille, France ; Institut National de la Santé et de la Recherche Médicale, U1104 Marseille, France ; Centre National de la Recherche Scientifique, UMR7280 Marseille, France
| | - Marc Dalod
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille University, UM2 Marseille, France ; Institut National de la Santé et de la Recherche Médicale, U1104 Marseille, France ; Centre National de la Recherche Scientifique, UMR7280 Marseille, France
| |
Collapse
|
35
|
Beverley PCL, Ruzsics Z, Hey A, Hutchings C, Boos S, Bolinger B, Marchi E, O'Hara G, Klenerman P, Koszinowski UH, Tchilian EZ. A novel murine cytomegalovirus vaccine vector protects against Mycobacterium tuberculosis. THE JOURNAL OF IMMUNOLOGY 2014; 193:2306-16. [PMID: 25070842 PMCID: PMC4134927 DOI: 10.4049/jimmunol.1302523] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Tuberculosis remains a global health problem so that a more effective vaccine than bacillus Calmette–Guérin is urgently needed. Cytomegaloviruses persist lifelong in vivo and induce powerful immune and increasing (“inflationary”) responses, making them attractive vaccine vectors. We have used an m1–m16-deleted recombinant murine CMV (MCMV) expressing Mycobacterium tuberculosis Ag 85A to show that infection of mice with this recombinant significantly reduces the mycobacterial load after challenge with M. tuberculosis, whereas control empty virus has a lesser effect. Both viruses induce immune responses to H-2d–restricted epitopes of MCMV pp89 and M18 Ags characteristic of infection with other MCMVs. A low frequency of 85A-specific memory cells could be revealed by in vivo or in vitro boosting or after challenge with M. tuberculosis. Kinetic analysis of M. tuberculosis growth in the lungs of CMV-infected mice shows early inhibition of M. tuberculosis growth abolished by treatment with NK-depleting anti–asialo ganglio-N-tetraosylceramide Ab. Microarray analysis of the lungs of naive and CMV-infected mice shows increased IL-21 mRNA in infected mice, whereas in vitro NK assays indicate increased levels of NK activity. These data indicate that activation of NK cells by MCMV provides early nonspecific protection against M. tuberculosis, potentiated by a weak 85A-specific T cell response, and they reinforce the view that the innate immune system plays an important role in both natural and vaccine-induced protection against M. tuberculosis.
Collapse
Affiliation(s)
- Peter C L Beverley
- Nuffield Department of Medicine, University of Oxford, Oxford OX1 3SY, United Kingdom; and
| | - Zsolt Ruzsics
- Max von Pettenkofer Institute, Ludwig Maximilians University, D-80336 Munich, Germany
| | - Ariann Hey
- Nuffield Department of Medicine, University of Oxford, Oxford OX1 3SY, United Kingdom; and
| | - Claire Hutchings
- Nuffield Department of Medicine, University of Oxford, Oxford OX1 3SY, United Kingdom; and
| | - Simone Boos
- Max von Pettenkofer Institute, Ludwig Maximilians University, D-80336 Munich, Germany
| | - Beatrice Bolinger
- Nuffield Department of Medicine, University of Oxford, Oxford OX1 3SY, United Kingdom; and
| | - Emanuele Marchi
- Nuffield Department of Medicine, University of Oxford, Oxford OX1 3SY, United Kingdom; and
| | - Geraldine O'Hara
- Nuffield Department of Medicine, University of Oxford, Oxford OX1 3SY, United Kingdom; and
| | - Paul Klenerman
- Nuffield Department of Medicine, University of Oxford, Oxford OX1 3SY, United Kingdom; and
| | - Ulrich H Koszinowski
- Max von Pettenkofer Institute, Ludwig Maximilians University, D-80336 Munich, Germany
| | - Elma Z Tchilian
- Nuffield Department of Medicine, University of Oxford, Oxford OX1 3SY, United Kingdom; and
| |
Collapse
|
36
|
Daley-Bauer LP, Roback LJ, Wynn GM, Mocarski ES. Cytomegalovirus hijacks CX3CR1(hi) patrolling monocytes as immune-privileged vehicles for dissemination in mice. Cell Host Microbe 2014; 15:351-62. [PMID: 24629341 PMCID: PMC3989205 DOI: 10.1016/j.chom.2014.02.002] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2013] [Revised: 12/10/2013] [Accepted: 02/04/2014] [Indexed: 12/24/2022]
Abstract
Peripheral blood myelomonocytic cells are important for cytomegalovirus dissemination to distal organs such as salivary glands where persistent replication and shedding dictates transmission patterns. We find that this process is markedly enhanced by the murine cytomegalovirus (MCMV)-encoded CC chemokine, MCK2, which promotes recruitment of CX3CR1(hi) patrolling monocytes to initial infection sites in the mouse. There, these cells become infected and traffic via the bloodstream to distal sites. In contrast, inflammatory monocytes, the other major myelomonocytic subset, remain virus negative. CX3CR1 deficiency prevents patrolling monocyte migration on the vascular endothelium and interrupts MCMV dissemination to the salivary glands independent of antiviral NK and T cell immune control. In this manner, CX3CR1(hi) patrolling monocytes serve as immune-privileged vehicles to transport MCMV via the bloodstream to distal organs. MCMV commandeers patrolling monocytes to mediate systemic infection and seed a persistent reservoir essential for horizontal transmission.
Collapse
Affiliation(s)
- Lisa P Daley-Bauer
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Linda J Roback
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Grace M Wynn
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Edward S Mocarski
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
37
|
Abstract
Although human cytomegalovirus (HCMV) primary infection is generally asymptomatic, in immune-compromised patients HCMV increases morbidity and mortality. As a member of the betaherpesvirus family, in vivo studies of HCMV are limited due to its species specificity. CMVs from other species are often used as surrogates to express HCMV genes/proteins or used as models for inferring HCMV protein function in humans. Using innovative experiments, these animal models have answered important questions about CMV's life cycle, dissemination, pathogenesis, immune evasion, and host immune response. This chapter provides CMV biologists with an overview of the insights gained using these animal models. Subsequent chapters will provide details of the specifics of the experimental methods developed for each of the animal models discussed here.
Collapse
Affiliation(s)
- Pranay Dogra
- Department of Microbiology, University of Tennessee, Knoxville, TN, USA
| | | |
Collapse
|
38
|
Cytomegalovirus expresses the chemokine homologue vXCL1 capable of attracting XCR1+ CD4- dendritic cells. J Virol 2013; 88:292-302. [PMID: 24155383 DOI: 10.1128/jvi.02330-13] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Cytomegaloviruses (CMV) have developed various strategies to escape the immune system of the host. One strategy involves the expression of virus-encoded chemokines to modulate the host chemokine network. We have identified in the English isolate of rat CMV (murid herpesvirus 8 [MuHV8]) an open reading frame encoding a protein homologous to the chemokine XCL1, the only known C chemokine. Viral XCL1 (vXCL1), a glycosylated protein of 96 amino acids, can be detected 13 h postinfection in the supernatant of MuHV8-infected rat embryo fibroblasts. vXCL1 exclusively binds to CD4(-) rat dendritic cells (DC), a subset of DC that express the corresponding chemokine receptor XCR1. Like endogenous rat XCL1, vXCL1 selectively chemoattracts XCR1(+) CD4(-) DC. Since XCR1(+) DC in mice and humans have been shown to excel in antigen cross-presentation and thus in the induction of cytotoxic CD8(+) T lymphocytes, the virus has apparently hijacked this gene to subvert cytotoxic immune responses. The biology of vXCL1 offers an interesting opportunity to study the role of XCL1 and XCR1(+) DC in the cross-presentation of viral antigens.
Collapse
|
39
|
An attenuated cytomegalovirus vaccine with a deletion of a viral chemokine gene is protective against congenital CMV transmission in a guinea pig model. Clin Dev Immunol 2013; 2013:906948. [PMID: 24000289 PMCID: PMC3755440 DOI: 10.1155/2013/906948] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 05/24/2013] [Accepted: 06/05/2013] [Indexed: 01/05/2023]
Abstract
Development of a vaccine against congenital cytomegalovirus (CMV) infection is a public health priority, but CMVs encode immune evasion genes that complicate live virus vaccine design. To resolve this problem, this study employed guanosyl phosphoribosyl transferase (gpt) mutagenesis to generate a recombinant guinea pig CMV (GPCMV) with a knockout of a viral chemokine gene, GPCMV MIP (gp1). MIP deletion virus replicated with wild-type kinetics in cell culture but was attenuated in nonpregnant guinea pigs, demonstrating reduced viremia and reduced inflammation and histopathology (compared to a control virus with an intact GPCMV MIP gene) following footpad inoculation. In spite of attenuation, the vaccine was immunogenic, eliciting antibody responses comparable to those observed in natural infection. To assess its protective potential as a vaccine, either recombinant virus or placebo was used to immunize seronegative female guinea pigs. Dams were challenged in the early 3rd trimester with salivary gland-adapted GPCMV. Immunization protected against DNAemia (1/15 in vaccine group versus 12/13 in the control group, P < 0.01). Mean birth weights were significantly higher in pups born to vaccinated dams compared to controls (98.7 g versus 71.2 g, P < 0.01). Vaccination reduced pup mortality, from 35/50 (70%) in controls to 8/52 (15%) in the immunization group. Congenital GPCMV infection was also reduced, from 35/50 (70%) in controls to 9/52 (17%) in the vaccine group (P < 0.0001). We conclude that deletion of an immune modulation gene can attenuate the pathogenicity of GPCMV while resulting in a viral vaccine that retains immunogenicity and demonstrates efficacy against congenital infection and disease.
Collapse
|
40
|
Wagner FM, Brizic I, Prager A, Trsan T, Arapovic M, Lemmermann NAW, Podlech J, Reddehase MJ, Lemnitzer F, Bosse JB, Gimpfl M, Marcinowski L, MacDonald M, Adler H, Koszinowski UH, Adler B. The viral chemokine MCK-2 of murine cytomegalovirus promotes infection as part of a gH/gL/MCK-2 complex. PLoS Pathog 2013; 9:e1003493. [PMID: 23935483 PMCID: PMC3723581 DOI: 10.1371/journal.ppat.1003493] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2012] [Accepted: 05/22/2013] [Indexed: 11/26/2022] Open
Abstract
Human cytomegalovirus (HCMV) forms two gH/gL glycoprotein complexes, gH/gL/gO and gH/gL/pUL(128,130,131A), which determine the tropism, the entry pathways and the mode of spread of the virus. For murine cytomegalovirus (MCMV), which serves as a model for HCMV, a gH/gL/gO complex functionally homologous to the HCMV gH/gL/gO complex has been described. Knock-out of MCMV gO does impair, but not abolish, virus spread indicating that also MCMV might form an alternative gH/gL complex. Here, we show that the MCMV CC chemokine MCK-2 forms a complex with the glycoprotein gH, a complex which is incorporated into the virion. We could additionally show that mutants lacking both, gO and MCK-2 are not able to produce infectious virus. Trans-complementation of these double mutants with either gO or MCK-2 showed that both proteins can promote infection of host cells, although through different entry pathways. MCK-2 has been extensively studied in vivo by others. It has been shown to be involved in attracting cells for virus dissemination and in regulating antiviral host responses. We now show that MCK-2, by forming a complex with gH, strongly promotes infection of macrophages in vitro and in vivo. Thus, MCK-2 may play a dual role in MCMV infection, as a chemokine regulating the host response and attracting specific target cells and as part of a glycoprotein complex promoting entry into cells crucial for virus dissemination. Several human herpesviruses form alternative gH/gL complexes which determine the tropism for different cell types. For murine cytomegalovirus (MCMV), a gH/gL/gO complex has recently been characterized. Here, we present the identification and characterization of an alternative gH/gL/MCK-2 complex which promotes MCMV spread and is important for efficient infection of macrophages in vitro and in vivo. Association of the MCMV CC chemokine MCK-2 with a glycoprotein complex promoting virus entry is a novel function for the well-characterized MCK-2. Virus mutants lacking MCK-2 have been shown to exhibit a reduced capacity to attract leukocytes and a disregulated T cell control of the MCMV infection in vivo. These defects can be attributed to the chemokine function of MCK-2. Yet, the observation that MCK-2 knock-out mutants additionally are impaired in infecting leukocytes in vivo is consistent with our new finding that MCK-2 forms a glycoprotein complex promoting entry into monocytic cells. gH/gL complexes associating with multifunctional proteins add a new level of complexity to the interpretation of infection phenotypes of the respective knock-out herpesviruses.
Collapse
Affiliation(s)
- Felicia M. Wagner
- Max von Pettenkofer-Institute for Virology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Ilija Brizic
- Max von Pettenkofer-Institute for Virology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Adrian Prager
- Max von Pettenkofer-Institute for Virology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Tihana Trsan
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Maja Arapovic
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Niels A. W. Lemmermann
- Institute for Virology and Research Center for Immunology (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Jürgen Podlech
- Institute for Virology and Research Center for Immunology (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Matthias J. Reddehase
- Institute for Virology and Research Center for Immunology (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Frederic Lemnitzer
- Max von Pettenkofer-Institute for Virology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Jens Bernhard Bosse
- Max von Pettenkofer-Institute for Virology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Martina Gimpfl
- Max von Pettenkofer-Institute for Virology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Lisa Marcinowski
- Max von Pettenkofer-Institute for Virology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Margaret MacDonald
- Laboratory of Virology and Infectious Disease, Rockefeller University, New York, New York, United States of America
| | - Heiko Adler
- Research Unit Gene Vectors, German Research Center for Environmental Health (GmbH), Munich, Germany
| | - Ulrich H. Koszinowski
- Max von Pettenkofer-Institute for Virology, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Barbara Adler
- Max von Pettenkofer-Institute for Virology, Ludwig-Maximilians-University Munich, Munich, Germany
- * E-mail:
| |
Collapse
|
41
|
A chemokine-like viral protein enhances alpha interferon production by plasmacytoid dendritic cells but delays CD8+ T cell activation and impairs viral clearance. J Virol 2013; 87:7911-20. [PMID: 23658453 DOI: 10.1128/jvi.00187-13] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Murine cytomegalovirus encodes numerous proteins that act on a variety of pathways to modulate the innate and adaptive immune responses. Here, we demonstrate that a chemokine-like protein encoded by murine cytomegalovirus activates the early innate immune response and delays adaptive immunity, thereby impairing viral clearance. The protein, m131/129 (also known as MCK-2), is not required to establish infection in the spleen; however, a mutant virus lacking m131/129 was cleared more rapidly from this organ. In the absence of m131/129 expression, there was enhanced activation of dendritic cells (DC), and virus-specific CD8(+) T cells were recruited into the immune response earlier. Viral mutants lacking m131/129 elicited weaker production of alpha interferon (IFN-α) at 40 h postinfection, indicating that this protein exerts its effects during early rounds of viral replication in the spleen. Furthermore, while wild-type and mutant viruses activated plasmacytoid dendritic cells (pDC) equally at this time, as measured by the upregulation of costimulatory molecules, the presence of m131/129 stimulated more pDC to secrete IFN-α, accounting for the stronger IFN-α response than from the wild-type virus. These data provide evidence for a novel immunomodulatory function of a viral chemokine and expose the multifunctionality of immune evasion proteins. In addition, these results broaden our understanding of the interplay between innate and adaptive immunity.
Collapse
|
42
|
Fleming P, Kvansakul M, Voigt V, Kile BT, Kluck RM, Huang DCS, Degli-Esposti MA, Andoniou CE. MCMV-mediated inhibition of the pro-apoptotic Bak protein is required for optimal in vivo replication. PLoS Pathog 2013; 9:e1003192. [PMID: 23468630 PMCID: PMC3585157 DOI: 10.1371/journal.ppat.1003192] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Accepted: 12/28/2012] [Indexed: 01/29/2023] Open
Abstract
Successful replication and transmission of large DNA viruses such as the cytomegaloviruses (CMV) family of viruses depends on the ability to interfere with multiple aspects of the host immune response. Apoptosis functions as a host innate defence mechanism against viral infection, and the capacity to interfere with this process is essential for the replication of many viruses. The Bcl-2 family of proteins are the principle regulators of apoptosis, with two pro-apoptotic members, Bax and Bak, essential for apoptosis to proceed. The m38.5 protein encoded by murine CMV (MCMV) has been identified as Bax-specific inhibitor of apoptosis. Recently, m41.1, a protein product encoded by the m41 open reading frame (ORF) of MCMV, has been shown to inhibit Bak activity in vitro. Here we show that m41.1 is critical for optimal MCMV replication in vivo. Growth of a m41.1 mutant was attenuated in multiple organs, a defect that was not apparent in Bak−/− mice. Thus, m41.1 promotes MCMV replication by inhibiting Bak-dependent apoptosis during in vivo infection. The results show that Bax and Bak mediate non-redundant functions during MCMV infection and that the virus produces distinct inhibitors for each protein to counter the activity of these proteins. The cytomegaloviruses (CMV) are a family of viruses that establish a latent infection that lasts for the life of the host, with the virus able to reactivate when the host is immunosuppressed. We have used murine CMV (MCMV) as a model to understand how CMV interferes with the anti-viral immune response. Apoptosis, or programmed cell death, is one of the defence mechanisms used by multicellular organisms to impair viral infection. In order for viral replication to proceed, many viruses have evolved mechanisms to prevent the apoptosis of infected host cells. Under most circumstances the activation of Bax, or the closely related protein Bak, is required for apoptosis to proceed. The m41.1 protein was recently identified as a candidate Bak inhibitor during in vitro infection. We have generated a mutant virus which is unable to produce the m41.1 protein and found that growth of this virus was attenuated in wild-type mice. Importantly, growth of the mutant virus was equivalent to that of the wild-type virus in mice lacking the Bak protein. These studies establish that m41.1 is an inhibitor of Bak and that the capacity to prevent apoptosis triggered by Bak is required for efficient replication of MCMV in vivo.
Collapse
Affiliation(s)
- Peter Fleming
- Immunology and Virology Program, Centre for Ophthalmology and Visual Science, The University of Western Australia, Nedlands, Western Australia, Australia
- Centre for Experimental Immunology, Lions Eye Institute, Nedlands, Western Australia, Australia
| | - Marc Kvansakul
- Department of Biochemistry, La Trobe University, Melbourne, Victoria, Australia
| | - Valentina Voigt
- Immunology and Virology Program, Centre for Ophthalmology and Visual Science, The University of Western Australia, Nedlands, Western Australia, Australia
- Centre for Experimental Immunology, Lions Eye Institute, Nedlands, Western Australia, Australia
| | - Benjamin T. Kile
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Ruth M. Kluck
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - David C. S. Huang
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Mariapia A. Degli-Esposti
- Immunology and Virology Program, Centre for Ophthalmology and Visual Science, The University of Western Australia, Nedlands, Western Australia, Australia
- Centre for Experimental Immunology, Lions Eye Institute, Nedlands, Western Australia, Australia
| | - Christopher E. Andoniou
- Immunology and Virology Program, Centre for Ophthalmology and Visual Science, The University of Western Australia, Nedlands, Western Australia, Australia
- Centre for Experimental Immunology, Lions Eye Institute, Nedlands, Western Australia, Australia
- * E-mail:
| |
Collapse
|
43
|
Crosby LN, McCormick AL, Mocarski ES. Gene products of the embedded m41/m41.1 locus of murine cytomegalovirus differentially influence replication and pathogenesis. Virology 2013; 436:274-83. [PMID: 23295021 DOI: 10.1016/j.virol.2012.12.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Revised: 09/30/2012] [Accepted: 12/02/2012] [Indexed: 01/08/2023]
Abstract
Cytomegaloviruses utilize overlapping and embedded reading frames as a way to efficiently package and express all genes necessary to carry out a complex lifecycle. Murine cytomegalovirus encodes a mitochondrial-localized inhibitor of Bak oligomerization (vIBO) from m41.1, a reading frame that is embedded within the m41 gene. The m41.1-encoded mitochondrial protein and m41-encoded Golgi-localized protein have both been implicated in cell death suppression; however, their contribution to viral infection within the host has not been investigated. Here, we report that mitochondrial-localized m41.1 (vIBO) is required for optimal viral replication in macrophages and has a modest impact on dissemination in infected mice. In contrast, Golgi-localized m41 protein is dispensable during acute infection and dissemination as well as for latency. All together, these data indicate that the primary evolutionary focus of this locus is to maintain mitochondrial function through inhibition of Bak-mediated death pathways in support of viral pathogenesis.
Collapse
Affiliation(s)
- Lynsey N Crosby
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | |
Collapse
|
44
|
Natural killer cell dependent within-host competition arises during multiple MCMV infection: consequences for viral transmission and evolution. PLoS Pathog 2013; 9:e1003111. [PMID: 23300458 PMCID: PMC3536701 DOI: 10.1371/journal.ppat.1003111] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Accepted: 11/18/2012] [Indexed: 11/18/2022] Open
Abstract
It is becoming increasingly clear that many diseases are the result of infection from multiple genetically distinct strains of a pathogen. Such multi-strain infections have the capacity to alter both disease and pathogen dynamics. Infection with multiple strains of human cytomegalovirus (HCMV) is common and has been linked to enhanced disease. Suggestions that disease enhancement in multi-strain infected patients is due to complementation have been supported by trans-complementation studies in mice during co-infection of wild type and gene knockout strains of murine CMV (MCMV). Complementation between naturally circulating strains of CMV has, however, not been assessed. In addition, many models of multi-strain infection predict that co-infecting strains will compete with each other and that this competition may contribute to selective transmission of more virulent pathogen strains. To assess the outcome of multi-strain infection, C57BL/6 mice were infected with up to four naturally circulating strains of MCMV. In this study, profound within-host competition was observed between co-infecting strains of MCMV. This competition was MCMV strain specific and resulted in the complete exclusion of certain strains of MCMV from the salivary glands of multi-strain infected mice. Competition was dependent on Ly49H+ natural killer (NK) cells as well as the expression of the ligand for Ly49H, the MCMV encoded product, m157. Strains of MCMV which expressed an m157 gene product capable of ligating Ly49H were outcompeted by strains of MCMV expressing variant m157 genes. Importantly, within-host competition prevented the shedding of the less virulent strains of MCMV, those recognized by Ly49H, into the saliva of multi-strain infected mice. These data demonstrate that NK cells have the strain specific recognition capacity required to meditate within-host competition between strains of MCMV. Furthermore, this within-host competition has the capacity to shape the dynamics of viral shedding and potentially select for the transmission of more virulent virus strains. Infection of the host with multiple strains of a pathogen is common and occurs with the herpesvirus, human cytomegalovirus (HCMV). However the effects of multi-strain infection on the host and the pathogen remain poorly studied. Here we show, in a mouse model, that infection of C57BL/6 mice with multiple strains of murine CMV (MCMV) results in profound within-host competition. Competition between the strains of MCMV is dependent on Ly49H+ natural killer (NK) cells. The NK cell activation receptor Ly49H receptor targets certain genotypes of the viral protein, m157. During multi-strain infection, strains of MCMV encoding an m157 capable of binding Ly49H are excluded from the salivary gland and the saliva of C57BL/6 mice, allowing for the shedding of only non-Ly49H binding strains of MCMV in the saliva. This within-host competition could therefore have significant impacts on the circulation of MCMV strains, as only the most virulent MCMV strains were present in the saliva.
Collapse
|
45
|
Cytomegalovirus impairs antiviral CD8+ T cell immunity by recruiting inflammatory monocytes. Immunity 2012; 37:122-33. [PMID: 22840843 DOI: 10.1016/j.immuni.2012.04.014] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2011] [Revised: 04/06/2012] [Accepted: 04/19/2012] [Indexed: 12/24/2022]
Abstract
Inflammatory monocytes are key early responders to infection that contribute to pathogen-host interactions in diverse ways. Here, we report that the murine cytomegalovirus-encoded CC chemokine, MCK2, enhanced CCR2-dependent recruitment of these cells to modulate antiviral immunity, impairing virus-specific CD8(+) T cell expansion and differentiation into effector cytotoxic T lymphocytes, thus reducing the capacity to eliminate viral antigen-bearing cells and slowing viral clearance. Adoptive transfer of inflammatory monocytes into Ccr2(-/-)Ccl2(-/-) mice impaired virus antigen-specific clearance. Cytomegalovirus therefore enhances a natural CCR2-dependent immune regulatory network to modulate adaptive immunity via nitric oxide production, reminiscent of the monocytic subtype of myeloid-derived suppressor cells primarily implicated in cancer immunomodulation.
Collapse
|
46
|
Abstract
Cytomegaloviruses manipulate the host chemokine/receptor axis by altering cellular chemokine expression and by encoding multiple chemokines and chemokine receptors. Similar to human cytomegalovirus (HCMV), rat cytomegalovirus (RCMV) encodes multiple CC chemokine-analogous proteins, including r129 (HCMV UL128 homologue) and r131 (HCMV UL130 and MCMV m129/130 homologues). Although these proteins play a role in CMV entry, their function as chemotactic cytokines remains unknown. In the current study, we examined the role of the RCMV chemokine r129 in promoting cellular migration and in accelerating transplant vascular sclerosis (TVS) in our rat heart transplant model. We determined that r129 protein is released into culture supernatants of infected cells and is expressed with late viral gene kinetics during RCMV infection and highly expressed in heart and salivary glands during in vivo rat infections. Using the recombinant r129 protein, we demonstrated that r129 induces migration of lymphocytes isolated from rat peripheral blood, spleen, and bone marrow and from a rat macrophage cell line. Using antibody-mediated cell sorting of rat splenocytes, we demonstrated that r129 induces migration of naïve/central memory CD4(+) T cells. Through ligand-binding assays, we determined that r129 binds rat CC chemokine receptors CCR3, CCR4, CCR5, and CCR7. In addition, mutational analyses identified functional domains of r129 resulting in recombinant proteins that fail to induce migration (r129-ΔNT and -C31A) or alter the chemotactic ability of the chemokine (r129-F43A). Two of the mutant proteins (r129-C31A and -ΔNT) also act as dominant negatives by inhibiting migration induced by wild-type r129. Furthermore, infection of rat heart transplant recipients with RCMV containing the r129-ΔNT mutation prevented CMV-induced acceleration of TVS. Together our findings indicate that RCMV r129 is highly chemotactic, which has important implications during RCMV infection and reactivation and acceleration of TVS.
Collapse
|
47
|
Chan G, Nogalski MT, Stevenson EV, Yurochko AD. Human cytomegalovirus induction of a unique signalsome during viral entry into monocytes mediates distinct functional changes: a strategy for viral dissemination. J Leukoc Biol 2012; 92:743-52. [PMID: 22715139 DOI: 10.1189/jlb.0112040] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
HCMV pathogenesis is a direct consequence of the hematogenous dissemination of the virus to multiple host organ sites. The presence of infected monocytes in the peripheral blood and organs of individuals exhibiting primary HCMV infection have long suggested that these blood sentinels are responsible for mediating viral spread. Despite monocytes being "at the right place at the right time", their short lifespan and the lack of productive viral infection in these cells complicate this scenario of a monocyte-driven approach to viral dissemination by HCMV. However, our laboratory has provided evidence that HCMV infection is able to induce a highly controlled polarization of monocytes toward a unique and long-lived proinflammatory macrophage, which we have demonstrated to be permissive for viral replication. These observations suggest that HCMV has evolved as a distinct mechanism to induce select proinflammatory characteristics that provide infected monocytes with the necessary tools to mediate viral spread following a primary infection. In the absence of viral gene products during the early stages of infection, the process by which HCMV "tunes" the inflammatory response in infected monocytes to promote viral spread and subsequently, viral persistence remains unclear. In this current review, we focus on the viral entry process of HCMV and the potential role of receptor-ligand interactions in modulating monocyte biology. Specifically, we examine the signaling pathways initiated by the distinct combination of cellular receptors simultaneously engaged and activated by HCMV during viral entry and how the acquisition of this distinct signalsome results in a nontraditional activation of monocytes leading to the induction of the unique, functional attributes observed in monocytes following HCMV infection.
Collapse
Affiliation(s)
- Gary Chan
- Department of Microbiology and Immunology, Center for Molecular and Tumor Virology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | | | | | | |
Collapse
|
48
|
Engel P, Angulo A. Viral Immunomodulatory Proteins: Usurping Host Genes as a Survival Strategy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 738:256-76. [DOI: 10.1007/978-1-4614-1680-7_15] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
49
|
Shedding light on the elusive role of endothelial cells in cytomegalovirus dissemination. PLoS Pathog 2011; 7:e1002366. [PMID: 22114552 PMCID: PMC3219709 DOI: 10.1371/journal.ppat.1002366] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 09/22/2011] [Indexed: 11/19/2022] Open
Abstract
Cytomegalovirus (CMV) is frequently transmitted by solid organ transplantation and is associated with graft failure. By forming the boundary between circulation and organ parenchyma, endothelial cells (EC) are suited for bidirectional virus spread from and to the transplant. We applied Cre/loxP-mediated green-fluorescence-tagging of EC-derived murine CMV (MCMV) to quantify the role of infected EC in transplantation-associated CMV dissemination in the mouse model. Both EC- and non-EC-derived virus originating from infected Tie2-cre(+) heart and kidney transplants were readily transmitted to MCMV-naïve recipients by primary viremia. In contrast, when a Tie2-cre(+) transplant was infected by primary viremia in an infected recipient, the recombined EC-derived virus poorly spread to recipient tissues. Similarly, in reverse direction, EC-derived virus from infected Tie2-cre(+) recipient tissues poorly spread to the transplant. These data contradict any privileged role of EC in CMV dissemination and challenge an indiscriminate applicability of the primary and secondary viremia concept of virus dissemination.
Collapse
|
50
|
Virus progeny of murine cytomegalovirus bacterial artificial chromosome pSM3fr show reduced growth in salivary Glands due to a fixed mutation of MCK-2. J Virol 2011; 85:10346-53. [PMID: 21813614 DOI: 10.1128/jvi.00545-11] [Citation(s) in RCA: 119] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Murine cytomegalovirus (MCMV) Smith strain has been cloned as a bacterial artificial chromosome (BAC) named pSM3fr and used for analysis of virus gene functions in vitro and in vivo. When sequencing the complete BAC genome, we identified a frameshift mutation within the open reading frame (ORF) encoding MCMV chemokine homologue MCK-2. This mutation would result in a truncated MCK-2 protein. When mice were infected with pSM3fr-derived virus, we observed reduced virus production in salivary glands, which could be reverted by repair of the frameshift mutation. When looking for the source of the mutation, we consistently found that virus stocks of cell culture-passaged MCMV Smith strain are mixtures of viruses with or without the MCK-2 mutation. We conclude that the MCK-2 mutation in the pSM3fr BAC is the result of clonal selection during the BAC cloning procedure.
Collapse
|