1
|
Mahmud J, Geiler BW, Biswas J, Miller MJ, Myers JE, Matthews SM, Wass AB, O’Connor CM, Chan GC. Delivery of US28 by incoming HCMV particles rapidly attenuates Akt activity to suppress HCMV lytic replication in monocytes. Sci Signal 2024; 17:eadn8727. [PMID: 39190708 PMCID: PMC11460310 DOI: 10.1126/scisignal.adn8727] [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: 01/04/2024] [Accepted: 08/08/2024] [Indexed: 08/29/2024]
Abstract
Establishing a nonproductive, quiescent infection within monocytes is essential for the spread of human cytomegalovirus (HCMV). We investigated the mechanisms through which HCMV establishes a quiescent infection in monocytes. US28 is a virally encoded G protein-coupled receptor (GPCR) that is essential for silent infections within cells of the myeloid lineage. We found that preformed US28 was rapidly delivered to monocytes by HCMV viral particles, whereas the de novo synthesis of US28 was delayed for several days. A recombinant mutant virus lacking US28 (US28Δ) was unable to establish a quiescent infection, resulting in a fully productive lytic infection able to produce progeny virus. Infection with US28Δ HCMV resulted in the phosphorylation of the serine and threonine kinase Akt at Ser473 and Thr308, in contrast with the phosphorylation of Akt only at Ser473 after WT viral infection. Inhibiting the dual phosphorylation of Akt prevented the lytic replication of US28Δ, and ectopic expression of a constitutively phosphorylated Akt variant triggered lytic replication of wild-type HCMV. Mechanistically, we found that US28 was necessary and sufficient to attenuate epidermal growth factor receptor (EGFR) signaling induced during the entry of WT virus, which led to the site-specific phosphorylation of Akt at Ser473. Thus, particle-delivered US28 fine-tunes Akt activity by limiting HCMV-induced EGFR activation during viral entry, enabling quiescent infection in monocytes.
Collapse
Affiliation(s)
- Jamil Mahmud
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY 13210
| | - Brittany W. Geiler
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY 13210
| | - Juthi Biswas
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY 13210
| | - Michael J. Miller
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY 13210
| | - Julia E. Myers
- Infection Biology, Lerner Research Institute, Sheikha Fatima bint Mubarak Global Center for Pathogen & Human Health Research, Cleveland Clinic, Cleveland, OH 44195
- Case Comprehensive Cancer Center, Cleveland, OH 44106
| | - Stephen M. Matthews
- Infection Biology, Lerner Research Institute, Sheikha Fatima bint Mubarak Global Center for Pathogen & Human Health Research, Cleveland Clinic, Cleveland, OH 44195
- Case Comprehensive Cancer Center, Cleveland, OH 44106
| | - Amanda B. Wass
- Infection Biology, Lerner Research Institute, Sheikha Fatima bint Mubarak Global Center for Pathogen & Human Health Research, Cleveland Clinic, Cleveland, OH 44195
- Case Comprehensive Cancer Center, Cleveland, OH 44106
| | - Christine M. O’Connor
- Infection Biology, Lerner Research Institute, Sheikha Fatima bint Mubarak Global Center for Pathogen & Human Health Research, Cleveland Clinic, Cleveland, OH 44195
- Case Comprehensive Cancer Center, Cleveland, OH 44106
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH 44195
| | - Gary C. Chan
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY 13210
| |
Collapse
|
2
|
Mahmud J, Geiler BW, Biswas J, Miller MJ, Myers JE, Matthews SM, Wass AB, O'Connor CM, Chan GC. Virion-associated US28 rapidly modulates Akt activity to suppress HCMV lytic replication in monocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.05.556359. [PMID: 37732204 PMCID: PMC10508783 DOI: 10.1101/2023.09.05.556359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Establishing a non-productive quiescent/silent infection within monocytes is essential for spread of human cytomegalovirus (HCMV). Yet, how HCMV establishes a quiescent infection in monocytes remains unclear. US28 is a viral G protein-coupled receptor (GPCR) essential for silent infections within cells of the myeloid lineage. We found virion-associated US28 was rapidly delivered to monocytes, while de novo synthesized US28 was delayed for several days. A recombinant mutant virus lacking US28 (US28Δ) was unable to establish a quiescent infection, resulting in a fully productive lytic replication cycle. Mechanistically, viral entry of US28Δ phosphorylated Akt at both serine 473 (S473) and threonine 308 (T308), which contrasted with the site-specific phosphorylation of Akt at S473 following WT infection. Preventing Akt bi-phosphorylation prevented lytic replication of US28Δ, and ectopic expression of a constitutively phosphorylated Akt variant triggered lytic replication of WT infection. Our data demonstrate that virion-delivered US28 fine-tunes Akt activity to permit HCMV infection to enter a quiescent state following primary infection of monocytes.
Collapse
|
3
|
Luganini A, Serra V, Scarpellino G, Bhat SM, Munaron L, Fiorio Pla A, Gribaudo G. The US21 viroporin of human cytomegalovirus stimulates cell migration and adhesion. mBio 2023; 14:e0074923. [PMID: 37477430 PMCID: PMC10470750 DOI: 10.1128/mbio.00749-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/02/2023] [Indexed: 07/22/2023] Open
Abstract
The human cytomegalovirus (HCMV) US12 gene family contributes to virus-host interactions by regulating the virus' cell tropism and its evasion of host innate immune responses. US21, one of the 10 US12 genes (US12-US21), is a descendant of a captured cellular transmembrane BAX inhibitor motif-containing gene. It encodes a 7TMD endoplasmic reticulum (ER)-resident viroporin (pUS21) capable of reducing the Ca2+ content of ER stores, which, in turn, protects cells against apoptosis. Since regulation of Ca2+ homeostasis affects a broad range of cellular responses, including cell motility, we investigated whether pUS21 might also interfere with this cytobiological consequence of Ca2+ signaling. Indeed, deletion of the US21 gene impaired the ability of HCMV-infected cells to migrate, whereas expression of US21 protein stimulated cell migration and adhesion, as well as focal adhesion (FA) dynamics, in a way that depended on its ability to manipulate ER Ca2+ content. Mechanistic studies revealed pUS21-mediated cell migration to involve calpain 2 activation since its inhibition prevented the viroporin's effects on cell motility. Pertinently, pUS21 expression stimulated a store-operated Ca2+ entry (SOCE) mechanism that may determine the activation of calpain 2 by promoting Ca2+ entry. Furthermore, pUS21 was observed to interact with talin-1, a calpain 2 substrate, and crucial protein component of FA complexes. A functional consequence of this interaction was confirmed by talin-1 knockdown, which abrogated the pUS21-mediated increase in cell migration. Together, these results indicate the US21-encoded viroporin to be a viral regulator of cell adhesion and migration in the context of HCMV infection. IMPORTANCE Human cytomegalovirus (HCMV) is an opportunistic pathogen that owes part of its success to the capture, duplication, and tuning of cellular genes to generate modern viral proteins which promote infection and persistence in the host by interfering with many cell biochemical and physiological pathways. The US21 viral protein provides an example of this evolutionary strategy: it is a cellular-derived calcium channel that manipulates intracellular calcium homeostasis to confer edges to HCMV replication. Here, we report on the characterization of a novel function of the US21 protein as a viral regulator of cell migration and adhesion through mechanisms involving its calcium channel activity. Characterization of HCMV multifunctional regulatory proteins, like US21, supports the better understanding of viral pathogenesis and may open avenues for the design of new antiviral strategies that exploit their functions.
Collapse
Affiliation(s)
- Anna Luganini
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
| | - Valentina Serra
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
| | - Giorgia Scarpellino
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
| | - Shree Madhu Bhat
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
| | - Luca Munaron
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
| | - Alessandra Fiorio Pla
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
| | - Giorgio Gribaudo
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
| |
Collapse
|
4
|
Abstract
While many viral infections are limited and eventually resolved by the host immune response or by death of the host, other viruses establish long-term relationships with the host by way of a persistent infection, that range from chronic viruses that may be eventually cleared to those that establish life-long persistent or latent infection. Viruses infecting hosts from bacteria to humans establish quiescent infections that must be reactivated to produce progeny. For mammalian viruses, most notably herpesviruses, this quiescent maintenance of viral genomes in the absence of virus replication is referred to as latency. The latent strategy allows the virus to persist quiescently within a single host until conditions indicate a need to reactivate to reach a new host or, to re-seed a reservoir within the host. Here, I review common themes in viral strategies to regulate the latent cycle and reactivate from it ranging from bacteriophage to herpesviruses with a focus on human cytomegalovirus (HCMV). Themes central to herpesvirus latency include, epigenetic repression of viral gene expression and mechanisms to regulate host signaling and survival. Critical to the success of a latent program are mechanisms by which the virus can "sense" fluctuations in host biology (within the host) or environment (outside the host) and make appropriate "decisions" to maintain latency or re-initiate the replicative program. The signals or environments that indicate the establishment of a latent state, the very nature of the latent state, as well as the signals driving reactivation have been topics of intense study from bacteriophage to human viruses, as these questions encompass the height of complexity in virus-host interactions-where the host and the virus coexist.
Collapse
Affiliation(s)
- Felicia Goodrum
- Department of Immunobiology, BIO5 Institute, University of Arizona, Tucson, AZ, United States.
| |
Collapse
|
5
|
Mavri M, Kubale V, Depledge DP, Zuo J, Huang CA, Breuer J, Vrecl M, Jarvis MA, Jovičić EJ, Petan T, Ehlers B, Rosenkilde MM, Spiess K. Epstein-Barr Virus-Encoded BILF1 Orthologues From Porcine Lymphotropic Herpesviruses Display Common Molecular Functionality. Front Endocrinol (Lausanne) 2022; 13:862940. [PMID: 35721730 PMCID: PMC9204316 DOI: 10.3389/fendo.2022.862940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Infection of immunosuppressed transplant patients with the human γ-herpesvirus Epstein-Barr virus (EBV) is associated with post-transplant lymphoproliferative disease (PTLD), an often fatal complication. Immunosuppressed miniature pigs infected with γ-herpesvirus porcine lymphotropic herpesvirus 1 (PLHV1) develop a similar disease, identifying pigs as a potential preclinical model for PTLD in humans. BILF1 is a G protein-coupled receptor (GPCR) encoded by EBV with constitutive activity linked to tumorigenesis and immunoevasive function downregulating MHC-I. In the present study, we compared BILF1-orthologues encoded by the three known PLHVs (PLHV1-3) with EBV-BILF1 to determine pharmacological suitability of BILF1 orthologues as model system to study EBV-BILF1 druggability. Cell surface localization, constitutive internalization, and MHC-I downregulation as well as membrane proximal constitutive Gαi signaling patterns were conserved across all BILFs. Only subtle differences between the individual BILFs were observed in downstream transcription factor activation. Using Illumina sequencing, PLHV1 was observed in lymphatic tissue from PTLD-diseased, but not non-diseased pigs. Importantly, these tissues showed enhanced expression of PLHV1-BILF1 supporting its involvement in PTLD infection.
Collapse
Affiliation(s)
- Maša Mavri
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Ljubljana, Slovenia
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Valentina Kubale
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Daniel P. Depledge
- Department of Medicine, New York University School of Medicine, New York, NY, United States
| | - Jianmin Zuo
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Christene A. Huang
- Department of Surgery, Division of Plastic & Reconstructive Surgery, Division of Transplant Surgery, Anschutz Medical Campus, University of Colorado, Denver, CO, United States
| | - Judith Breuer
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Milka Vrecl
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Michael A. Jarvis
- The Vaccine Group Ltd, Plymouth; and the University of Plymouth, Plymouth, United Kingdom
| | - Eva Jarc Jovičić
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Toni Petan
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Bernhard Ehlers
- Division 12, Measles, Mumps, Rubella, and Viruses Affecting Immunocompromised Patients, Robert Koch Institute, Berlin, Germany
| | - Mette M. Rosenkilde
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- *Correspondence: Katja Spiess, ; ; Mette M. Rosenkilde,
| | - Katja Spiess
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- *Correspondence: Katja Spiess, ; ; Mette M. Rosenkilde,
| |
Collapse
|
6
|
Hanka I, Stamminger T, Ramsperger-Gleixner M, Kuckhahn AV, Müller R, Weyand M, Heim C. Role of CMV chemokine receptor M33 in airway graft rejection in a mouse transplant model. Transpl Immunol 2021; 67:101415. [PMID: 34033867 DOI: 10.1016/j.trim.2021.101415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/20/2021] [Accepted: 05/20/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Cytomegalovirus (CMV) infection is a risk factor for bronchiolitis obliterans (BO), one form of chronic lung allograft dysfunction (CLAD). The viral chemokine receptor M33 is essential for successful spread of murine CMV to host salivary glands. In the present study we investigated the impact of M33 on chronic airway rejection. METHODS MHC I-mismatched tracheas of C·B10-H2b/LilMcdJ mice were transplanted into BALB/c (H2d) recipients and infected at different dates with wild type (WT) or M33-deleted (delM33) MCMV representing clinical settings of viral recipient (R)-donor (D)-serostatus: (D-/R+) or (D+/R-). Grafts were recovered for gene expression and histological / immunofluorescence analysis, respectively. RESULTS Evaluations showed significantly increased signs of chronic rejection in WT-infected mice compared to uninfected allografts seen in lower epithelium/lamina propria-ratio (ELR) (ELR 0.46 ± 0.07 [WT post] vs. ELR 0.66 ± 0.10 [non-inf.]; p < 0.05). The rejection in delM33-infected groups was significantly reduced vs. WT-infected groups (0.67 ± 0.04 [delM33 post]; vs. WT post p < 0.05). Furthermore, decreased rejection was observed in WT pre-infected compared to post-infected groups (0.56 ± 0.08 [WT pre]; vs. WT post p < 0.05). CD8+ T cell infiltration was significantly higher in WT-post compared to the delM33 infected or non-infected allografts. CONCLUSIONS These data support the role of the CMV in accelerating CLAD. The deletion of chemokine receptor M33 leads to attenuated rejection.
Collapse
Affiliation(s)
- Isabella Hanka
- Department of Cardiac Surgery, Friedrich-Alexander University Erlangen-Nürnberg, Krankenhausstaße 12, 91054 Erlangen, Germany
| | - Thomas Stamminger
- Institute for Virology, University of Ulm, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Martina Ramsperger-Gleixner
- Department of Cardiac Surgery, Friedrich-Alexander University Erlangen-Nürnberg, Krankenhausstaße 12, 91054 Erlangen, Germany
| | - Annika V Kuckhahn
- Department of Cardiac Surgery, Friedrich-Alexander University Erlangen-Nürnberg, Krankenhausstaße 12, 91054 Erlangen, Germany
| | - Regina Müller
- Institute of Clinical and Molecular Virology, Friedrich-Alexander University Erlangen-Nürnberg, Schlossgarten 4, 91054 Erlangen, Germany
| | - Michael Weyand
- Department of Cardiac Surgery, Friedrich-Alexander University Erlangen-Nürnberg, Krankenhausstaße 12, 91054 Erlangen, Germany
| | - Christian Heim
- Department of Cardiac Surgery, Friedrich-Alexander University Erlangen-Nürnberg, Krankenhausstaße 12, 91054 Erlangen, Germany.
| |
Collapse
|
7
|
Vasilieva E, Gianella S, Freeman ML. Novel Strategies to Combat CMV-Related Cardiovascular Disease. Pathog Immun 2020; 5:240-274. [PMID: 33089035 PMCID: PMC7556413 DOI: 10.20411/pai.v5i1.382] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 06/08/2020] [Indexed: 12/20/2022] Open
Abstract
Cytomegalovirus (CMV), a ubiquitous human pathogen that is never cleared from the host, has long been thought to be relatively innocuous in immunocompetent adults, but causes severe complications including blindness, end-organ disease, and death in newborns and in immuno-compromised individuals, such as organ transplant recipients and those suffering from AIDS. Yet even in persons with intact immunity, CMV infection is associated with profound stimulation of immune and inflammatory pathways. Carriers of CMV infection also have an elevated risk of developing cardiovascular complications. In this review, we define the proposed mechanisms of how CMV contributes to cardiovascular disease (CVD), describe current approaches to target CMV, and discuss how these strategies may or may not alleviate cardiovascular complications in those with CMV infection. In addition, we discuss the special situation of CMV coinfection in people with HIV infection receiving antiretroviral therapy, and describe how these 2 viral infections may interact to potentiate CVD in this especially vulnerable population.
Collapse
Affiliation(s)
- Elena Vasilieva
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow 127473, Russia
| | - Sara Gianella
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Michael L. Freeman
- Division of Infectious Diseases and HIV Medicine; Department of Medicine; Case Western Reserve University, Cleveland, Ohio, United States
| |
Collapse
|
8
|
Plouffe B, Thomsen ARB, Irannejad R. Emerging Role of Compartmentalized G Protein-Coupled Receptor Signaling in the Cardiovascular Field. ACS Pharmacol Transl Sci 2020; 3:221-236. [PMID: 32296764 PMCID: PMC7155194 DOI: 10.1021/acsptsci.0c00006] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Indexed: 02/06/2023]
Abstract
G protein-coupled receptors (GPCRs) are cell surface receptors that for many years have been considered to function exclusively at the plasma membrane, where they bind to extracellular ligands and activate G protein signaling cascades. According to the conventional model, these signaling events are rapidly terminated by β-arrestin (β-arr) recruitment to the activated GPCR resulting in signal desensitization and receptor internalization. However, during the past decade, emerging evidence suggest that many GPCRs can continue to activate G proteins from intracellular compartments after they have been internalized. G protein signaling from intracellular compartments is in general more sustained compared to G protein signaling at the plasma membrane. Notably, the particular location closer to the nucleus is beneficial for selective cellular functions such as regulation of gene transcription. Here, we review key GPCRs that undergo compartmentalized G protein signaling and discuss molecular considerations and requirements for this signaling to occur. Our main focus will be on receptors involved in the regulation of important physiological and pathological cardiovascular functions. We also discuss how sustained G protein activation from intracellular compartments may be involved in cellular functions that are distinct from functions regulated by plasma membrane G protein signaling, and the corresponding significance in cardiovascular physiology.
Collapse
Affiliation(s)
- Bianca Plouffe
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast BT9 7BL, United Kingdom
| | - Alex R B Thomsen
- Department of Basic Science and Craniofacial Biology, NYU College of Dentistry, New York, New York 10010, United States
| | - Roshanak Irannejad
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, California 94158, United States
| |
Collapse
|
9
|
Min CK, Shakya AK, Lee BJ, Streblow DN, Caposio P, Yurochko AD. The Differentiation of Human Cytomegalovirus Infected-Monocytes Is Required for Viral Replication. Front Cell Infect Microbiol 2020; 10:368. [PMID: 32850474 PMCID: PMC7411144 DOI: 10.3389/fcimb.2020.00368] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 06/15/2020] [Indexed: 12/24/2022] Open
Abstract
Viral dissemination is a key mechanism responsible for persistence and disease following human cytomegalovirus (HCMV) infection. Monocytes play a pivotal role in viral dissemination to organ tissue during primary infection and following reactivation from latency. For example, during primary infection, infected monocytes migrate into tissues and differentiate into macrophages, which then become a source of viral replication. In addition, because differentiated macrophages can survive for months to years, they provide a potential persistent infection source in various organ systems. We broadly note that there are three phases to infection and differentiation of HCMV-infected monocytes: (1) Virus enters and traffics to the nucleus through a virus receptor ligand engagement event that activates a unique signalsome that initiates the monocyte-to-macrophage differentiation process. (2) Following initial infection, HCMV undergoes a "quiescence-like state" in monocytes lasting for several weeks and promotes monocyte differentiation into macrophages. While, the initial event is triggered by the receptor-ligand engagement, the long-term cellular activation is maintained by chronic viral-mediated signaling events. (3) Once HCMV infected monocytes differentiate into macrophages, the expression of immediate early viral (IE) genes is detectable, followed by viral replication and long term infectious viral particles release. Herein, we review the detailed mechanisms of each phase during infection and differentiation into macrophages and discuss the biological significance of the differentiation of monocytes in the pathogenesis of HCMV.
Collapse
Affiliation(s)
- Chan-Ki Min
- Department of Microbiology and Immunology, Center for Molecular and Tumor Virology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Akhalesh K Shakya
- Department of Microbiology and Immunology, Center for Molecular and Tumor Virology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Byeong-Jae Lee
- Department of Microbiology and Immunology, Center for Molecular and Tumor Virology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Daniel N Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR, United States
| | - Patrizia Caposio
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, OR, United States
| | - Andrew D Yurochko
- Department of Microbiology and Immunology, Center for Molecular and Tumor Virology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
- Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
- Center of Excellence in Arthritis and Rheumatology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| |
Collapse
|
10
|
Paradowska E, Jabłońska A, Studzińska M, Wilczyński M, Wilczyński JR. Detection and genotyping of CMV and HPV in tumors and fallopian tubes from epithelial ovarian cancer patients. Sci Rep 2019; 9:19935. [PMID: 31882737 PMCID: PMC6934444 DOI: 10.1038/s41598-019-56448-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 12/11/2019] [Indexed: 01/02/2023] Open
Abstract
Viral and bacterial infections are detected in epithelial ovarian cancer (EOC) tissues. Since the fallopian tubes are often affected by pelvic inflammatory disease (PID) and the majority of serous EOCs appear to originate from dysplastic lesions in the distal tube, it is relevant to consider the potential role that infectious agents may play in ovarian carcinogenesis. We sought to analyze the prevalence of human papillomavirus (HPV) and cytomegalovirus (CMV) in EOC tissue and fallopian tube specimens obtained at tumor resection. Ovarian cancer and fallopian tube tissue samples obtained from patients with EOC were analyzed by both qualitative and quantitative PCR to detect and quantify viral DNA. The presence of CMV and HPV DNA was detected in 70% and 74% cancerous ovarian tissues, respectively, and was significantly higher in EOC than in benign tumor cases (P ≤ 0.01). CMV or HPV infection was observed also in the fallopian tube samples. Infection with HPV16 was determined in 70% of EOC cases. Almost two thirds of EOC patients demonstrated coinfection with CMV and HPV in the pathological samples. The results revealed that the presence of CMV and HPV in EOC samples is common. CMV and HPV infections can be potential risks for EOC development.
Collapse
Affiliation(s)
- Edyta Paradowska
- Laboratory of Virology, Institute of Medical Biology of the Polish Academy of Sciences, Lodz, Poland.
| | - Agnieszka Jabłońska
- Laboratory of Virology, Institute of Medical Biology of the Polish Academy of Sciences, Lodz, Poland
| | - Mirosława Studzińska
- Laboratory of Virology, Institute of Medical Biology of the Polish Academy of Sciences, Lodz, Poland
| | - Miłosz Wilczyński
- Department of Surgical, Endoscopic and Oncological Gynecology, Polish Mother's Health Center Research Institute, Lodz, Poland
| | - Jacek R Wilczyński
- Department of Surgical and Oncological Gynecology, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
11
|
Crawford LB, Caposio P, Kreklywich C, Pham AH, Hancock MH, Jones TA, Smith PP, Yurochko AD, Nelson JA, Streblow DN. Human Cytomegalovirus US28 Ligand Binding Activity Is Required for Latency in CD34 + Hematopoietic Progenitor Cells and Humanized NSG Mice. mBio 2019; 10:e01889-19. [PMID: 31431555 PMCID: PMC6703429 DOI: 10.1128/mbio.01889-19] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 07/22/2019] [Indexed: 01/13/2023] Open
Abstract
Human cytomegalovirus (HCMV) infection of CD34+ hematopoietic progenitor cells (CD34+ HPCs) provides a critical reservoir of virus in stem cell transplant patients, and viral reactivation remains a significant cause of morbidity and mortality. The HCMV chemokine receptor US28 is implicated in the regulation of viral latency and reactivation. To explore the role of US28 signaling in latency and reactivation, we analyzed protein tyrosine kinase signaling in CD34+ HPCs expressing US28. US28-ligand signaling in CD34+ HPCs induced changes in key regulators of cellular activation and differentiation. In vitro latency and reactivation assays utilizing CD34+ HPCs indicated that US28 was required for viral reactivation but not latency establishment or maintenance. Similarly, humanized NSG mice (huNSG) infected with TB40E-GFP-US28stop failed to reactivate upon treatment with granulocyte-colony-stimulating factor, but viral genome levels were maintained. Interestingly, HCMV-mediated changes in hematopoiesis during latency in vivo and in vitro was also dependent upon US28, as US28 directly promoted differentiation toward the myeloid lineage. To determine whether US28 constitutive activity and/or ligand-binding activity were required for latency and reactivation, we infected both huNSG mice and CD34+ HPCs in vitro with HCMV TB40E-GFP containing the US28-R129A mutation (no CA) or Y16F mutation (no ligand binding). TB40E-GFP-US28-R129A was maintained during latency and exhibited normal reactivation kinetics. In contrast, TB40E-GFP-US28-Y16F exhibited high levels of viral genome during latency and reactivation, indicating that the virus did not establish latency. These data indicate that US28 is necessary for viral reactivation and ligand binding activity is required for viral latency, highlighting the complex role of US28 during HCMV latency and reactivation.IMPORTANCE Human cytomegalovirus (HCMV) can establish latency following infection of CD34+ hematopoietic progenitor cells (HPCs), and reactivation from latency is a significant cause of viral disease and accelerated graft failure in bone marrow and solid-organ transplant patients. The precise molecular mechanisms of HCMV infection in HPCs are not well defined; however, select viral gene products are known to regulate aspects of latency and reactivation. The HCMV-encoded chemokine receptor US28, which binds multiple CC chemokines as well as CX3CR1, is expressed both during latent and lytic phases of the virus life cycle and plays a role in latency and reactivation. However, the specific timing of US28 expression and the role of ligand binding in these processes are not well defined. In this report, we determined that US28 is required for reactivation but not for maintaining latency. However, when present during latency, US28 ligand binding activity is critical to maintaining the virus in a quiescent state. We attribute the regulation of both latency and reactivation to the role of US28 in promoting myeloid lineage cell differentiation. These data highlight the dynamic and multifunctional nature of US28 during HCMV latency and reactivation.
Collapse
Affiliation(s)
- Lindsey B Crawford
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Patrizia Caposio
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Craig Kreklywich
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Andrew H Pham
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Meaghan H Hancock
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Taylor A Jones
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Patricia P Smith
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Andrew D Yurochko
- Department of Microbiology and Immunology, Louisiana State University at Shreveport, Shreveport, Louisiana, USA
| | - Jay A Nelson
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Daniel N Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| |
Collapse
|
12
|
Abstract
Human cytomegalovirus (HCMV) latency and reactivation is regulated by the chromatin structure at the major immediate early promoter (MIEP) within myeloid cells. Both cellular and viral factors are known to control this promoter during latency, here we will review the known mechanisms for MIEP regulation during latency. We will then focus on the virally encoded G-protein coupled receptor, US28, which suppresses the MIEP in early myeloid lineage cells. The importance of this function is underlined by the fact that US28 is essential for HCMV latency in CD34+ progenitor cells and CD14+ monocytes. We will describe cellular signalling pathways modulated by US28 to direct MIEP suppression during latency and demonstrate how US28 is able to ‘regulate the regulators’ of HCMV latency. Finally, we will describe how cell-surface US28 can be a target for antiviral therapies directed at the latent viral reservoir.
Collapse
|
13
|
Krishna BA, Miller WE, O'Connor CM. US28: HCMV's Swiss Army Knife. Viruses 2018; 10:E445. [PMID: 30127279 PMCID: PMC6116241 DOI: 10.3390/v10080445] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 08/08/2018] [Accepted: 08/17/2018] [Indexed: 12/13/2022] Open
Abstract
US28 is one of four G protein coupled receptors (GPCRs) encoded by human cytomegalovirus (HCMV). The US28 protein (pUS28) is a potent signaling molecule that alters a variety of cellular pathways that ultimately alter the host cell environment. This viral GPCR is expressed not only in the context of lytic replication but also during viral latency, highlighting its multifunctional properties. pUS28 is a functional GPCR, and its manipulation of multiple signaling pathways likely impacts HCMV pathogenesis. Herein, we will discuss the impact of pUS28 on both lytic and latent infection, pUS28-mediated signaling and its downstream consequences, and the influence this viral GPCR may have on disease states, including cardiovascular disease and cancer. We will also discuss the potential for and progress towards exploiting pUS28 as a novel therapeutic to combat HCMV.
Collapse
Affiliation(s)
- Benjamin A Krishna
- Genomic Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| | - William E Miller
- Department of Molecular Genetics, Biochemistry, & Microbiology, University of Cincinnati, Cincinnati, OH 45267, USA.
| | - Christine M O'Connor
- Genomic Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
14
|
A Abdullah A, Abdullah R, A Nazariah Z, N Balakrishnan K, Firdaus J Abdullah F, A Bala J, Mohd-Lila MA. Cyclophilin A as a target in the treatment of cytomegalovirus infections. Antivir Chem Chemother 2018; 26:2040206618811413. [PMID: 30449131 PMCID: PMC6243413 DOI: 10.1177/2040206618811413] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 10/12/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Viruses are obligate parasites that depend on the cellular machinery of the host to regenerate and manufacture their proteins. Most antiviral drugs on the market today target viral proteins. However, the more recent strategies involve targeting the host cell proteins or pathways that mediate viral replication. This new approach would be effective for most viruses while minimizing drug resistance and toxicity. METHODS Cytomegalovirus replication, latency, and immune response are mediated by the intermediate early protein 2, the main protein that determines the effectiveness of drugs in cytomegalovirus inhibition. This review explains how intermediate early protein 2 can modify the action of cyclosporin A, an immunosuppressive, and antiviral drug. It also links all the pathways mediated by cyclosporin A, cytomegalovirus replication, and its encoded proteins. RESULTS Intermediate early protein 2 can influence the cellular cyclophilin A pathway, affecting cyclosporin A as a mediator of viral replication or anti-cytomegalovirus drug. CONCLUSION Cyclosporin A has a dual function in cytomegalovirus pathogenesis. It has the immunosuppressive effect that establishes virus replication through the inhibition of T-cell function. It also has an anti-cytomegalovirus effect mediated by intermediate early protein 2. Both of these functions involve cyclophilin A pathway.
Collapse
Affiliation(s)
- Ashwaq A Abdullah
- 1 Institute of Bioscience, University Putra Malaysia, Serdang, Selangor D.E, Malaysia
- 2 Department of Microbiology, Faculty of Applied Science, Taiz University, Taiz, Yemen
| | - Rasedee Abdullah
- 1 Institute of Bioscience, University Putra Malaysia, Serdang, Selangor D.E, Malaysia
- 3 Department of Veterinary Laboratory Diagnosis, Universiti Putra Malaysia, Serdang, Selangor D.E, Malaysia
| | - Zeenathul A Nazariah
- 1 Institute of Bioscience, University Putra Malaysia, Serdang, Selangor D.E, Malaysia
- 4 Department of Pathology and Microbiology, Universiti Putra Malaysia, Serdang, Selangor D.E, Malaysia
| | - Krishnan N Balakrishnan
- 4 Department of Pathology and Microbiology, Universiti Putra Malaysia, Serdang, Selangor D.E, Malaysia
| | - Faez Firdaus J Abdullah
- 5 Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Serdang, Selangor D.E, Malaysia
| | - Jamilu A Bala
- 4 Department of Pathology and Microbiology, Universiti Putra Malaysia, Serdang, Selangor D.E, Malaysia
- 6 Department of Medical Laboratory Science, Faculty of Allied Health Sciences, Bayero University Kano, Kano, Nigeria
| | - Mohd-Azmi Mohd-Lila
- 1 Institute of Bioscience, University Putra Malaysia, Serdang, Selangor D.E, Malaysia
- 4 Department of Pathology and Microbiology, Universiti Putra Malaysia, Serdang, Selangor D.E, Malaysia
| |
Collapse
|
15
|
Latency-Associated Expression of Human Cytomegalovirus US28 Attenuates Cell Signaling Pathways To Maintain Latent Infection. mBio 2017; 8:mBio.01754-17. [PMID: 29208743 PMCID: PMC5717388 DOI: 10.1128/mbio.01754-17] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Reactivation of human cytomegalovirus (HCMV) latent infection from early myeloid lineage cells constitutes a threat to immunocompromised or immune-suppressed individuals. Consequently, understanding the control of latency and reactivation to allow targeting and killing of latently infected cells could have far-reaching clinical benefits. US28 is one of the few viral genes that is expressed during latency and encodes a cell surface G protein-coupled receptor (GPCR), which, during lytic infection, is a constitutive cell-signaling activator. Here we now show that in monocytes, which are recognized sites of HCMV latency in vivo, US28 attenuates multiple cell signaling pathways, including mitogen-activated protein (MAP) kinase and NF-κB, and that this is required to establish a latent infection; viruses deleted for US28 initiate a lytic infection in infected monocytes. We also show that these monocytes then become potent targets for the HCMV-specific host immune response and that latently infected cells treated with an inverse agonist of US28 also reactivate lytic infection and similarly become immune targets. Consequently, we suggest that the use of inhibitors of US28 could be a novel immunotherapeutic strategy to reactivate the latent viral reservoir, allowing it to be targeted by preexisting HCMV-specific T cells. Human cytomegalovirus (HCMV) is a betaherpesvirus and a leading cause of morbidity and mortality among immunosuppressed individuals. HCMV can establish latent infection, where the viral genome is maintained in an infected cell, without production of infectious virus. A number of genes, including US28, are expressed by HCMV during latent infection. US28 has been shown to activate many cellular signaling pathways during lytic infection, promoting lytic gene expression and virus production. As such, the role of US28 remains unclear and seems at odds with latency. Here, we show that US28 has the opposite phenotype in cells that support latent infection—it attenuates cellular signaling, thereby maintaining latency. Inhibition of US28 with a small-molecule inhibitor causes HCMV latent infection to reactivate, allowing latently infected cells to be detected and killed by the immune system. This approach could be used to treat latent HCMV to clear it from human transplants.
Collapse
|
16
|
Krishna BA, Spiess K, Poole EL, Lau B, Voigt S, Kledal TN, Rosenkilde MM, Sinclair JH. Targeting the latent cytomegalovirus reservoir with an antiviral fusion toxin protein. Nat Commun 2017; 8:14321. [PMID: 28148951 PMCID: PMC5296658 DOI: 10.1038/ncomms14321] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 12/16/2016] [Indexed: 12/26/2022] Open
Abstract
Reactivation of human cytomegalovirus (HCMV) in transplant recipients can cause life-threatening disease. Consequently, for transplant recipients, killing latently infected cells could have far-reaching clinical benefits. In vivo, myeloid cells and their progenitors are an important site of HCMV latency, and one viral gene expressed by latently infected myeloid cells is US28. This viral gene encodes a cell surface G protein-coupled receptor (GPCR) that binds chemokines, triggering its endocytosis. We show that the expression of US28 on the surface of latently infected cells allows monocytes and their progenitor CD34+ cells to be targeted and killed by F49A-FTP, a highly specific fusion toxin protein that binds this viral GPCR. As expected, this specific targeting of latently infected cells by F49A-FTP also robustly reduces virus reactivation in vitro. Consequently, such specific fusion toxin proteins could form the basis of a therapeutic strategy for eliminating latently infected cells before haematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- B A Krishna
- Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge CB20QQ, UK
| | - K Spiess
- Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark
| | - E L Poole
- Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge CB20QQ, UK
| | - B Lau
- Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge CB20QQ, UK
| | - S Voigt
- Department of Infectious Diseases, Robert Koch Institute, Nordufer 20, Berlin 13353, Germany.,Department of Pediatric Oncology/Hematology/SCT, Charité-Universitätsmedizin, Berlin 13353, Germany
| | - T N Kledal
- Section for Virology, The National Veterinary Institute, Technical University of Denmark, Frederiksberg DK-1870, Denmark
| | - M M Rosenkilde
- Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark
| | - J H Sinclair
- Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge CB20QQ, UK
| |
Collapse
|
17
|
Lee S, Chung YH, Lee C. US28, a Virally-Encoded GPCR as an Antiviral Target for Human Cytomegalovirus Infection. Biomol Ther (Seoul) 2017; 25:69-79. [PMID: 28035083 PMCID: PMC5207464 DOI: 10.4062/biomolther.2016.208] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/19/2016] [Accepted: 11/22/2016] [Indexed: 11/05/2022] Open
Abstract
Viruses continue to evolve a new strategy to take advantage of every aspect of host cells in order to maximize their survival. Due to their central roles in transducing a variety of transmembrane signals, GPCRs seem to be a prime target for viruses to pirate for their own use. Incorporation of GPCR functionality into the genome of herpesviruses has been demonstrated to be essential for pathogenesis of many herpesviruses-induced diseases. Here, we introduce US28 of human cytomegalovirus (HCMV) as the best-studied example of virally-encoded GPCRs to manipulate host GPCR signaling. In this review, we wish to summarize a number of US28-related topics including its regulation of host signaling pathways, its constitutive internalization, its structural and functional analysis, its roles in HCMV biology and pathogenesis, its proliferative activities and role in oncogenesis, and pharmacological modulation of its biological activities. This review will aid in our understanding of how pathogenic viruses usurp the host GPCR signaling for successful viral infection. This kind of knowledge will enable us to build a better strategy to control viral infection by normalizing the virally-dysregulated host GPCR signaling.
Collapse
Affiliation(s)
- Sungjin Lee
- College of Pharmacy, Dongguk University, Goyang 10326, Republic of Korea
| | - Yoon Hee Chung
- Department of Anatomy, College of Medicine, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Choongho Lee
- College of Pharmacy, Dongguk University, Goyang 10326, Republic of Korea
| |
Collapse
|
18
|
Abstract
Herpesviruses have evolved exquisite virus-host interactions that co-opt or evade a number of host pathways to enable the viruses to persist. Persistence of human cytomegalovirus (CMV), the prototypical betaherpesvirus, is particularly complex in the host organism. Depending on host physiology and the cell types infected, CMV persistence comprises latent, chronic, and productive states that may occur concurrently. Viral latency is a central strategy by which herpesviruses ensure their lifelong persistence. Although much remains to be defined about the virus-host interactions important to CMV latency, it is clear that checkpoints composed of viral and cellular factors exist to either maintain a latent state or initiate productive replication in response to host cues. CMV offers a rich platform for defining the virus-host interactions and understanding the host biology important to viral latency. This review describes current understanding of the virus-host interactions that contribute to viral latency and reactivation.
Collapse
Affiliation(s)
- Felicia Goodrum
- Department of Immunobiology, BIO5 Institute, University of Arizona, Tucson, Arizona 85721;
| |
Collapse
|
19
|
Lückmann M, Amarandi RM, Papargyri N, Jakobsen MH, Christiansen E, Jensen LJ, Pui A, Schwartz TW, Rosenkilde MM, Frimurer TM. Structure-based discovery of novel US28 small molecule ligands with different modes of action. Chem Biol Drug Des 2016; 89:289-296. [PMID: 27569905 DOI: 10.1111/cbdd.12848] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 08/02/2016] [Accepted: 08/25/2016] [Indexed: 12/11/2022]
Abstract
The human cytomegalovirus-encoded G protein-coupled receptor US28 is a constitutively active receptor, which can recognize various chemokines. Despite the recent determination of its 2.9 Å crystal structure, potent and US28-specific tool compounds are still scarce. Here, we used structural information from a refined US28:VUF2274 complex for virtual screening of >12 million commercially available small molecule compounds. Using a combined receptor- and ligand-based approach, we tested 98 of the top 0.1% ranked compounds, revealing novel chemotypes as compared to the ~1.45 million known ligands in the ChEMBL database. Two compounds were confirmed as agonist and inverse agonist, respectively, in both IP accumulation and Ca2+ mobilization assays. The screening setup presented in this work is computationally inexpensive and therefore particularly useful in an academic setting as it enables simultaneous testing in binding as well as in different functional assays and/or species without actual chemical synthesis.
Collapse
Affiliation(s)
- Michael Lückmann
- Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Roxana-Maria Amarandi
- Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark.,Faculty of Chemistry, Alexandru Ioan Cuza University of Iaşi, Iaşi, Romania
| | - Natalia Papargyri
- Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark.,Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark
| | - Mette H Jakobsen
- Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Elisabeth Christiansen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Odense M, Denmark
| | - Lars J Jensen
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Aurel Pui
- Faculty of Chemistry, Alexandru Ioan Cuza University of Iaşi, Iaşi, Romania
| | - Thue W Schwartz
- Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Mette M Rosenkilde
- Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Thomas M Frimurer
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
20
|
Wu SE, Miller WE. The HCMV US28 vGPCR induces potent Gαq/PLC-β signaling in monocytes leading to increased adhesion to endothelial cells. Virology 2016; 497:233-243. [PMID: 27497185 DOI: 10.1016/j.virol.2016.07.025] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 07/18/2016] [Accepted: 07/26/2016] [Indexed: 01/12/2023]
Abstract
US28 transcripts have been detected in primary monocytes and in THP-1 monocytes infected with HCMV but US28 protein expression has not yet been demonstrated in these cell types. Moreover, the mechanism(s) by which US28 signals and contributes to viral pathogenesis in monocytes remains unclear. Here, we show that US28 protein is robustly expressed in HCMV infected THP-1 monocytes and that US28 can trigger Gαq dependent signaling in THP-1 cells infected with HCMV and in THP-1 cells stably expressing US28. US28 signaling in these cells is dependent on G-protein coupling, but independent of chemokine binding. Importantly, we demonstrate that this US28 signaling is functionally important as it stimulates the adhesion of monocytes to an endothelial monolayer. Our studies, which demonstrate that US28-driven Gαq signaling has profound effects on monocyte biology, suggest that US28 driven phenotypic changes in HCMV infected monocytes may play important roles in HCMV dissemination and/or pathogenesis.
Collapse
Affiliation(s)
- Shu-En 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.
| |
Collapse
|
21
|
Luganini A, Terlizzi ME, Gribaudo G. Bioactive Molecules Released From Cells Infected with the Human Cytomegalovirus. Front Microbiol 2016; 7:715. [PMID: 27242736 PMCID: PMC4865657 DOI: 10.3389/fmicb.2016.00715] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 04/29/2016] [Indexed: 01/01/2023] Open
Abstract
Following primary infection in humans, the human cytomegalovirus (HCMV) persists in a latent state throughout the host’s lifetime despite a strong and efficient immune response. If the host experiences some form of immune dysregulation, such as immunosuppression or immunodeficiency, HCMV reactivates, thereby emerging from latency. Thus, in the absence of effective functional immune responses, as occurs in immunocompromised or immunoimmature individuals, both HCMV primary infections and reactivations from latency can cause significant morbidity and mortality. However, even in immunocompetent hosts, HCMV represents a relevant risk factor for the development of several chronic inflammatory diseases and certain forms of neoplasia. HCMV infection may shift between the lytic and latent state, regulated by a delicate and intricate balance between virus-mediated immunomodulation and host immune defenses. Indeed, HCMV is a master in manipulating innate and adaptive host defense pathways, and a large portion of its genome is devoted to encoding immunomodulatory proteins; such proteins may thus represent important virulence determinants. However, the pathogenesis of HCMV-related diseases is strengthened by the activities of bioactive molecules, of both viral and cellular origin, that are secreted from infected cells and collectively named as the secretome. Here, we review the state of knowledge on the composition and functions of HCMV-derived secretomes. In lytic infections of fibroblasts and different types of endothelial cells, the majority of HCMV-induced secreted proteins act in a paracrine fashion to stimulate the generation of an inflammatory microenvironment around infected cells; this may lead to vascular inflammation and angiogenesis that, in turn, foster HCMV replication and its dissemination through host tissues. Conversely, the HCMV secretome derived from latently infected hematopoietic progenitor cells induces an immunosuppressive extracellular environment that interferes with immune recognition and elimination of latently infected cells, thereby promoting viral persistence. Characterization of the composition and biological activities of HCMV secretomes from different types of infected cells will lay the foundation for future advances in our knowledge about the pathogenesis HCMV diseases and may provide targets for the development of novel antiviral intervention strategies.
Collapse
Affiliation(s)
- Anna Luganini
- Laboratory of Microbiology and Virology, Department of Life Sciences and Systems Biology, University of Turin Turin, Italy
| | - Maria E Terlizzi
- Laboratory of Microbiology and Virology, Department of Life Sciences and Systems Biology, University of Turin Turin, Italy
| | - Giorgio Gribaudo
- Laboratory of Microbiology and Virology, Department of Life Sciences and Systems Biology, University of Turin Turin, Italy
| |
Collapse
|
22
|
Bagdonaite I, Nordén R, Joshi HJ, King SL, Vakhrushev SY, Olofsson S, Wandall HH. Global Mapping of O-Glycosylation of Varicella Zoster Virus, Human Cytomegalovirus, and Epstein-Barr Virus. J Biol Chem 2016; 291:12014-28. [PMID: 27129252 DOI: 10.1074/jbc.m116.721746] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Indexed: 12/27/2022] Open
Abstract
Herpesviruses are among the most complex and widespread viruses, infection and propagation of which depend on envelope proteins. These proteins serve as mediators of cell entry as well as modulators of the immune response and are attractive vaccine targets. Although envelope proteins are known to carry glycans, little is known about the distribution, nature, and functions of these modifications. This is particularly true for O-glycans; thus we have recently developed a "bottom up" mass spectrometry-based technique for mapping O-glycosylation sites on herpes simplex virus type 1. We found wide distribution of O-glycans on herpes simplex virus type 1 glycoproteins and demonstrated that elongated O-glycans were essential for the propagation of the virus. Here, we applied our proteome-wide discovery platform for mapping O-glycosites on representative and clinically significant members of the herpesvirus family: varicella zoster virus, human cytomegalovirus, and Epstein-Barr virus. We identified a large number of O-glycosites distributed on most envelope proteins in all viruses and further demonstrated conserved patterns of O-glycans on distinct homologous proteins. Because glycosylation is highly dependent on the host cell, we tested varicella zoster virus-infected cell lysates and clinically isolated virus and found evidence of consistent O-glycosites. These results present a comprehensive view of herpesvirus O-glycosylation and point to the widespread occurrence of O-glycans in regions of envelope proteins important for virus entry, formation, and recognition by the host immune system. This knowledge enables dissection of specific functional roles of individual glycosites and, moreover, provides a framework for design of glycoprotein vaccines with representative glycosylation.
Collapse
Affiliation(s)
- Ieva Bagdonaite
- From the Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, 2200 Copenhagen, Denmark and
| | - Rickard Nordén
- the Department of Infectious Diseases, Institute of Biomedicine, University of Gothenburg, 413 45 Gothenburg, Sweden
| | - Hiren J Joshi
- From the Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, 2200 Copenhagen, Denmark and
| | - Sarah L King
- From the Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, 2200 Copenhagen, Denmark and
| | - Sergey Y Vakhrushev
- From the Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, 2200 Copenhagen, Denmark and
| | - Sigvard Olofsson
- the Department of Infectious Diseases, Institute of Biomedicine, University of Gothenburg, 413 45 Gothenburg, Sweden
| | - Hans H Wandall
- From the Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, 2200 Copenhagen, Denmark and
| |
Collapse
|
23
|
Christiaansen A, Varga SM, Spencer JV. Viral manipulation of the host immune response. Curr Opin Immunol 2015; 36:54-60. [PMID: 26177523 DOI: 10.1016/j.coi.2015.06.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 06/20/2015] [Accepted: 06/23/2015] [Indexed: 12/01/2022]
Abstract
Viruses are obligate intracellular parasites that require a host for essential machinery to replicate and ultimately be transmitted to new susceptible hosts. At the same time, the immune system has evolved to protect the human body from invasion by viruses and other pathogens. To counter this, viruses have developed an arsenal of strategies to not only avoid immune detection but to actively manipulate host immune responses to create an environment more favorable for infection. Here, we describe recent advances uncovering novel mechanisms by which viruses skew host immune responses through modulation of cytokine and chemokine signaling networks, interference with antigen presentation and T cell responses, and preventing antibody production.
Collapse
Affiliation(s)
- Allison Christiaansen
- Department of Microbiology, The University of Iowa, 51 Newton Road, 3-532 Bowen Science Building, Iowa City, IA 52242, USA
| | - Steven M Varga
- Department of Microbiology, The University of Iowa, 51 Newton Road, 3-532 Bowen Science Building, Iowa City, IA 52242, USA; Department of Pathology, The University of Iowa, 51 Newton Road, 3-532 Bowen Science Building, Iowa City, IA 52242, USA; Interdisciplinary Graduate Program in Immunology, The University of Iowa, 51 Newton Road, 3-532 Bowen Science Building, Iowa City, IA 52242, USA
| | - Juliet V Spencer
- Department of Biology, University of San Francisco, Harney Science Center, 2130 Fulton Street, San Francisco, CA 94117, USA.
| |
Collapse
|
24
|
Mølleskov-Jensen AS, Oliveira MT, Farrell HE, Davis-Poynter N. Virus-Encoded 7 Transmembrane Receptors. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 129:353-93. [DOI: 10.1016/bs.pmbts.2014.10.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
25
|
Allosteric modulation of the G protein-coupled US28 receptor of human cytomegalovirus: are the small-weight inverse agonist of US28 'camouflaged' agonists? Bioorg Med Chem Lett 2014; 24:3744-7. [PMID: 25052428 DOI: 10.1016/j.bmcl.2014.06.082] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 06/26/2014] [Accepted: 06/28/2014] [Indexed: 02/07/2023]
Abstract
The highly constitutively active G protein-coupled receptor US28 of human cytomegalovirus (HCMV) is thought to camouflage agonism by mediating constitutive endocytosis. With the use of the US28Δ300 mutant, which is largely devoid of constitutive internalization, I have demonstrated that the coupling of the receptor to its downstream signaling partners is responsible for the inverse agonism to agonism efficacy switch in some small-weight ligands of US28.
Collapse
|
26
|
Kralj A, Kurt E, Tschammer N, Heinrich MR. Synthesis and Biological Evaluation of Biphenyl Amides That Modulate the US28 Receptor. ChemMedChem 2013; 9:151-68. [DOI: 10.1002/cmdc.201300369] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 10/23/2013] [Indexed: 11/10/2022]
|
27
|
Kralj A, Nguyen MT, Tschammer N, Ocampo N, Gesiotto Q, Heinrich MR, Phanstiel O. Development of flavonoid-based inverse agonists of the key signaling receptor US28 of human cytomegalovirus. J Med Chem 2013; 56:5019-32. [PMID: 23768434 DOI: 10.1021/jm4003457] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A series of 31 chalcone- and flavonoid-based derivatives were synthesized in good overall yields and screened for their inverse agonist activity on the US28 receptor of human cytomegalovirus (HCMV). With one exception (e.g., 2-(5-bromo-2-methoxyphenyl)-3-hydroxy-4H-chromen-4-one), halogen-substituted flavonoids were typically more potent inverse agonists than their related hydro derivatives. While toxicity could be used to partially explain the inverse agonist activity of some members of the series, 5-(benzyloxy)-2-(5-bromo-2-methoxyphenyl)-4H-chromen-4-one (11b) acted on the US28 receptor as a nontoxic, inverse agonist. The full inverse agonism (efficacy, -89%) and potency (EC50 = 3.5 μM) observed with flavonoid 11b is especially important as it provides both a new tool to study US28 signaling and a potential platform for the future development of HCMV-targeting drugs.
Collapse
Affiliation(s)
- Ana Kralj
- Department of Chemistry and Pharmacy, Friedrich Alexander University, Erlangen 91052, Germany
| | | | | | | | | | | | | |
Collapse
|
28
|
US28 is a potent activator of phospholipase C during HCMV infection of clinically relevant target cells. PLoS One 2012; 7:e50524. [PMID: 23209769 PMCID: PMC3510093 DOI: 10.1371/journal.pone.0050524] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Accepted: 10/23/2012] [Indexed: 01/14/2023] Open
Abstract
Members of the cytomegalovirus family each encode two or more genes with significant homology to G-protein coupled receptors (GPCRs). In rodent models of pathogenesis, these viral encoded GPCRs play functionally significant roles, as their deletion results in crippled viruses that cannot traffic properly and/or replicate in virally important target cells. Of the four HCMV encoded GPCRs, US28 has garnered the most attention due to the fact that it exhibits both agonist-independent and agonist-dependent signaling activity and has been demonstrated to promote cellular migration and proliferation. Thus, it appears that the CMV GPCRs play important roles in viral replication in vivo as well as promote the development of virus-associated pathology. In the current study we have utilized a series of HCMV/US28 recombinants to investigate the expression profile and signaling activities of US28 in a number of cell types relevant to HCMV infection including smooth muscle cells, endothelial cells and cells derived from glioblastoma multiforme (GBM) tumors. The results indicate that US28 is expressed and exhibits constitutive agonist-independent signaling activity through PLC-β in all cell types tested. Moreover, while CCL5/RANTES and CX3CL1/Fractalkine both promote US28-dependent Ca++ release in smooth muscle cells, this agonist-dependent effect appears to be cell-specific as we fail to detect US28 driven Ca++ release in the GBM cells. We have also investigated the effects of US28 on signaling via endogenous GPCRs including those in the LPA receptor family. Our data indicate that US28 can enhance signaling via endogenous LPA receptors. Taken together, our results indicate that US28 induces a variety of signaling events in all cell types tested suggesting that US28 signaling likely plays a significant role during HCMV infection and dissemination in vivo.
Collapse
|
29
|
McSharry BP, Avdic S, Slobedman B. Human cytomegalovirus encoded homologs of cytokines, chemokines and their receptors: roles in immunomodulation. Viruses 2012. [PMID: 23202490 PMCID: PMC3509658 DOI: 10.3390/v4112448] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Human cytomegalovirus (HCMV), the largest human herpesvirus, infects a majority of the world’s population. Like all herpesviruses, following primary productive infection, HCMV establishes a life-long latent infection, from which it can reactivate years later to produce new, infectious virus. Despite the presence of a massive and sustained anti-HCMV immune response, productively infected individuals can shed virus for extended periods of time, and once latent infection is established, it is never cleared from the host. It has been proposed that HCMV must therefore encode functions which help to evade immune mediated clearance during productive virus replication and latency. Molecular mimicry is a strategy used by many viruses to subvert and regulate anti-viral immunity and HCMV has hijacked/developed a range of functions that imitate host encoded immunomodulatory proteins. This review will focus on the HCMV encoded homologs of cellular cytokines/chemokines and their receptors, with an emphasis on how these virus encoded homologs may facilitate viral evasion of immune clearance.
Collapse
Affiliation(s)
- Brian P. McSharry
- Discipline of Infectious Diseases and Immunology, University of Sydney, Australia; (B.P.McS); (S.A.); (B.S.)
- Centre for Virus Research, Westmead Millennium Institute, Sydney, Australia
| | - Selmir Avdic
- Discipline of Infectious Diseases and Immunology, University of Sydney, Australia; (B.P.McS); (S.A.); (B.S.)
- Centre for Virus Research, Westmead Millennium Institute, Sydney, Australia
| | - Barry Slobedman
- Discipline of Infectious Diseases and Immunology, University of Sydney, Australia; (B.P.McS); (S.A.); (B.S.)
- Author to whom correspondence should be addressed; ; Tel.: +1-61-93514334
| |
Collapse
|
30
|
Human cytomegalovirus-encoded UL33 and UL78 heteromerize with host CCR5 and CXCR4 impairing their HIV coreceptor activity. Blood 2012; 119:4908-18. [PMID: 22496149 DOI: 10.1182/blood-2011-08-372516] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Human cytomegalovirus (HCMV) encodes four 7-transmembrane-spanning (7TM) proteins, US28, US27, UL33, and UL78, which present important sequence homology with human chemokine receptors. Whereas US28 binds a large range of chemokines and disturbs host cell signaling at different levels, the others are orphans with largely unknown functions. Assembly of 2 different 7TM proteins into hetero-oligomeric complexes may profoundly change their respective functional properties. We show that HCMV-encoded UL33 and UL78 form heteromers with CCR5 and CXCR4 chemokine receptors in transfected human embryonic kidney 293T cells and monocytic THP-1 cells. Expression of UL33 and UL78 had pleiotropic, predominantly negative, effects on CCR5 and CXCR4 cell surface expression, ligand-induced internalization, signal transduction, and migration without modifying the chemokine binding properties of CCR5 and CXCR4. Importantly, the coreceptor activity of CCR5 and CXCR4 for HIV was largely impaired in the presence of UL33 and UL78 without affecting expression of the primary HIV entry receptor CD4 and its interaction with CCR5 and CXCR4. Collectively, we identified the first molecular function for the HCMV-encoded orphan UL33 and UL78 7TM proteins, namely the regulation of cellular chemokine receptors through receptor heteromerization.
Collapse
|
31
|
Tschammer N. Virally Encoded G Protein-Coupled Receptors. ANNUAL REPORTS IN MEDICINAL CHEMISTRY VOLUME 47 2012. [DOI: 10.1016/b978-0-12-396492-2.00025-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
32
|
Abstract
Cytomegalovirus (CMV) infection is the most common opportunistic infection in immunosuppressed individuals, such as transplant recipients or people living with HIV/AIDS, and congenital CMV is the leading viral cause of developmental disabilities in infants. Due to the highly species-specific nature of CMV, animal models that closely recapitulate human CMV (HCMV) are of growing importance for vaccine development. Here we present the genomic sequence of a novel nonhuman primate CMV from cynomolgus macaques (Macaca fascicularis; CyCMV). CyCMV (Ottawa strain) was isolated from the urine of a healthy, captive-bred, 4-year-old cynomolgus macaque of Philippine origin, and the viral genome was sequenced using next-generation Illumina sequencing to an average of 516-fold coverage. The CyCMV genome is 218,041 bp in length, with 49.5% G+C content and 84% protein-coding density. We have identified 262 putative open reading frames (ORFs) with an average coding length of 789 bp. The genomic organization of CyCMV is largely colinear with that of rhesus macaque CMV (RhCMV). Of the 262 CyCMV ORFs, 137 are homologous to HCMV genes, 243 are homologous to RhCMV 68.1, and 200 are homologous to RhCMV 180.92. CyCMV encodes four ORFs that are not present in RhCMV strain 68.1 or 180.92 but have homologies with HCMV (UL30, UL74A, UL126, and UL146). Similar to HCMV, CyCMV does not produce the RhCMV-specific viral homologue of cyclooxygenase-2. This newly characterized CMV may provide a novel model in which to study CMV biology and HCMV vaccine development.
Collapse
|
33
|
Schleiss MR. Congenital cytomegalovirus infection: molecular mechanisms mediating viral pathogenesis. Infect Disord Drug Targets 2011; 11:449-465. [PMID: 21827434 PMCID: PMC3869401 DOI: 10.2174/187152611797636721] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2010] [Accepted: 03/21/2011] [Indexed: 05/31/2023]
Abstract
Human cytomegalovirus (CMV) is responsible for approximately 40,000 congenital infections in the United States each year. Congenital CMV disease frequently produces serious neurodevelopmental disability, as well as vision impairment and sensorineural hearing loss. Development of a CMV vaccine is therefore considered to be a major public health priority. The mechanisms by which CMV injures the fetus are complex and likely include a combination of direct fetal injury induced by pathologic virally-encoded gene products, an inability of the maternal immune response to control infection, and the direct impact of infection on placental function. CMV encodes gene products that function, both at the RNA and the protein level, to interfere with many cellular processes. These include gene products that modify the cell cycle; interfere with apoptosis; induce an inflammatory response; mediate vascular injury; induce site-specific breakage of chromosomes; promote oncogenesis; dysregulate cellular proliferation; and facilitate evasion of host immune responses. This minireview summarizes current concepts regarding these aspects of the molecular virology of CMV and the potential pathogenic impact of viral gene expression on the developing fetus. Areas for potential development of novel therapeutic intervention are suggested for improving the outcome of this disabling congenital infection.
Collapse
Affiliation(s)
- Mark R Schleiss
- Center for Infectious Diseases and Microbiology Translational Research, University of Minnesota Medical School, Department of Pediatrics, Division of Pediatric Infectious Diseases and Immunology, Minneapolis, MN 55455, USA.
| |
Collapse
|
34
|
Soroceanu L, Matlaf L, Bezrookove V, Harkins L, Martinez R, Greene M, Soteropoulos P, Cobbs CS. Human cytomegalovirus US28 found in glioblastoma promotes an invasive and angiogenic phenotype. Cancer Res 2011; 71:6643-53. [PMID: 21900396 DOI: 10.1158/0008-5472.can-11-0744] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Human cytomegalovirus (HCMV) infections are seen often in glioblastoma multiforme (GBM) tumors, but whether the virus contributes to GBM pathogenesis is unclear. In this study, we explored an oncogenic role for the G-protein-coupled receptor-like protein US28 encoded by HCMV that we found to be expressed widely in human GBMs. Immunohistochemical and reverse transcriptase PCR approaches established that US28 was expressed in approximately 60% of human GBM tissues and primary cultures examined. In either uninfected GBM cells or neural progenitor cells, thought to be the GBM precursor cells, HCMV infection or US28 overexpression was sufficient to promote secretion of biologically active VEGF and to activate multiple cellular kinases that promote glioma growth and invasion, including phosphorylated STAT3 (p-STAT3) and endothelial nitric oxide synthase (e-NOS). Consistent with these findings, US28 overexpression increased primary GBM cell invasion in Matrigel. Notably, this invasive phenotype was further enhanced by exposure to CCL5/RANTES, a US28 ligand, associated with poor patient outcome in GBM. Conversely, RNA interference-mediated knockdown of US28 in human glioma cells persistently infected with HCMV led to an inhibition in VEGF expression and glioma cell invasion in response to CCL5 stimulation. Analysis of clinical GBM specimens further revealed that US28 colocalized in situ with several markers of angiogenesis and inflammation, including VEGF, p-STAT3, COX2, and e-NOS. Taken together, our results indicate that US28 expression from HCMV contributes to GBM pathogenesis by inducing an invasive, angiogenic phenotype. In addition, these findings argue that US28-CCL5 paracrine signaling may contribute to glioma progression and suggest that targeting US28 may provide therapeutic benefits in GBM treatment.
Collapse
Affiliation(s)
- Liliana Soroceanu
- California Pacific Medical Center Research Institute, University of California, San Francisco, California, USA.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Kralj A, Wetzel A, Mahmoudian S, Stamminger T, Tschammer N, Heinrich MR. Identification of novel allosteric modulators for the G-protein coupled US28 receptor of human cytomegalovirus. Bioorg Med Chem Lett 2011; 21:5446-50. [DOI: 10.1016/j.bmcl.2011.06.120] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Revised: 06/27/2011] [Accepted: 06/28/2011] [Indexed: 10/18/2022]
|
36
|
Human cytomegalovirus pUS27 G protein-coupled receptor homologue is required for efficient spread by the extracellular route but not for direct cell-to-cell spread. J Virol 2011; 85:3700-7. [PMID: 21307184 DOI: 10.1128/jvi.02442-10] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Human cytomegalovirus (HCMV) encodes multiple G protein-coupled receptor (GPCR) homologues, including pUS27, pUS28, pUL33, and pUL78. To explore the function of pUS27, we constructed pUS27-deficient derivates of two clinical isolates of HCMV. BFX-GFPstopUS27 is a FIX variant with a single base pair change in the US27 open reading frame, generating a stop codon that ablates accumulation of the GPCR homologue, and TB40/E-mCherrydlUS27 lacks the entire US27 coding region. BFX-GFPstopUS27 generated 10-fold less extracellular progeny in fibroblasts, and TB40/E-mCherrydlUS27 exhibited a similar defect in endothelial cells. The pUS27-deficient FIX derivative produced normal quantities of viral DNA and viral proteins tested, and a late virion protein was appropriately localized to the cytoplasmic assembly zone. After infection at a low multiplicity with wild-type FIX virus, neutralizing antibody reduced the accumulation of intracellular viral DNA and intracellular virions, as would be expected if the virus is limited to direct cell-to-cell spread by neutralization of extracellular virus. In contrast, the antibody had little effect on the spread of the BFX-GFPstopUS27 virus. Further, after infection at a low multiplicity, the pUS27-deficient TB40/E virus exhibited a growth defect in endothelial cells, where the clinical isolate normally generates extracellular virus, but the TB40/E derivative exhibited little defect in epithelial cells, where the wild-type virus does not produce extracellular virus. Thus, mutants lacking pUS27 rely primarily on direct cell-to-cell spread, and we conclude that the viral GCPR homologue acts at a late stage of the HCMV replication cycle to support spread of virus by the extracellular route.
Collapse
|
37
|
The battle between virus and host: modulation of Toll-like receptor signaling pathways by virus infection. Mediators Inflamm 2010; 2010:184328. [PMID: 20672047 PMCID: PMC2903949 DOI: 10.1155/2010/184328] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2009] [Accepted: 04/07/2010] [Indexed: 02/05/2023] Open
Abstract
In order to establish an infection, viruses need to either suppress or escape from host immune defense systems. Recent immunological research has focused on innate immunity as the first line of host defense, especially pattern recognition molecules such as Toll-like receptors (TLRs), RIG-I-like receptors (RLRs), and NOD-like receptors (NLRs). Various microbial components are recognized by their vague and common molecular shapes so-called, pathogen-associated molecular patterns (PAMPs). PAMPs induce inflammatory reactions mediated by the activation of the transcription factor, NF-κB, and by interferons, which lead to an antiviral immune response. Viruses have the capacity to suppress or escape from this pattern recognition molecule-mediated antimicrobial response in various ways. In this paper, we review the various strategies used by viruses to modulate the pattern recognition molecule-mediated innate immune response.
Collapse
|