1
|
Penner I, Krämer N, Hirsch J, Büscher N, Schmidt H, Plachter B. Deletion of the Human Cytomegalovirus US2 to US11 Gene Family Members Impairs the Type-I Interferon Response. Viruses 2025; 17:426. [PMID: 40143353 PMCID: PMC11945591 DOI: 10.3390/v17030426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/07/2025] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
Infection of cells with the human cytomegalovirus (HCMV) triggers the expression of interferon-stimulated genes (ISGs). ISGs encode proteins with antiviral functions, such as inhibiting viral replication, promoting cell death of infected cells and enhancing immune responses. HCMV has evolved mechanisms to evade the antiviral effects of ISGs. The viral proteins encoded by the viral genes US7, US8, and US9 have been shown to interfere with interferon induction. US7 to US9 are embedded in a cluster of HCMV genes, termed US2 to US11. The individual members of this gene family interfere on multiple levels with innate and adaptive immune responses to HCMV infection. Using viral mutants with different deletions in US2 to US11, we addressed the question if genes other than US7 to US9 would also influence the IFN responses. Surprisingly, deletion of the complete US2 to US11 gene region led to reduced levels of selected ISGs. Cells infected with viruses in which individual US2 to US11 genes were deleted showed a less pronounced reduction of the selected ISGs. The experiments including RNA-seq analyses indicate that genes of the US2 to US11 gene family have a complex interaction with the IFN-ISG response which is likely regulated on the level of ISG protein stability. As US2-US11 are dispensable for replication in cell culture, the genomic region was frequently used for the insertion of bacterial artificial chromosome vectors in the process of cloning the complete HCMV genome. The results shown here must be considered when viruses derived from BACs with US2-US11 deletions are used and whether appropriate controls must be applied.
Collapse
|
2
|
Gáspár Z, Szabó BG, Ceglédi A, Lakatos B. Human herpesvirus reactivation and its potential role in the pathogenesis of post-acute sequelae of SARS-CoV-2 infection. GeroScience 2025; 47:167-187. [PMID: 39207648 PMCID: PMC11872864 DOI: 10.1007/s11357-024-01323-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
The emergence of SARS-CoV-2 has precipitated a global pandemic with substantial long-term health implications, including the condition known as post-acute sequelae of SARS-CoV-2 infection (PASC), commonly referred to as Long COVID. PASC is marked by persistent symptoms such as fatigue, neurological issues, and autonomic dysfunction that persist for months beyond the acute phase of COVID-19. This review examines the potential role of herpesvirus reactivation, specifically Epstein-Barr virus (EBV) and cytomegalovirus (CMV), in the pathogenesis of PASC. Elevated antibody titers and specific T cell responses suggest recent herpesvirus reactivation in some PASC patients, although viremia is not consistently detected. SARS-CoV-2 exhibits endothelial trophism, directly affecting the vascular endothelium and contributing to microvascular pathologies. These pathologies are significant in PASC, where microvascular dysfunction may underlie various chronic symptoms. Similarly, herpesviruses like CMV also exhibit endothelial trophism, which may exacerbate endothelial damage when reactivated. Evidence suggests that EBV and CMV reactivation could indirectly contribute to the immune dysregulation, immunosenescence, and autoimmune responses observed in PASC. Additionally, EBV may play a role in the genesis of neurological symptoms through creating mitochondrial dysfunction, though direct confirmation remains elusive. The reviewed evidence suggests that while herpesviruses may not play a direct role in the pathogenesis of PASC, their potential indirect effects, especially in the context of endothelial involvement, warrant further investigation.
Collapse
Affiliation(s)
- Zsófia Gáspár
- School of PhD Studies, Semmelweis University, Üllői Street 26, 1085, Budapest, Hungary
- South Pest Central Hospital, National Institute of Haematology and Infectious Diseases, Albert Flórián Street 5-7, 1097, Budapest, Hungary
| | - Bálint Gergely Szabó
- School of PhD Studies, Semmelweis University, Üllői Street 26, 1085, Budapest, Hungary.
- South Pest Central Hospital, National Institute of Haematology and Infectious Diseases, Albert Flórián Street 5-7, 1097, Budapest, Hungary.
- Departmental Group of Infectious Diseases, Department of Internal Medicine and Haematology, Semmelweis University, Albert Flórián Street 5-7, 1097, Budapest, Hungary.
| | - Andrea Ceglédi
- South Pest Central Hospital, National Institute of Haematology and Infectious Diseases, Albert Flórián Street 5-7, 1097, Budapest, Hungary
| | - Botond Lakatos
- School of PhD Studies, Semmelweis University, Üllői Street 26, 1085, Budapest, Hungary
- South Pest Central Hospital, National Institute of Haematology and Infectious Diseases, Albert Flórián Street 5-7, 1097, Budapest, Hungary
- Departmental Group of Infectious Diseases, Department of Internal Medicine and Haematology, Semmelweis University, Albert Flórián Street 5-7, 1097, Budapest, Hungary
| |
Collapse
|
3
|
Diggins NL, Pham AH, Mitchell J, Parkins CJ, Slind L, Turner R, Lee BJ, Yurochko AD, Caposio P, Nelson JA, Hancock MH. Viral microRNA regulation of Akt is necessary for reactivation of Human Cytomegalovirus from latency in CD34+ hematopoietic progenitor cells and humanized mice. PLoS Pathog 2024; 20:e1012285. [PMID: 39661658 PMCID: PMC11666035 DOI: 10.1371/journal.ppat.1012285] [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: 05/23/2024] [Revised: 12/23/2024] [Accepted: 11/26/2024] [Indexed: 12/13/2024] Open
Abstract
Human cytomegalovirus (HCMV) actively manipulates cellular signaling pathways to benefit viral replication. Phosphatidyl-inositol 3-kinase (PI3K)/Akt signaling is an important negative regulator of HCMV replication, and during lytic infection the virus utilizes pUL38 to limit Akt phosphorylation and activity. During latency, PI3K/Akt signaling also limits virus replication, but how this is overcome at the time of reactivation is unknown. Virally encoded microRNAs (miRNAs) are a key component of the virus arsenal used to alter signaling during latency and reactivation. In the present study we show that three HCMV miRNAs (miR-UL36, miR-UL112 and miR-UL148D) downregulate Akt expression and attenuate downstream signaling, resulting in the activation of FOXO3a and enhanced internal promoter-driven IE transcription. A virus lacking expression of all three miRNAs is unable to reactivate from latency both in CD34+ hematopoietic progenitor cells and in a humanized mouse model of HCMV infection, however downregulating Akt restores the ability of the mutant virus to replicate. These findings highlight the negative role Akt signaling plays in HCMV replication in lytic and latent infection and how the virus has evolved miRNA-mediated countermeasures to promote successful reactivation.
Collapse
Affiliation(s)
- Nicole L. Diggins
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Andrew H. Pham
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Jennifer Mitchell
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Christopher J. Parkins
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Luke Slind
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Rebekah Turner
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Byeong-Jae Lee
- Department of Microbiology & Immunology, Center for Applied Immunology and Pathological Processes, Center for Emerging Viral Threats, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, United States of America
| | - Andrew D. Yurochko
- Department of Microbiology & Immunology, Center for Applied Immunology and Pathological Processes, Center for Emerging Viral Threats, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, United States of America
| | - Patrizia Caposio
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Jay A. Nelson
- 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
| |
Collapse
|
4
|
Mocarski ES. Cytomegalovirus Biology Viewed Through a Cell Death Suppression Lens. Viruses 2024; 16:1820. [PMID: 39772130 PMCID: PMC11680106 DOI: 10.3390/v16121820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/22/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
Cytomegaloviruses, species-specific members of the betaherpesviruses, encode an impressive array of immune evasion strategies committed to the manipulation of the host immune system enabling these viruses to remain for life in a stand-off with host innate and adaptive immune mechanisms. Even though they are species-restricted, cytomegaloviruses are distributed across a wide range of different mammalian species in which they cause systemic infection involving many different cell types. Regulated, or programmed cell death has a recognized potential to eliminate infected cells prior to completion of viral replication and release of progeny. Cell death also naturally terminates replication during the final stages of replication. Over the past two decades, the host defense potential of known programmed cell death pathways (apoptosis, necroptosis, and pyroptosis), as well as a novel mitochondrial serine protease pathway have been defined through studies of cytomegalovirus-encoded cell death suppressors. Such virus-encoded inhibitors prevent virus-induced, cytokine-induced, and stress-induced death of infected cells while also moderating inflammation. By evading cell death and consequent inflammation as well as innate and adaptive immune clearance, cytomegaloviruses represent successful pathogens that become a critical disease threat when the host immune system is compromised. This review will discuss cell death programs acquired for mammalian host defense against cytomegaloviruses and enumerate the range of modulatory strategies this type of virus employs to balance host defense in favor of lifelong persistence.
Collapse
Affiliation(s)
- Edward S. Mocarski
- Department of Microbiology & Immunology, Stanford Medical School, Stanford University, Stanford, CA 94305, USA;
- Department of Microbiology & Immunology, Emory Medical School, Emory Vaccine Center, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
5
|
Yoo WS, Kwon LH, Eom Y, Thng ZX, Or C, Nguyen QD, Kim SJ. Cytomegalovirus Corneal Endotheliitis: A Comprehensive Review. Ocul Immunol Inflamm 2024; 32:2228-2237. [PMID: 38417101 DOI: 10.1080/09273948.2024.2320704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/08/2024] [Accepted: 02/14/2024] [Indexed: 03/01/2024]
Abstract
Cytomegalovirus (CMV) anterior uveitis and corneal endotheliitis are the most common ocular diseases caused by CMV infections in immunocompetent patients. The incidence of CMV corneal endotheliitis is relatively high in middle-aged men. CMV corneal endotheliitis presents with mild anterior chamber inflammation, corneal edema, keratic precipitates, and elevated intraocular pressure. It resembles Posner-Schlossman syndrome and Fuchs uveitis because of the elevated intraocular pressure. Without proper diagnosis and treatment, it may progress to bullous keratopathy or glaucoma, necessitating keratoplasty or glaucoma surgery. Therefore, early diagnosis and treatment are important for a good prognosis. Aqueous humor analysis can facilitate the diagnosis of CMV corneal endotheliitis, and early antiviral treatment can decrease the risk of corneal compensation or glaucomatous optic atrophy. In this article, we review the epidemiology, pathogenesis, clinical manifestations, diagnosis, treatment, and prognosis of CMV corneal endotheliitis along with the evidence for early clinical diagnosis and active antiviral treatment.
Collapse
Affiliation(s)
- Woong-Sun Yoo
- Department of Ophthalmology, Gyeongsang National University College of Medicine, and Gyeongsang National University Hospital, Jinju, Republic of Korea
- Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, California, USA
| | - Lee-Ha Kwon
- Department of Ophthalmology, Gyeongsang National University College of Medicine, and Gyeongsang National University Hospital, Jinju, Republic of Korea
- Institute of Health Sciences, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Youngsub Eom
- Department of Ophthalmology, Korea University College of Medicine, Seoul, Republic of Korea
- Department of Ophthalmology, Korea University Ansan Hospital, Ansan, Republic of Korea
| | - Zheng Xian Thng
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, California, USA
- National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore, Singapore
| | - Christopher Or
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, California, USA
| | - Quan Dong Nguyen
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, California, USA
| | - Seong-Jae Kim
- Department of Ophthalmology, Gyeongsang National University College of Medicine, and Gyeongsang National University Hospital, Jinju, Republic of Korea
| |
Collapse
|
6
|
Sabbaghian M, Gheitasi H, Fadaee M, Javadi Henafard H, Tavakoli A, Shekarchi AA, Poortahmasebi V. Human cytomegalovirus microRNAs: strategies for immune evasion and viral latency. Arch Virol 2024; 169:157. [PMID: 38969819 DOI: 10.1007/s00705-024-06080-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 05/17/2024] [Indexed: 07/07/2024]
Abstract
Viruses use various strategies and mechanisms to deal with cells and proteins of the immune system that form a barrier against infection. One of these mechanisms is the encoding and production of viral microRNAs (miRNAs), whose function is to regulate the gene expression of the host cell and the virus, thus creating a suitable environment for survival and spreading viral infection. miRNAs are short, single-stranded, non-coding RNA molecules that can regulate the expression of host and viral proteins, and due to their non-immunogenic nature, they are not eliminated by the cells of the immune system. More than half of the viral miRNAs are encoded and produced by Orthoherpesviridae family members. Human cytomegalovirus (HCMV) produces miRNAs that mediate various processes in infected cells to contribute to HCMV pathogenicity, including immune escape, viral latency, and cell apoptosis. Here, we discuss which cellular and viral proteins or cellular pathways and processes these mysterious molecules target to evade immunity and support viral latency in infected cells. We also discuss current evidence that their function of bypassing the host's innate and adaptive immune system is essential for the survival and multiplication of the virus and the spread of HCMV infection.
Collapse
Affiliation(s)
- Mohammad Sabbaghian
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamidreza Gheitasi
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Manouchehr Fadaee
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran
| | | | - Ahmad Tavakoli
- Research Center of Pediatric Infectious Diseases, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Akbar Shekarchi
- Department of Pathology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahdat Poortahmasebi
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Pantalone MR, Almazan NM, Lattanzio R, Taher C, De Fabritiis S, Valentinuzzi S, Bishehsari F, Mahdavinia M, Verginelli F, Rahbar A, Mariani-Costantini R, Söderberg-Naucler C. Human cytomegalovirus infection enhances 5‑lipoxygenase and cycloxygenase‑2 expression in colorectal cancer. Int J Oncol 2023; 63:116. [PMID: 37654195 PMCID: PMC10546380 DOI: 10.3892/ijo.2023.5564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 07/07/2023] [Indexed: 09/02/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common and fatal types of cancer. Inflammation promotes CRC development, however, the underlying etiological factors are unknown. Human cytomegalovirus (HCMV), a virus that induces inflammation and other cancer hallmarks, has been detected in several types of malignancy, including CRC. The present study investigated whether HCMV infection was associated with expression of the pro‑inflammatory enzymes 5‑lipoxygenase (5‑LO) and cyclooxygenase‑2 (COX‑2) and other molecular, genetic and clinicopathological CRC features. The present study assessed 146 individual paraffin‑embedded CRC tissue microarray (TMA) cores already characterized for TP53 and KRAS mutations, microsatellite instability (MSI) status, Ki‑67 index and EGFR by immunohistochemistry (IHC). The cores were further analyzed by IHC for the expression of two HCMV proteins (Immediate Early, IE and pp65) and the inflammatory markers 5‑LO and COX‑2. The CRC cell lines Caco‑2 and LS‑174T were infected with HCMV strain VR1814, treated with antiviral drug ganciclovir (GCV) and/or anti‑inflammatory drug celecoxib (CCX) and analyzed by reverse transcription‑quantitative PCR and immunofluorescence for 5‑LO, COX‑2, IE and pp65 transcripts and proteins. HCMV IE and pp65 proteins were detected in ~90% of the CRC cases tested; this was correlated with COX‑2, 5‑LO and KI‑67 expression, but not with EGFR immunostaining, TP53 and KRAS mutations or MSI status. In vitro, HCMV infection upregulated 5‑LO and COX‑2 transcript and proteins in both Caco‑2 and LS‑174T cells and enhanced cell proliferation as determined by MTT assay. Treatment with GCV and CCX significantly decreased the transcript levels of COX‑2, 5‑LO, HCMV IE and pp65 in infected cells. HCMV was widely expressed in CRC and may promote inflammation and serve as a potential new target for CRC therapy.
Collapse
Affiliation(s)
- Mattia Russel Pantalone
- Department of Medicine, Solna, Microbial Pathogenesis Unit, Karolinska Institutet, 17164 Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, 17164 Stockholm, Sweden
- Center for Advanced Studies and Technology, G. d'Annunzio University, I-66100 Chieti, Italy
| | - Nerea Martin Almazan
- Department of Medicine, Solna, Microbial Pathogenesis Unit, Karolinska Institutet, 17164 Stockholm, Sweden
- Department of Laboratory Medicine, Unit of Microbial Pathogenesis, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Rossano Lattanzio
- Center for Advanced Studies and Technology, G. d'Annunzio University, I-66100 Chieti, Italy
- Department of Innovative Technologies in Medicine & Dentistry, G. d'Annunzio University, I-66100 Chieti, Italy
| | - Chato Taher
- Department of Basic Sciences, Hawler Medical University, Erbil 44001, Iraq
| | - Simone De Fabritiis
- Center for Advanced Studies and Technology, G. d'Annunzio University, I-66100 Chieti, Italy
- Department of Innovative Technologies in Medicine & Dentistry, G. d'Annunzio University, I-66100 Chieti, Italy
| | - Silvia Valentinuzzi
- Center for Advanced Studies and Technology, G. d'Annunzio University, I-66100 Chieti, Italy
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, I-66100 Chieti, Italy
| | - Faraz Bishehsari
- Division of Digestive Diseases, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612, USA
- Digestive Disease Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran 14114, Iran
| | - Mahboobeh Mahdavinia
- Digestive Disease Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran 14114, Iran
- Department of Internal Medicine, Division of Allergy and Immunology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Fabio Verginelli
- Center for Advanced Studies and Technology, G. d'Annunzio University, I-66100 Chieti, Italy
- Department of Pharmacy, G. d'Annunzio University of Chieti-Pescara, I-66100 Chieti, Italy
| | - Afsar Rahbar
- Department of Medicine, Solna, Microbial Pathogenesis Unit, Karolinska Institutet, 17164 Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, 17164 Stockholm, Sweden
| | | | - Cecilia Söderberg-Naucler
- Department of Medicine, Solna, Microbial Pathogenesis Unit, Karolinska Institutet, 17164 Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, 17164 Stockholm, Sweden
- MediCity Research Laboratory, University of Turku, FI-20014 Turku, Finland
- Institute of Biomedicine, University of Turku, FI-20014 Turku, Finland
| |
Collapse
|
8
|
Medica S, Crawford LB, Denton M, Min CK, Jones TA, Alexander T, Parkins CJ, Diggins NL, Streblow GJ, Mayo AT, Kreklywich CN, Smith P, Jeng S, McWeeney S, Hancock MH, Yurochko A, Cohen MS, Caposio P, Streblow DN. Proximity-dependent mapping of the HCMV US28 interactome identifies RhoGEF signaling as a requirement for efficient viral reactivation. PLoS Pathog 2023; 19:e1011682. [PMID: 37782657 PMCID: PMC10569644 DOI: 10.1371/journal.ppat.1011682] [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: 01/03/2023] [Revised: 10/12/2023] [Accepted: 09/12/2023] [Indexed: 10/04/2023] Open
Abstract
Human cytomegalovirus (HCMV) encodes multiple putative G protein-coupled receptors (GPCRs). US28 functions as a viral chemokine receptor and is expressed during both latent and lytic phases of virus infection. US28 actively promotes cellular migration, transformation, and plays a major role in mediating viral latency and reactivation; however, knowledge about the interaction partners involved in these processes is still incomplete. Herein, we utilized a proximity-dependent biotinylating enzyme (TurboID) to characterize the US28 interactome when expressed in isolation, and during both latent (CD34+ hematopoietic progenitor cells) and lytic (fibroblasts) HCMV infection. Our analyses indicate that the US28 signalosome converges with RhoA and EGFR signal transduction pathways, sharing multiple mediators that are major actors in processes such as cellular proliferation and differentiation. Integral members of the US28 signaling complex were validated in functional assays by immunoblot and small-molecule inhibitors. Importantly, we identified RhoGEFs as key US28 signaling intermediaries. In vitro latency and reactivation assays utilizing primary CD34+ hematopoietic progenitor cells (HPCs) treated with the small-molecule inhibitors Rhosin or Y16 indicated that US28 -RhoGEF interactions are required for efficient viral reactivation. These findings were recapitulated in vivo using a humanized mouse model where inhibition of RhoGEFs resulted in a failure of the virus to reactivate. Together, our data identifies multiple new proteins in the US28 interactome that play major roles in viral latency and reactivation, highlights the utility of proximity-sensor labeling to characterize protein interactomes, and provides insight into targets for the development of novel anti-HCMV therapeutics.
Collapse
Affiliation(s)
- Samuel Medica
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Lindsey B. Crawford
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Michael Denton
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Chan-Ki Min
- Department of Microbiology & Immunology, Center for Molecular & Tumor Virology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, United States of America
| | - Taylor A. Jones
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Timothy Alexander
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Christopher J. Parkins
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Nicole L. Diggins
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Gabriel J. Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Adam T. Mayo
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Craig N. Kreklywich
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Patricia Smith
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Sophia Jeng
- Department of Bioinformatics and Computational Biology, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Shannon McWeeney
- Department of Bioinformatics and Computational Biology, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Meaghan H. Hancock
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Andrew Yurochko
- Department of Microbiology & Immunology, Center for Molecular & Tumor Virology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, United States of America
| | - Michael S. Cohen
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Patrizia Caposio
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Daniel N. Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, United States of America
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| |
Collapse
|
9
|
Diggins NL, Hancock MH. Viral miRNA regulation of host gene expression. Semin Cell Dev Biol 2023; 146:2-19. [PMID: 36463091 PMCID: PMC10101914 DOI: 10.1016/j.semcdb.2022.11.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/16/2022] [Accepted: 11/22/2022] [Indexed: 12/05/2022]
Abstract
Viruses have evolved a multitude of mechanisms to combat barriers to productive infection in the host cell. Virally-encoded miRNAs are one such means to regulate host gene expression in ways that benefit the virus lifecycle. miRNAs are small non-coding RNAs that regulate protein expression but do not trigger the adaptive immune response, making them powerful tools encoded by viruses to regulate cellular processes. Diverse viruses encode for miRNAs but little sequence homology exists between miRNAs of different viral species. Despite this, common cellular pathways are targeted for regulation, including apoptosis, immune evasion, cell growth and differentiation. Herein we will highlight the viruses that encode miRNAs and provide mechanistic insight into how viral miRNAs aid in lytic and latent infection by targeting common cellular processes. We also highlight how viral miRNAs can mimic host cell miRNAs as well as how viral miRNAs have evolved to regulate host miRNA expression. These studies dispel the myth that viral miRNAs are subtle regulators of gene expression, and highlight the critical importance of viral miRNAs to the virus lifecycle.
Collapse
Affiliation(s)
- Nicole L Diggins
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Portland, OR, USA
| | - Meaghan H Hancock
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
10
|
Pandita S, Verma A, Kumar N. Role of miRNAs in regulating virus replication. ANIMAL GENE 2023; 30:200162. [DOI: 10.1016/j.angen.2023.200162] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
11
|
Crawford LB. Hematopoietic stem cells and betaherpesvirus latency. Front Cell Infect Microbiol 2023; 13:1189805. [PMID: 37346032 PMCID: PMC10279960 DOI: 10.3389/fcimb.2023.1189805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/11/2023] [Indexed: 06/23/2023] Open
Abstract
The human betaherpesviruses including human cytomegalovirus (HCMV), human herpesvirus (HHV)-6a and HHV-6b, and HHV-7 infect and establish latency in CD34+ hematopoietic stem and progenitor cells (HPCs). The diverse repertoire of HPCs in humans and the complex interactions between these viruses and host HPCs regulate the viral lifecycle, including latency. Precise manipulation of host and viral factors contribute to preferential maintenance of the viral genome, increased host cell survival, and specific manipulation of the cellular environment including suppression of neighboring cells and immune control. The dynamic control of these processes by the virus regulate inter- and intra-host signals critical to the establishment of chronic infection. Regulation occurs through direct viral protein interactions and cellular signaling, miRNA regulation, and viral mimics of cellular receptors and ligands, all leading to control of cell proliferation, survival, and differentiation. Hematopoietic stem cells have unique biological properties and the tandem control of virus and host make this a unique environment for chronic herpesvirus infection in the bone marrow. This review highlights the elegant complexities of the betaherpesvirus latency and HPC virus-host interactions.
Collapse
Affiliation(s)
- Lindsey B Crawford
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, United States
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, United States
- Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE, United States
| |
Collapse
|
12
|
Schinas G, Moustaka V, Polyzou E, Almyroudi MP, Dimopoulos G, Akinosoglou K. Targeting CMV Reactivation to Optimize Care for Critically Ill COVID-19 Patients: A Review on the Therapeutic Potential of Antiviral Treatment. Viruses 2023; 15:v15051165. [PMID: 37243251 DOI: 10.3390/v15051165] [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: 05/03/2023] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Cytomegalovirus (CMV) reactivation has been linked to adverse clinical outcomes in critically ill patients, with emerging evidence suggesting a potential connection with severe COVID-19. Mechanisms driving this association may include primary lung injury, amplification of systemic inflammation, and secondary immunosuppression. Diagnostic challenges in detecting and assessing CMV reactivation necessitate a comprehensive approach to improve accuracy and inform treatment decisions. Currently, there is limited evidence on the efficacy and safety of CMV pharmacotherapy in critically ill COVID-19 patients. Although insights from non-COVID-19 critical illness studies suggest a potential role for antiviral treatment or prophylaxis, the risks and benefits must be carefully balanced in this vulnerable patient population. Understanding the pathophysiological role of CMV in the context of COVID-19 and exploring the advantages of antiviral treatment are crucial for optimizing care in critically ill patients. This review provides a comprehensive synthesis of available evidence, emphasizing the need for additional investigation to establish the role of CMV treatment or prophylaxis in the management of severe COVID-19 and to develop a framework for future research on this topic.
Collapse
Affiliation(s)
| | - Vasiliki Moustaka
- Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Eleni Polyzou
- Medical School, University of Patras, 26504 Patras, Greece
- Department of Internal Medicine and Infectious Diseases, University General Hospital of Patras, 26504 Patras, Greece
| | - Maria Panagiota Almyroudi
- Department of Emergency Medicine, University Hospital ATTIKON, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - George Dimopoulos
- 3rd Department of Critical Care, EVGENIDIO Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Karolina Akinosoglou
- Medical School, University of Patras, 26504 Patras, Greece
- Department of Internal Medicine and Infectious Diseases, University General Hospital of Patras, 26504 Patras, Greece
| |
Collapse
|
13
|
Herb S, Zeleznjak J, Hennig T, L'Hernault A, Lodha M, Jürges C, Trsan T, Juranic Lisnic V, Jonjic S, Erhard F, Krmpotic A, Dölken L. Two murine cytomegalovirus microRNAs target the major viral immediate early 3 gene. J Gen Virol 2022; 103. [DOI: 10.1099/jgv.0.001804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Human cytomegalovirus is responsible for morbidity and mortality in immune compromised patients and is the leading viral cause of congenital infection. Virus-encoded microRNAs (miRNAs) represent interesting targets for novel antiviral agents. While many cellular targets that augment productive infection have been identified in recent years, regulation of viral genes such as the major viral immediate early protein 72 (IE72) by hcmv-miR-UL112-1 may contribute to both the establishment and the maintenance of latent infection. We employed photoactivated ribonucleotide-enhanced individual nucleotide resolution crosslinking (PAR-iCLIP) to identify murine cytomegalovirus (MCMV) miRNA targets during lytic infection. While the PAR-iCLIP data were of insufficient quality to obtain a comprehensive list of cellular and viral miRNA targets, the most prominent PAR-iCLIP peak in the MCMV genome mapped to the 3′ untranslated region of the major viral immediate early 3 (ie3) transcript. We show that this results from two closely positioned binding sites for the abundant MCMV miRNAs miR-M23-2-3p and miR-m01-2-3p. Their pre-expression significantly impaired viral plaque formation. However, mutation of the respective binding sites did not alter viral fitness during acute or subacute infection in vivo. Furthermore, no differences in the induction of virus-specific CD8+ T cells were observed. Future studies will probably need to go beyond studying immunocompetent laboratory mice housed in pathogen-free conditions to reveal the functional relevance of viral miRNA-mediated regulation of key viral immediate early genes.
Collapse
Affiliation(s)
- Stefanie Herb
- Institute for Virology and Immunobiology, Julius-Maximilians-University Würzburg, Versbacherstr. 7, 97078, Würzburg, Germany
| | - Jelena Zeleznjak
- Department of Histology and Embryology/Center for Proteomics, Faculty of Medicine, University of Rijeka, B. Branchetta 20, 51 000 Rijeka, Croatia
| | - Thomas Hennig
- Institute for Virology and Immunobiology, Julius-Maximilians-University Würzburg, Versbacherstr. 7, 97078, Würzburg, Germany
| | - Anne L'Hernault
- Department of Medicine, University of Cambridge, Box 157, Addenbrookes Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Manivel Lodha
- Institute for Virology and Immunobiology, Julius-Maximilians-University Würzburg, Versbacherstr. 7, 97078, Würzburg, Germany
| | - Christopher Jürges
- Institute for Virology and Immunobiology, Julius-Maximilians-University Würzburg, Versbacherstr. 7, 97078, Würzburg, Germany
| | - Tihana Trsan
- Department of Histology and Embryology/Center for Proteomics, Faculty of Medicine, University of Rijeka, B. Branchetta 20, 51 000 Rijeka, Croatia
| | - Vanda Juranic Lisnic
- Department of Histology and Embryology/Center for Proteomics, Faculty of Medicine, University of Rijeka, B. Branchetta 20, 51 000 Rijeka, Croatia
| | - Stipan Jonjic
- Department of Histology and Embryology/Center for Proteomics, Faculty of Medicine, University of Rijeka, B. Branchetta 20, 51 000 Rijeka, Croatia
| | - Florian Erhard
- Institute for Virology and Immunobiology, Julius-Maximilians-University Würzburg, Versbacherstr. 7, 97078, Würzburg, Germany
| | - Astrid Krmpotic
- Department of Histology and Embryology/Center for Proteomics, Faculty of Medicine, University of Rijeka, B. Branchetta 20, 51 000 Rijeka, Croatia
| | - Lars Dölken
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), 97080 Würzburg, Germany
- Institute for Virology and Immunobiology, Julius-Maximilians-University Würzburg, Versbacherstr. 7, 97078, Würzburg, Germany
- Department of Medicine, University of Cambridge, Box 157, Addenbrookes Hospital, Hills Road, Cambridge CB2 0QQ, UK
| |
Collapse
|
14
|
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
|
15
|
microRNA, a Subtle Indicator of Human Cytomegalovirus against Host Immune Cells. Vaccines (Basel) 2022; 10:vaccines10020144. [PMID: 35214602 PMCID: PMC8874957 DOI: 10.3390/vaccines10020144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 11/17/2022] Open
Abstract
Human cytomegalovirus (HCMV) is a double-stranded DNA virus that belongs to the β-herpesvirus family and infects 40–90% of the adult population worldwide. HCMV infection is usually asymptomatic in healthy individuals but causes serious problems in immunocompromised people. We restricted this narrative review (PubMed, January 2022) to demonstrate the interaction and molecular mechanisms between the virus and host immune cells with a focus on HCMV-encoded miRNAs. We found a series of HCMV-encoded miRNAs (e.g., miR-UL112 and miR-UL148D) are explicitly involved in the regulation of viral DNA replication, immune evasion, as well as host cell fate. MiRNA-targeted therapies have been explored for the treatment of atherosclerosis, cardiovascular disease, cancer, diabetes, and hepatitis C virus infection. It is feasible to develop an alternative vaccine to restart peripheral immunity or to inhibit HCMV activity, which may contribute to the antiviral intervention for serious HCMV-related diseases.
Collapse
|
16
|
Vinogradskaya GR, Ivanov AV, Kushch AA. Mechanisms of Survival of Cytomegalovirus-Infected Tumor Cells. Mol Biol 2022; 56:668-683. [PMID: 36217337 PMCID: PMC9534468 DOI: 10.1134/s0026893322050132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/19/2022] [Accepted: 04/19/2022] [Indexed: 11/04/2022]
Abstract
Human cytomegalovirus (HCMV) DNA and proteins are often detected in malignant tumors, warranting studies of the role that HCMV plays in carcinogenesis and tumor progression. HCMV proteins were shown to regulate the key processes involved in tumorigenesis. While HCMV as an oncogenic factor just came into focus, its ability to promote tumor progression is generally recognized. The review discusses the viral factors and cell molecular pathways that affect the resistance of cancer cells to therapy. CMV inhibits apoptosis of tumor cells, that not only promotes tumor progression, but also reduces the sensitivity of cells to antitumor therapy. Autophagy was found to facilitate either cell survival or cell death in different tumor cells. In leukemia cells, HCMV induces a "protective" autophagy that suppresses apoptosis. Viral factors that mediate drug resistance and their interactions with key cell death pathways are necessary to further investigate in order to develop agents that can restore the tumor sensitivity to anticancer drugs.
Collapse
Affiliation(s)
- G. R. Vinogradskaya
- Konstantinov St. Petersburg Institute of Nuclear Physics, National Research Center “Kurchatov Institute”, 188300 Gatchina, Leningrad oblast Russia
| | - A. V. Ivanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - A. A Kushch
- Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia
| |
Collapse
|
17
|
Pandeya A, Khalko RK, Mishra A, Singh N, Singh S, Saha S, Yadav S, Saxena S, Gosipatala SB. Human Cytomegalovirus miR-UL70-3p Downregulates the H 2O 2-Induced Apoptosis by Targeting the Modulator of Apoptosis-1 (MOAP1). Int J Mol Sci 2021; 23:ijms23010018. [PMID: 35008453 PMCID: PMC8744590 DOI: 10.3390/ijms23010018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/08/2021] [Accepted: 12/15/2021] [Indexed: 01/01/2023] Open
Abstract
Human Cytomegalovirus (HCMV) is a prototypic beta herpesvirus, causing persistent infections in humans. There are medications that are used to treat the symptoms; however, there is no cure yet. Thus, understanding the molecular mechanisms of HCMV replication and its persistence may reveal new prevention strategies. HCMV evasive strategies on the antiviral responses of the human host largely rely on its significant portion of genome. Numerous studies have highlighted the importance of miRNA-mediated regulation of apoptosis, which is an innate immune mechanism that eradicates virus-infected cells. In this study, we explore the antiapoptotic role of hcmv-miR-UL70-3p in HEK293T cells. We establish that hcmv-miR-UL70-3p targets the proapoptotic gene Modulator of Apoptosis-1 (MOAP1) through interaction with its 3'UTR region of mRNA. The ectopic expression of hcmv-miR-UL70-3p mimic significantly downregulates the H2O2-induced apoptosis through the translational repression of MOAP1. Silencing of MOAP1 through siRNA also inhibits the H2O2-induced apoptosis, which further supports the hcmv-miR-UL70-3p mediated antiapoptotic effect by regulating MOAP1 expression. These results uncover a role for hcmv-miR-UL70-3p and its target MOAP1 in regulating apoptosis.
Collapse
Affiliation(s)
- Abhishek Pandeya
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow 226025, India; (A.P.); (R.K.K.); (A.M.); (S.S.)
| | - Raj Kumar Khalko
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow 226025, India; (A.P.); (R.K.K.); (A.M.); (S.S.)
| | - Anup Mishra
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow 226025, India; (A.P.); (R.K.K.); (A.M.); (S.S.)
| | - Nishant Singh
- Developmental Toxicology Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow 226001, India; (N.S.); (S.S.); (S.Y.)
| | - Sukhveer Singh
- Developmental Toxicology Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow 226001, India; (N.S.); (S.S.); (S.Y.)
| | - Sudipta Saha
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow 226025, India;
| | - Sanjay Yadav
- Developmental Toxicology Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Lucknow 226001, India; (N.S.); (S.S.); (S.Y.)
- Department of Biochemistry, All India Institute of Medical Sciences, Rae Bareli 229405, India
| | - Sangeeta Saxena
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow 226025, India; (A.P.); (R.K.K.); (A.M.); (S.S.)
| | - Sunil Babu Gosipatala
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow 226025, India; (A.P.); (R.K.K.); (A.M.); (S.S.)
- Correspondence: or ; Tel.: +91-9455036926
| |
Collapse
|
18
|
Smith NA, Chan GC, O’Connor CM. Modulation of host cell signaling during cytomegalovirus latency and reactivation. Virol J 2021. [DOI: 10.1186/s12985-021-01674-1
expr 947873540 + 978833141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
AbstractBackgroundHuman cytomegalovirus (HCMV) resides latently in cells of the myeloid compartment, including CD34+hematopoietic progenitor cells and circulating monocytes. Healthy hosts maintain the virus latently, and this infection is, for the most part, asymptomatic. However, given the proper external cues, HCMV reactivates from latency, at which point the virus disseminates, causing disease. The viral and cellular factors dictating the balance between these phases of infection are incompletely understood, though a large body of literature support a role for viral-mediated manipulation of host cell signaling.Main bodyTo establish and maintain latency, HCMV has evolved various means by which it usurps host cell factors to alter the cellular environment to its own advantage, including altering host cell signaling cascades. As early as virus entry into myeloid cells, HCMV usurps cellular signaling to change the cellular milieu, and this regulation includes upregulation, as well as downregulation, of different signaling cascades. Indeed, given proper reactivation cues, this signaling is again altered to allow for transactivation of viral lytic genes.ConclusionsHCMV modulation of host cell signaling is not binary, and many of the cellular pathways altered are finely regulated, wherein the slightest modification imparts profound changes to the cellular milieu. It is also evident that viral-mediated cell signaling differs not only between these phases of infection, but also is myeloid cell type specific. Nonetheless, understanding the exact pathways and the means by which HCMV mediates them will undoubtedly provide novel targets for therapeutic intervention.
Collapse
|
19
|
Smith NA, Chan GC, O'Connor CM. Modulation of host cell signaling during cytomegalovirus latency and reactivation. Virol J 2021; 18:207. [PMID: 34663377 PMCID: PMC8524946 DOI: 10.1186/s12985-021-01674-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/30/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Human cytomegalovirus (HCMV) resides latently in cells of the myeloid compartment, including CD34+ hematopoietic progenitor cells and circulating monocytes. Healthy hosts maintain the virus latently, and this infection is, for the most part, asymptomatic. However, given the proper external cues, HCMV reactivates from latency, at which point the virus disseminates, causing disease. The viral and cellular factors dictating the balance between these phases of infection are incompletely understood, though a large body of literature support a role for viral-mediated manipulation of host cell signaling. MAIN BODY To establish and maintain latency, HCMV has evolved various means by which it usurps host cell factors to alter the cellular environment to its own advantage, including altering host cell signaling cascades. As early as virus entry into myeloid cells, HCMV usurps cellular signaling to change the cellular milieu, and this regulation includes upregulation, as well as downregulation, of different signaling cascades. Indeed, given proper reactivation cues, this signaling is again altered to allow for transactivation of viral lytic genes. CONCLUSIONS HCMV modulation of host cell signaling is not binary, and many of the cellular pathways altered are finely regulated, wherein the slightest modification imparts profound changes to the cellular milieu. It is also evident that viral-mediated cell signaling differs not only between these phases of infection, but also is myeloid cell type specific. Nonetheless, understanding the exact pathways and the means by which HCMV mediates them will undoubtedly provide novel targets for therapeutic intervention.
Collapse
Affiliation(s)
- Nicholas A Smith
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Gary C Chan
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA.
| | - Christine M O'Connor
- Department of Genomic Medicine, Infection Biology Program, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.
| |
Collapse
|
20
|
Immune Prophylaxis and Therapy for Human Cytomegalovirus Infection. Int J Mol Sci 2021; 22:ijms22168728. [PMID: 34445434 PMCID: PMC8395925 DOI: 10.3390/ijms22168728] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/29/2021] [Accepted: 07/30/2021] [Indexed: 02/06/2023] Open
Abstract
Human Cytomegalovirus (HCMV) infection is widespread and can result in severe sequelae in susceptible populations. Primary HCMV infection of naïve individuals results in life-long latency characterized by frequent and sporadic reactivations. HCMV infection elicits a robust antibody response, including neutralizing antibodies that can block the infection of susceptible cells in vitro and in vivo. Thus, antibody products and vaccines hold great promise for the prevention and treatment of HCMV, but to date, most attempts to demonstrate their safety and efficacy in clinical trials have been unsuccessful. In this review we summarize publicly available data on these products and highlight new developments and approaches that could assist in successful translation of HCMV immunotherapies.
Collapse
|
21
|
Cytomegalovirus (CMV) Infection and Latency. Pathogens 2021; 10:pathogens10030342. [PMID: 33804183 PMCID: PMC8001800 DOI: 10.3390/pathogens10030342] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 03/13/2021] [Indexed: 01/31/2023] Open
Abstract
Cytomegalovirus (CMV) is a herpesvirus that infects a majority of the human population worldwide [...].
Collapse
|