1
|
Shang Z, Li X. Human cytomegalovirus: pathogenesis, prevention, and treatment. MOLECULAR BIOMEDICINE 2024; 5:61. [PMID: 39585514 PMCID: PMC11589059 DOI: 10.1186/s43556-024-00226-7] [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: 07/16/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/26/2024] Open
Abstract
Human cytomegalovirus (HCMV) infection remains a significant global health challenge, particularly for immunocompromised individuals and newborns. This comprehensive review synthesizes current knowledge on HCMV pathogenesis, prevention, and treatment strategies. We examine the molecular mechanisms of HCMV entry, focusing on the structure and function of key envelope glycoproteins (gB, gH/gL/gO, gH/gL/pUL128-131) and their interactions with cellular receptors such as PDGFRα, NRP2, and THBD. The review explores HCMV's sophisticated immune evasion strategies, including interference with pattern recognition receptor signaling, modulation of antigen presentation, and regulation of NK and T cell responses. We highlight recent advancements in developing neutralizing antibodies, various vaccine strategies (live-attenuated, subunit, vector-based, DNA, and mRNA), antiviral compounds (both virus-targeted and host-targeted), and emerging cellular therapies such as TCR-T cell approaches. By integrating insights from structural biology, immunology, and clinical research, we identify critical knowledge gaps and propose future research directions. This analysis aims to stimulate cross-disciplinary collaborations and accelerate the development of more effective prevention and treatment strategies for HCMV infections, addressing a significant unmet medical need.
Collapse
Affiliation(s)
- Zifang Shang
- Research Experiment Center, Meizhou Academy of Medical Sciences, Meizhou People's Hospital, Meizhou, 514031, Guangdong, China.
- Guangdong Engineering Technological Research Center of Clinical Molecular Diagnosis and Antibody Drugs, Meizhou, 514031, Guangdong, China.
| | - Xin Li
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, 100101, China
| |
Collapse
|
2
|
Trevisan M, Pianezzola A, Onorati M, Apolloni L, Pistello M, Arav-Boger R, Palù G, Mercorelli B, Loregian A. Human neural progenitor cell models to study the antiviral effects and neuroprotective potential of approved and investigational human cytomegalovirus inhibitors. Antiviral Res 2024; 223:105816. [PMID: 38286212 DOI: 10.1016/j.antiviral.2024.105816] [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: 11/04/2023] [Revised: 12/19/2023] [Accepted: 01/18/2024] [Indexed: 01/31/2024]
Abstract
Human cytomegalovirus (HCMV) is the viral leading cause of congenital defects in newborns worldwide. Many aspects of congenital CMV (cCMV) infection, which currently lacks a specific treatment, as well as the main determinants of neuropathogenesis in the developing brain during HCMV infection are unclear. In this study, we modeled HCMV infection at different stages of neural development. Moreover, we evaluated the effects of both approved and investigational anti-HCMV drugs on viral replication and gene expression in two different neural progenitor cell lines, i.e., human embryonic stem cells-derived neural stem cells (NSCs) and fetus-derived neuroepithelial stem (NES) cells. Ganciclovir, letermovir, nitazoxanide, and the ozonide OZ418 reduced viral DNA synthesis and the production of infectious virus in both lines of neural progenitors. HCMV infection dysregulated the expression of genes that either are markers of neural progenitors, such as SOX2, NESTIN, PAX-6, or play a role in neurogenesis, such as Doublecortin. Treatment with antiviral drugs had different effects on HCMV-induced dysregulation of the genes under investigation. This study contributes to the understanding of the molecular mechanisms of cCMV neuropathogenesis and paves the way for further consideration of anti-HCMV drugs as candidate therapeutic agents for the amelioration of cCMV-associated neurological manifestations.
Collapse
Affiliation(s)
- Marta Trevisan
- Department of Molecular Medicine, University of Padua, Padua, Italy.
| | - Anna Pianezzola
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Marco Onorati
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, Pisa, 56127, Italy
| | - Lorenzo Apolloni
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Mauro Pistello
- Centro Retrovirus, Department of Translational Research, University of Pisa, Pisa, 56127, Italy
| | - Ravit Arav-Boger
- Department of Pediatrics, Division of Infectious Disease, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Giorgio Palù
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | | | - Arianna Loregian
- Department of Molecular Medicine, University of Padua, Padua, Italy.
| |
Collapse
|
3
|
Zhang S, Nan F, Jiang S, Zhou X, Niu D, Li J, Wang H, Zhang X, Zhang X, Wang B. CRM197-conjugated peptides vaccine of HCMV pp65 and gH induce maturation of DC and effective viral-specific T cell responses. Virulence 2023; 14:2169488. [PMID: 36723437 PMCID: PMC9897769 DOI: 10.1080/21505594.2023.2169488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Human cytomegalovirus (HCMV) infection is prevalent worldwide, and there is currently no licenced HCMV vaccine to control it. Therefore, developing an effective HCMV vaccine is a significant priority. Because of their excellent immunogenicity, the crucial components of HCMV, phosphoprotein 65 (pp65) and glycoproteins H (gH) are potential target proteins for HCMV vaccine design. In this study, we predicted and screened the dominant antigenic epitopes of B and T cells from pp65 and gH conjugated with the carrier protein cross-reacting material 197 (CRM197) to form three peptide-CRM197 vaccines (pp65-CRM197, gH-CRM197, and pp65-CRM197+gH-CRM197). Furthermore, the immunogenicity of the peptide-CRM197 vaccines and their effects on dendritic cells (DCs) were explored. The results showed that three peptide-CRM197 vaccines could induce maturation of DCs through the p38 MAPK signalling pathway and promote the release of proinflammatory factors, such as TNF-α and interleukin (IL) -6. Meanwhile, the peptide-CRM197 vaccines could effectively activate T cell and humoral immunity, which were far better than the inactivated HCMV vaccine. In conclusion, we constructed three peptide-CRM197 vaccines, which could induce multiple immune effects, providing a novel approach for HCMV vaccine design.
Collapse
Affiliation(s)
- Shuyun Zhang
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China,CONTACT Bin Wang Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine Qingdao University, Qingdao, China
| | - Fulong Nan
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China,CONTACT Bin Wang Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine Qingdao University, Qingdao, China
| | - Shasha Jiang
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xiaoqiong Zhou
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Delei Niu
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jun Li
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Hui Wang
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xueming Zhang
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xianjuan Zhang
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Bin Wang
- Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China,CONTACT Bin Wang Department of Pathogenic Biology, Department of Special Medicine, School of Basic Medicine Qingdao University, Qingdao, China
| |
Collapse
|
4
|
Turner DL, Mathias RA. The human cytomegalovirus decathlon: Ten critical replication events provide opportunities for restriction. Front Cell Dev Biol 2022; 10:1053139. [PMID: 36506089 PMCID: PMC9732275 DOI: 10.3389/fcell.2022.1053139] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 11/10/2022] [Indexed: 11/27/2022] Open
Abstract
Human cytomegalovirus (HCMV) is a ubiquitous human pathogen that can cause severe disease in immunocompromised individuals, transplant recipients, and to the developing foetus during pregnancy. There is no protective vaccine currently available, and with only a limited number of antiviral drug options, resistant strains are constantly emerging. Successful completion of HCMV replication is an elegant feat from a molecular perspective, with both host and viral processes required at various stages. Remarkably, HCMV and other herpesviruses have protracted replication cycles, large genomes, complex virion structure and complicated nuclear and cytoplasmic replication events. In this review, we outline the 10 essential stages the virus must navigate to successfully complete replication. As each individual event along the replication continuum poses as a potential barrier for restriction, these essential checkpoints represent potential targets for antiviral development.
Collapse
Affiliation(s)
- Declan L. Turner
- Department of Microbiology, Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Rommel A. Mathias
- Department of Microbiology, Infection and Immunity Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
5
|
Abuduxikuer K, Wang L, Zou L, Cao CY, Yu L, Guo HM, Liang XM, Wang JS, Chen L. Updated clinical and glycomic features of mannosyl-oligosaccharide glucosidase deficiency: Two case reports. World J Clin Cases 2022; 10:7397-7408. [PMID: 36158009 PMCID: PMC9353930 DOI: 10.12998/wjcc.v10.i21.7397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/16/2021] [Accepted: 06/17/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Mannosyl-oligosaccharide glucosidase (MOGS) deficiency is an extremely rare type of congenital disorder of glycosylation (CDG), with only 12 reported cases. Its clinical, genetic, and glycomic features are still expanding. Our aim is to update the novel clinical and glycosylation features of 2 previously reported patients with MOGS-CDG.
CASE SUMMARY We collected comprehensive clinical information, and conducted the immunoglobulin G1 glycosylation assay using nano-electrospray ionization source quadruple time-of-flight mass spectrometry. Novel dysmorphic features included an enlarged tongue, forwardly rotated earlobes, a birth mark, overlapped toes, and abnormal fat distribution. Novel imaging findings included pericardial effusion, a deep interarytenoid groove, mild congenital subglottic stenosis, and laryngomalacia. Novel laboratory findings included peripheral leukocytosis with neutrophil predominance, elevated C-reactive protein and creatine kinase, dyslipidemia, coagulopathy, complement 3 and complement 4 deficiencies, decreased proportions of T lymphocytes and natural killer cells, and increased serum interleukin 6. Glycosylation studies showed a significant increase of hypermannosylated glycopeptides (Glc3Man7GlcNAc2/N2H10 and Man5GlcNAc2/N2H5) and hypersialylated glycopeptides. A compensatory glycosylation pathway leading to an increase in Man5GlcNAc2/N2H5 was indicated with the glycosylation profile.
CONCLUSION We confirmed abnormal glycomics in 1 patient, expanding the clinical and glycomic spectrum of MOGS-CDG. We also postulated a compensatory glycosylation pathway, leading to a possible serum biomarker for future diagnosis.
Collapse
Affiliation(s)
| | - Lei Wang
- Department of Research and Development, SysDiagno Biomedtech, Nanjing 211800, Jiangsu Province, China
| | - Lin Zou
- Department of Medical Microbiology, Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Cui-Yan Cao
- Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian 116023, Liaoning Province, China
| | - Long Yu
- Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian 116023, Liaoning Province, China
| | - Hong-Mei Guo
- Department of Gastroenterology, Children's Hospital Affiliated to Nanjing Medical University, Nanjing 210008, Jiangsu Province, China
| | - Xin-Miao Liang
- Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian 116023, Liaoning Province, China
| | - Jian-She Wang
- Department of Hepatology, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Li Chen
- Department of Medical Microbiology, Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
6
|
Papadakis G, Gerasi M, Snoeck R, Marakos P, Andrei G, Lougiakis N, Pouli N. Synthesis of New Imidazopyridine Nucleoside Derivatives Designed as Maribavir Analogues. Molecules 2020; 25:molecules25194531. [PMID: 33022923 PMCID: PMC7582934 DOI: 10.3390/molecules25194531] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 01/26/2023] Open
Abstract
The strong inhibition of Human Cytomegalovirus (HCMV) replication by benzimidazole nucleosides, like Triciribine and Maribavir, has prompted us to expand the structure-activity relationships of the benzimidazole series, using as a central core the imidazo[4,5-b]pyridine scaffold. We have thus synthesized a number of novel amino substituted imidazopyridine nucleoside derivatives, which can be considered as 4-(or 7)-aza-d-isosters of Maribavir and have evaluated their potential antiviral activity. The target compounds were synthesized upon glycosylation of suitably substituted 2-aminoimidazopyridines, which were prepared in six steps starting from 2-amino-6-chloropyridine. Even if the new compounds possessed only a slight structural modification when compared to the original drug, they were not endowed with interesting antiviral activity. Even so, three derivatives showed promising cytotoxic potential.
Collapse
Affiliation(s)
- Georgios Papadakis
- Division of Pharmaceutical Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, 15771 Athens, Greece; (G.P.); (M.G.); (P.M.); (N.P.)
| | - Maria Gerasi
- Division of Pharmaceutical Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, 15771 Athens, Greece; (G.P.); (M.G.); (P.M.); (N.P.)
| | - Robert Snoeck
- Laboratory of Virology & Chemotherapy, Rega Institute, KU Leuven, 3000 Leuven, Belgium; (R.S.); (G.A.)
| | - Panagiotis Marakos
- Division of Pharmaceutical Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, 15771 Athens, Greece; (G.P.); (M.G.); (P.M.); (N.P.)
| | - Graciela Andrei
- Laboratory of Virology & Chemotherapy, Rega Institute, KU Leuven, 3000 Leuven, Belgium; (R.S.); (G.A.)
| | - Nikolaos Lougiakis
- Division of Pharmaceutical Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, 15771 Athens, Greece; (G.P.); (M.G.); (P.M.); (N.P.)
- Correspondence:
| | - Nicole Pouli
- Division of Pharmaceutical Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, 15771 Athens, Greece; (G.P.); (M.G.); (P.M.); (N.P.)
| |
Collapse
|
7
|
The Clinically Approved Antifungal Drug Posaconazole Inhibits Human Cytomegalovirus Replication. Antimicrob Agents Chemother 2020; 64:AAC.00056-20. [PMID: 32690644 DOI: 10.1128/aac.00056-20] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 05/11/2020] [Indexed: 12/14/2022] Open
Abstract
Posaconazole (PCZ) is a clinically approved drug used predominantly for prophylaxis and salvage therapy of fungal infections. Here, we report its previously undescribed anti-human cytomegalovirus (HCMV) activity. By using antiviral assays, we demonstrated that PCZ, along with other azolic antifungals, has a broad anti-HCMV activity, being active against different strains, including low-passage-number clinical isolates and strains resistant to viral DNA polymerase inhibitors. Using a pharmacological approach, we identified the inhibition of human cytochrome P450 51 (hCYP51), or lanosterol 14α demethylase, a cellular target of posaconazole in infected cells, as a mechanism of anti-HCMV activity of the drug. Indeed, hCYP51 expression was stimulated upon HCMV infection, and the inhibition of its enzymatic activity by either the lanosterol analog VFV {(R)-N-(1-(3,4'-difluoro-[1,1'-biphenyl]-4-yl)-2-(1H-imidazol-1-yl)ethyl)-4-(5-phenyl-1,3,4-oxadiazol-2-yl)benzamide} or PCZ decreased HCMV yield and infectivity of released virus particles. Importantly, we observed that the activity of the first-line anti-HCMV drug ganciclovir was boosted tenfold by PCZ and that ganciclovir (GCV) and PCZ act synergistically in inhibiting HCMV replication. Taken together, these findings suggest that this clinically approved drug deserves further investigation in the development of host-directed antiviral strategies as a candidate anti-HCMV drug with a dual antimicrobial effect.
Collapse
|
8
|
The Cytomegalovirus Protein Kinase pUL97:Host Interactions, Regulatory Mechanisms and Antiviral Drug Targeting. Microorganisms 2020; 8:microorganisms8040515. [PMID: 32260430 PMCID: PMC7232230 DOI: 10.3390/microorganisms8040515] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/02/2020] [Accepted: 04/02/2020] [Indexed: 12/25/2022] Open
Abstract
Human cytomegalovirus (HCMV) expresses a variety of viral regulatory proteins that undergo close interaction with host factors including viral-cellular multiprotein complexes. The HCMV protein kinase pUL97 represents a viral cyclin-dependent kinase ortholog (vCDK) that determines the efficiency of HCMV replication via phosphorylation of viral and cellular substrates. A hierarchy of functional importance of individual pUL97-mediated phosphorylation events has been discussed; however, the most pronounced pUL97-dependent phenotype could be assigned to viral nuclear egress, as illustrated by deletion of the UL97 gene or pharmacological pUL97 inhibition. Despite earlier data pointing to a cyclin-independent functionality, experimental evidence increasingly emphasized the role of pUL97-cyclin complexes. Consequently, the knowledge about pUL97 involvement in host interaction, viral nuclear egress and additional replicative steps led to the postulation of pUL97 as an antiviral target. Indeed, validation experiments in vitro and in vivo confirmed the sustainability of this approach. Consequently, current investigations of pUL97 in antiviral treatment go beyond the known pUL97-mediated ganciclovir prodrug activation and henceforward include pUL97-specific kinase inhibitors. Among a number of interesting small molecules analyzed in experimental and preclinical stages, maribavir is presently investigated in clinical studies and, in the near future, might represent a first kinase inhibitor applied in the field of antiviral therapy.
Collapse
|
9
|
Lee JH, Pasquarella JR, Kalejta RF. Cell Line Models for Human Cytomegalovirus Latency Faithfully Mimic Viral Entry by Macropinocytosis and Endocytosis. J Virol 2019; 93:e01021-19. [PMID: 31391271 PMCID: PMC6803280 DOI: 10.1128/jvi.01021-19] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 08/05/2019] [Indexed: 02/06/2023] Open
Abstract
Human cytomegalovirus (HCMV) enters primary CD34+ hematopoietic progenitor cells by macropinocytosis, where it establishes latency in part because its tegument-transactivating protein, pp71, remains associated with endosomes and is therefore unable to initiate productive, lytic replication. Here we show that multiple HCMV strains also enter cell line models used to study latency by macropinocytosis and endocytosis. In all latency models tested, tegument-delivered pp71 was found to be colocalized with endosomal markers and was not associated with the seven other cytoplasmic localization markers tested. Like the capsid-associated pp150 tegument protein, we initially detected capsid proteins in association with endosomes but later detected them in the nucleus. Inhibitors of macropinocytosis and endocytosis reduced latent viral gene expression and precluded reactivation. Importantly, we utilized electron microscopy to observe entry by macropinocytosis and endocytosis, providing additional visual corroboration of the findings of our functional studies. Our demonstration that HCMV enters cell line models for latency in a manner indistinguishable from that of its entry into primary cells illustrates the utility of these cell lines for probing the mechanisms, host genetics, and small-molecule-mediated inhibition of HCMV entry into the cell types where it establishes latency.IMPORTANCE Primary cells cultured in vitro currently provide the highest available relevance for examining molecular and genetic requirements for the establishment, maintenance, and reactivation of HCMV latency. However, their expense, heterogeneity, and intransigence to both long-term culture and molecular or genetic modification create rigor and reproducibility challenges for HCMV latency studies. There are several cell line models for latency not obstructed by deficiencies inherent in primary cells. However, many researchers view cell line studies of latency to be physiologically irrelevant because of the perception that these models display numerous and significant differences from primary cells. Here, we show that the very first step in a latent HCMV infection, entry of the virus into cells, occurs in cell line models in a manner indistinguishable from that in which it occurs in primary CD34+ hematopoietic progenitor cells. Our data argue that experimental HCMV latency is much more similar than it is different in cell lines and primary cells.
Collapse
Affiliation(s)
- Jeong-Hee Lee
- Institute for Molecular Virology and McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Joseph R Pasquarella
- Institute for Molecular Virology and McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Robert F Kalejta
- Institute for Molecular Virology and McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
10
|
Britt WJ, Prichard MN. New therapies for human cytomegalovirus infections. Antiviral Res 2018; 159:153-174. [PMID: 30227153 DOI: 10.1016/j.antiviral.2018.09.003] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 08/28/2018] [Accepted: 09/07/2018] [Indexed: 02/07/2023]
Abstract
The recent approval of letermovir marks a new era of therapy for human cytomegalovirus (HCMV) infections, particularly for the prevention of HCMV disease in hematopoietic stem cell transplant recipients. For almost 30 years ganciclovir has been the therapy of choice for these infections and by today's standards this drug exhibits only modest antiviral activity that is often insufficient to completely suppress viral replication, and drives the selection of drug-resistant variants that continue to replicate and contribute to disease. While ganciclovir remains the therapy of choice, additional drugs that inhibit novel molecular targets, such as letermovir, will be required as highly effective combination therapies are developed not only for the treatment of immunocompromised hosts, but also for congenitally infected infants. Sustained efforts, largely in the biotech industry and academia, have identified additional highly active lead compounds that have progressed into clinical studies with varying levels of success and at least two have the potential to be approved in the near future. Some of the new drugs in the pipeline inhibit new molecular targets, remain effective against isolates that have developed resistance to existing therapies, and promise to augment existing therapeutic regimens. Here, we will describe some of the unique features of HCMV biology and discuss their effect on therapeutic needs. Existing drugs will also be discussed and some of the more promising candidates will be reviewed with an emphasis on those progressing through clinical studies. The in vitro and in vivo antiviral activity, spectrum of antiviral activity, and mechanism of action of new compounds will be reviewed to provide an update on potential new therapies for HCMV infections that have progressed significantly in recent years.
Collapse
Affiliation(s)
- William J Britt
- Department of Pediatrics, University of Alabama School of Medicine, Birmingham AL 35233-1711, USA
| | - Mark N Prichard
- Department of Pediatrics, University of Alabama School of Medicine, Birmingham AL 35233-1711, USA.
| |
Collapse
|
11
|
Murine Cytomegalovirus Protein pM91 Interacts with pM79 and Is Critical for Viral Late Gene Expression. J Virol 2018; 92:JVI.00675-18. [PMID: 29997217 DOI: 10.1128/jvi.00675-18] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 07/05/2018] [Indexed: 02/02/2023] Open
Abstract
Viral gene expression is tightly regulated during cytomegalovirus (CMV) lytic replication, but the detailed mechanism of late gene transcription remains to be fully understood. Previous studies reported that six viral proteins (named viral transactivation factors [vTFs]) supporting late gene expression were conserved in beta- and gammaherpesviruses but not in alphaherpesviruses. Here, we performed coimmunoprecipitation experiments to elucidate the organization of these six proteins in murine CMV. Our results showed that these proteins formed a complex by both direct and indirect interactions. Specifically, pM91 strongly bound to pM79 even in the absence of other vTFs. Similar to pM79, pM91 exhibited early-late expression kinetics and localized within nuclear viral replication compartments during infection. Functional analysis was also performed using the pM91-deficient virus. Real-time PCR results revealed that abrogation of M91 expression markedly reduced viral late gene expression and progeny virus production without affecting viral DNA synthesis. Using mutagenesis, we found that residues E61, D62, D89, and D96 in pM91 were required for the pM91-pM79 interaction. Disruption of the interaction via E61A/D62A or D89A/D96A double mutation in the context of virus infection inhibited progeny virus production. Our data indicate that pM91 is a component of the viral late gene transcription factor complex and that the pM91-pM79 interaction is essential for viral late gene expression.IMPORTANCE Cytomegalovirus (CMV) infection is the leading cause of birth defects and causes morbidity and mortality in immunocompromised patients. The regulation of viral late gene transcription is not well elucidated, and understanding of this process benefits the development of novel therapeutics against CMV infection. This study (i) identified that six viral transactivation factors encoded by murine CMV form a complex, (ii) demonstrated that pM91 interacts with pM79 and that pM91 and pM79 colocalize in the nuclear viral replication compartments, (iii) confirmed that pM91 is critical for viral late gene expression but dispensable for viral DNA replication, and (iv) revealed that the pM91-pM79 interaction is required for progeny virus production. These findings give an explanation of how CMV regulates late gene expression and have important implications for the design of antiviral strategies.
Collapse
|
12
|
Kornfeind EM, Visalli RJ. Human herpesvirus portal proteins: Structure, function, and antiviral prospects. Rev Med Virol 2018; 28:e1972. [PMID: 29573302 DOI: 10.1002/rmv.1972] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 01/26/2018] [Accepted: 01/27/2018] [Indexed: 01/28/2023]
Abstract
Herpesviruses (Herpesvirales) and tailed bacteriophages (Caudovirales) package their dsDNA genomes through an evolutionarily conserved mechanism. Much is known about the biochemistry and structural biology of phage portal proteins and the DNA encapsidation (viral genome cleavage and packaging) process. Although not at the same level of detail, studies on HSV-1, CMV, VZV, and HHV-8 have revealed important information on the function and structure of herpesvirus portal proteins. During dsDNA phage and herpesviral genome replication, concatamers of viral dsDNA are cleaved into single length units by a virus-encoded terminase and packaged into preformed procapsids through a channel located at a single capsid vertex (portal). Oligomeric portals are formed by the interaction of identical portal protein monomers. Comparing portal protein primary aa sequences between phage and herpesviruses reveals little to no sequence similarity. In contrast, the secondary and tertiary structures of known portals are remarkable. In all cases, function is highly conserved in that portals are essential for DNA packaging and also play a role in releasing viral genomic DNA during infection. Preclinical studies have described small molecules that target the HSV-1 and VZV portals and prevent viral replication by inhibiting encapsidation. This review summarizes what is known concerning the structure and function of herpesvirus portal proteins primarily based on their conserved bacteriophage counterparts and the potential to develop novel portal-specific DNA encapsidation inhibitors.
Collapse
Affiliation(s)
- Ellyn M Kornfeind
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA, USA
| | - Robert J Visalli
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA, USA
| |
Collapse
|
13
|
Yamada KH, Majima R, Yamaguchi T, Inoue N. Characterization of phenyl pyrimidine derivatives that inhibit cytomegalovirus immediate-early gene expression. Antivir Chem Chemother 2018; 26:2040206618763193. [PMID: 29546767 PMCID: PMC5890547 DOI: 10.1177/2040206618763193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Background Previously, we established a reporter cell line for human cytomegalovirus and screened anti-human cytomegalovirus compounds using the cell line. In this study, we characterized one of the identified compounds, 2,4-diamino-6–(4-methoxyphenyl)pyrimidine (coded as 35C10). Methods 50% Effective concentrations (EC50s) and 50% cytotoxic concentrations (CC50s) of 35C10 and its derivatives in human fibroblasts were determined by X-gal staining of the cells infected with human cytomegalovirus Towne strain expressing β-galactosidase. Results EC50 and CC50 of 35C10 were 4.3 µM and >200 µM, respectively. Among several 35C10 derivatives, only one lacking 4-amino group of pyrimidine showed a similar EC50. 35C10 weakly inhibited murine cytomegalovirus, herpes simplex virus type 1, and varicella-zoster virus. A “time of addition” experiment suggested that 35C10 inhibited an early phase of the infection. Although 35C10 did not inhibit viral attachment to the cells nor the delivery of viral DNA to the nuclei, it decreased the number of infected cells expressing immediate-early 1 and 2 (IE1/IE2) proteins. 35C10 also inhibited the activation of a promoter for TRL4 in the reporter cells upon human cytomegalovirus infection, but not in the same reporter cells transfected with a plasmid expressing IE2. Conclusion Our findings suggest that 35C10 is a novel compound that inhibits IE gene expression in human cytomegalovirus-infected cells.
Collapse
Affiliation(s)
- Koh-Hei Yamada
- 1 Department of Microbiology and Immunology, Gifu Pharmaceutical University, Gifu, Japan
| | - Ryuichi Majima
- 1 Department of Microbiology and Immunology, Gifu Pharmaceutical University, Gifu, Japan
| | - Toyofumi Yamaguchi
- 2 Department of Biosciences, Teikyo University of Science and Technology, Tokyo, Japan
| | - Naoki Inoue
- 1 Department of Microbiology and Immunology, Gifu Pharmaceutical University, Gifu, Japan
| |
Collapse
|
14
|
Mercorelli B, Luganini A, Celegato M, Palù G, Gribaudo G, Loregian A. Repurposing the clinically approved calcium antagonist manidipine dihydrochloride as a new early inhibitor of human cytomegalovirus targeting the Immediate-Early 2 (IE2) protein. Antiviral Res 2018; 150:130-136. [DOI: 10.1016/j.antiviral.2017.12.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 12/14/2017] [Accepted: 12/19/2017] [Indexed: 01/04/2023]
|
15
|
Strang BL. RO0504985 is an inhibitor of CMGC kinase proteins and has anti-human cytomegalovirus activity. Antiviral Res 2017; 144:21-26. [DOI: 10.1016/j.antiviral.2017.05.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 05/08/2017] [Indexed: 12/17/2022]
|
16
|
Abstract
Viruses are major pathogenic agents causing a variety of serious diseases in humans, other animals, and plants. Drugs that combat viral infections are called antiviral drugs. There are no effective antiviral drugs for many viral infections. However, there are several drugs for influenza, a couple of drugs for herpesviruses, and some new antiviral drugs for treatment of HIV and hepatitis C infections. The arsenal of antivirals is complex. As of March 2014, it consists of approximately 50 drugs approved by the FDA, approximately half of which are directed against HIV. Antiviral drug creation strategies are focused on two different approaches: targeting the viruses themselves or targeting host cell factors. Direct virus-targeting antiviral drugs include attachment inhibitors, entry inhibitors, uncoating inhibitors, protease inhibitors, polymerase inhibitors, nucleoside and nucleotide reverse transcriptase inhibitors, nonnucleoside reverse-transcriptase inhibitors, and integrase inhibitors. Protease inhibitors (darunavir, atazanavir, and ritonavir), viral DNA polymerase inhibitors (acyclovir, valacyclovir, valganciclovir, and tenofovir), and an integrase inhibitor (raltegravir) are included in the list of Top 200 Drugs by sales for the 2010s.
Collapse
|
17
|
Ciáurriz M, Zabalza A, Beloki L, Mansilla C, Pérez-Valderrama E, Lachén M, Bandrés E, Olavarría E, Ramírez N. The immune response to cytomegalovirus in allogeneic hematopoietic stem cell transplant recipients. Cell Mol Life Sci 2015; 72:4049-62. [PMID: 26174234 PMCID: PMC11113937 DOI: 10.1007/s00018-015-1986-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 06/22/2015] [Accepted: 07/03/2015] [Indexed: 02/08/2023]
Abstract
Approximately, up to 70 % of the human population is infected with cytomegalovirus (CMV) that persists for life in a latent state. In healthy people, CMV reactivation induces the expansion of CMV-specific T cells up to 10 % of the entire T cell repertoire. On the contrary, CMV infection is a major opportunistic viral pathogen that remains a leading cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation. Due to the delayed CMV-specific immune recovery, the incidence of CMV reactivation during post-transplant period is very high. Several methods are currently available for the monitoring of CMV-specific responses that help in clinical monitoring. In this review, essential aspects in the immune recovery against CMV are discussed to improve the better understanding of the immune system relying on CMV infection and, thereby, helping the avoidance of CMV disease or reactivation following hematopoietic stem cell transplantation with severe consequences for the transplanted patients.
Collapse
Affiliation(s)
- Miriam Ciáurriz
- Oncohematology Research Group, Navarrabiomed-Fundación Miguel Servet, IDISNA (Navarra's Health Research Institute), Irunlarrea 3 Street, 31008, Pamplona, Navarra, Spain
| | - Amaya Zabalza
- Oncohematology Research Group, Navarrabiomed-Fundación Miguel Servet, IDISNA (Navarra's Health Research Institute), Irunlarrea 3 Street, 31008, Pamplona, Navarra, Spain
- Hematology Department, Complejo Hospitalario de Navarra, Navarra Health Service, IDISNA, Pamplona, Navarra, Spain
| | - Lorea Beloki
- Oncohematology Research Group, Navarrabiomed-Fundación Miguel Servet, IDISNA (Navarra's Health Research Institute), Irunlarrea 3 Street, 31008, Pamplona, Navarra, Spain
| | - Cristina Mansilla
- Oncohematology Research Group, Navarrabiomed-Fundación Miguel Servet, IDISNA (Navarra's Health Research Institute), Irunlarrea 3 Street, 31008, Pamplona, Navarra, Spain
| | - Estela Pérez-Valderrama
- Oncohematology Research Group, Navarrabiomed-Fundación Miguel Servet, IDISNA (Navarra's Health Research Institute), Irunlarrea 3 Street, 31008, Pamplona, Navarra, Spain
| | - Mercedes Lachén
- Oncohematology Research Group, Navarrabiomed-Fundación Miguel Servet, IDISNA (Navarra's Health Research Institute), Irunlarrea 3 Street, 31008, Pamplona, Navarra, Spain
| | - Eva Bandrés
- Oncohematology Research Group, Navarrabiomed-Fundación Miguel Servet, IDISNA (Navarra's Health Research Institute), Irunlarrea 3 Street, 31008, Pamplona, Navarra, Spain
- Hematology Department, Complejo Hospitalario de Navarra, Navarra Health Service, IDISNA, Pamplona, Navarra, Spain
- Immunity Unit, Complejo Hospitalario de Navarra, Navarra Health Service, IDISNA, Pamplona, Navarra, Spain
| | - Eduardo Olavarría
- Oncohematology Research Group, Navarrabiomed-Fundación Miguel Servet, IDISNA (Navarra's Health Research Institute), Irunlarrea 3 Street, 31008, Pamplona, Navarra, Spain
- Hammersmith Hospital-Imperial College Healthcare NHS, London, UK
| | - Natalia Ramírez
- Oncohematology Research Group, Navarrabiomed-Fundación Miguel Servet, IDISNA (Navarra's Health Research Institute), Irunlarrea 3 Street, 31008, Pamplona, Navarra, Spain.
| |
Collapse
|
18
|
Tabata T, Petitt M, Zydek M, Fang-Hoover J, Larocque N, Tsuge M, Gormley M, Kauvar LM, Pereira L. Human cytomegalovirus infection interferes with the maintenance and differentiation of trophoblast progenitor cells of the human placenta. J Virol 2015; 89:5134-47. [PMID: 25741001 PMCID: PMC4403461 DOI: 10.1128/jvi.03674-14] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 01/19/2015] [Indexed: 12/12/2022] Open
Abstract
UNLABELLED Human cytomegalovirus (HCMV) is a major cause of birth defects that include severe neurological deficits, hearing and vision loss, and intrauterine growth restriction. Viral infection of the placenta leads to development of avascular villi, edema, and hypoxia associated with symptomatic congenital infection. Studies of primary cytotrophoblasts (CTBs) revealed that HCMV infection impedes terminal stages of differentiation and invasion by various molecular mechanisms. We recently discovered that HCMV arrests earlier stages involving development of human trophoblast progenitor cells (TBPCs), which give rise to the mature cell types of chorionic villi-syncytiotrophoblasts on the surfaces of floating villi and invasive CTBs that remodel the uterine vasculature. Here, we show that viral proteins are present in TBPCs of the chorion in cases of symptomatic congenital infection. In vitro studies revealed that HCMV replicates in continuously self-renewing TBPC lines derived from the chorion and alters expression and subcellular localization of proteins required for cell cycle progression, pluripotency, and early differentiation. In addition, treatment with a human monoclonal antibody to HCMV glycoprotein B rescues differentiation capacity, and thus, TBPCs have potential utility for evaluation of the efficacies of novel antiviral antibodies in protecting and restoring placental development. Our results suggest that HCMV replicates in TBPCs in the chorion in vivo, interfering with the earliest steps in the growth of new villi, contributing to virus transmission and impairing compensatory development. In cases of congenital infection, reduced responsiveness of the placenta to hypoxia limits the transport of substances from maternal blood and contributes to fetal growth restriction. IMPORTANCE Human cytomegalovirus (HCMV) is a leading cause of birth defects in the United States. Congenital infection can result in permanent neurological defects, mental retardation, hearing loss, visual impairment, and pregnancy complications, including intrauterine growth restriction, preterm delivery, and stillbirth. Currently, there is neither a vaccine nor any approved treatment for congenital HCMV infection during gestation. The molecular mechanisms underlying structural deficiencies in the placenta that undermine fetal development are poorly understood. Here we report that HCMV replicates in trophoblast progenitor cells (TBPCs)-precursors of the mature placental cells, syncytiotrophoblasts and cytotrophoblasts, in chorionic villi-in clinical cases of congenital infection. Virus replication in TBPCs in vitro dysregulates key proteins required for self-renewal and differentiation and inhibits normal division and development into mature placental cells. Our findings provide insights into the underlying molecular mechanisms by which HCMV replication interferes with placental maturation and transport functions.
Collapse
Affiliation(s)
- Takako Tabata
- Department of Cell and Tissue Biology, School of Dentistry, University of California, San Francisco, San Francisco, California, USA
| | - Matthew Petitt
- Department of Cell and Tissue Biology, School of Dentistry, University of California, San Francisco, San Francisco, California, USA
| | - Martin Zydek
- Department of Cell and Tissue Biology, School of Dentistry, University of California, San Francisco, San Francisco, California, USA
| | - June Fang-Hoover
- Department of Cell and Tissue Biology, School of Dentistry, University of California, San Francisco, San Francisco, California, USA
| | - Nicholas Larocque
- Center for Reproductive Sciences, University of California, San Francisco, San Francisco, California, USA Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, California, USA The Eli & Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, USA
| | - Mitsuru Tsuge
- Department of Cell and Tissue Biology, School of Dentistry, University of California, San Francisco, San Francisco, California, USA
| | - Matthew Gormley
- Center for Reproductive Sciences, University of California, San Francisco, San Francisco, California, USA Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, San Francisco, California, USA The Eli & Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, USA
| | | | - Lenore Pereira
- Department of Cell and Tissue Biology, School of Dentistry, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
19
|
Gambarino S, Callea S, Rizzo G, Montanari P, Loiacono E, Bergallo M. Evaluation of UL99 transcript as a target for antiviral treatment efficacy. J Virol Methods 2014; 207:104-9. [PMID: 24977314 DOI: 10.1016/j.jviromet.2014.06.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 01/31/2014] [Accepted: 06/06/2014] [Indexed: 11/20/2022]
Abstract
Human cytomegalovirus (HCMV) is a virus belonging to the Beta Herpes virus family. Its genome contains many different genes clustered in immediate early, early and late genes. This last cluster includes UL99, a late gene that encodes for a tegument protein called pp28. In immunocompetent patients, HCMV infection occurs asymptomatically, while its reactivation in immunocompromised patients can be a cause of pneumonia, retinitis and gastrointestinal diseases. To prevent or to contrast HCMV infection, several drugs (such as Ganciclovir, Acyclovir, Foscarnet) are available, and their efficiency is evaluated by HCMV DNA load monitoring, as also for antiviral resistance onset that may occur after the therapy. In this study is described the development of a Real Time PCR for the detection and quantification of UL99 transcript and the clearance of this target compared to HCMV DNA, both in vitro and in vivo on bronchoalveolar lavage samples.
Collapse
Affiliation(s)
- Stefano Gambarino
- Department of Public Health and Pediatric Sciences, University of Turin, Medical School, Turin, Italy.
| | - Stefano Callea
- Department of Public Health and Pediatric Sciences, University of Turin, Medical School, Turin, Italy
| | - Giovanna Rizzo
- Department of Public Health and Pediatric Sciences, University of Turin, Medical School, Turin, Italy
| | - Paola Montanari
- Department of Public Health and Pediatric Sciences, University of Turin, Medical School, Turin, Italy
| | - Elisa Loiacono
- Department of Public Health and Pediatric Sciences, University of Turin, Medical School, Turin, Italy
| | - Massimiliano Bergallo
- Department of Public Health and Pediatric Sciences, University of Turin, Medical School, Turin, Italy
| |
Collapse
|
20
|
Zydek M, Petitt M, Fang-Hoover J, Adler B, Kauvar LM, Pereira L, Tabata T. HCMV infection of human trophoblast progenitor cells of the placenta is neutralized by a human monoclonal antibody to glycoprotein B and not by antibodies to the pentamer complex. Viruses 2014; 6:1346-64. [PMID: 24651029 PMCID: PMC3970154 DOI: 10.3390/v6031346] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 02/27/2014] [Accepted: 02/27/2014] [Indexed: 12/17/2022] Open
Abstract
Human cytomegalovirus (HCMV) is the major viral cause of congenital infection and birth defects. Primary maternal infection often results in virus transmission, and symptomatic babies can have permanent neurological deficiencies and deafness. Congenital infection can also lead to intrauterine growth restriction, a defect in placental transport. HCMV replicates in primary cytotrophoblasts (CTBs), the specialized cells of the placenta, and inhibits differentiation/invasion. Human trophoblast progenitor cells (TBPCs) give rise to the mature cell types of the chorionic villi, CTBs and multi-nucleated syncytiotrophoblasts (STBs). Here we report that TBPCs are fully permissive for pathogenic and attenuated HCMV strains. Studies with a mutant virus lacking a functional pentamer complex (gH/gL/pUL128-131A) showed that virion entry into TBPCs is independent of the pentamer. In addition, infection is blocked by a potent human neutralizing monoclonal antibody (mAb), TRL345, reactive with glycoprotein B (gB), but not mAbs to the pentamer proteins pUL130/pUL131A. Functional studies revealed that neutralization of infection preserved the capacity of TBPCs to differentiate and assemble into trophospheres composed of CTBs and STBs in vitro. Our results indicate that mAbs to gB protect trophoblast progenitors of the placenta and could be included in antibody treatments developed to suppress congenital infection and prevent disease.
Collapse
Affiliation(s)
- Martin Zydek
- Department of Cell and Tissue Biology, University of California San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA.
| | - Matthew Petitt
- Department of Cell and Tissue Biology, University of California San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA.
| | - June Fang-Hoover
- Department of Cell and Tissue Biology, University of California San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA.
| | - Barbara Adler
- Division of Virology, Max von Pettenkofer-Institute, Ludwig-Maximilians-University Munich, Pettenkoferstr. 9A, D-80336 Munich, Germany.
| | - Lawrence M Kauvar
- Trellis Bioscience, LLC, 2-B Corporate Drive, South San Francisco, CA 94080, USA.
| | - Lenore Pereira
- Department of Cell and Tissue Biology, University of California San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA.
| | - Takako Tabata
- Department of Cell and Tissue Biology, University of California San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA.
| |
Collapse
|
21
|
Herr W, Plachter B. Cytomegalovirus and varicella–zoster virus vaccines in hematopoietic stem cell transplantation. Expert Rev Vaccines 2014; 8:999-1021. [DOI: 10.1586/erv.09.58] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
22
|
Mercorelli B, Gribaudo G, Palù G, Loregian A. Approaches for the generation of new anti-cytomegalovirus agents: identification of protein-protein interaction inhibitors and compounds against the HCMV IE2 protein. Methods Mol Biol 2014; 1119:349-363. [PMID: 24639231 DOI: 10.1007/978-1-62703-788-4_18] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Human cytomegalovirus (HCMV) infection is responsible for severe, often even fatal, diseases in immunocompromised subjects and also represents the major cause of viral-associated congenital malformations in newborn children. The few drugs licensed for anti-HCMV therapy suffer from many drawbacks and none of them have been approved for the treatment of congenital infections. Furthermore, the emergence of drug-resistant viral strains represents a major concern for disease management. Thus, there is a strong need for new anti-HCMV drugs. Here we describe three different assays for the discovery of novel anti-HCMV compounds: two are in vitro assays, i.e., a fluorescence polarization (FP)-based assay and an enzyme-linked immunosorbent assay (ELISA), which are designed to search for compounds that act by disrupting the interactions between the HCMV DNA polymerase subunits, but in general can be employed to find inhibitors of any protein-protein interaction of interest; the third is a cell-based assay designed to identify inhibitors of the viral immediate-early 2 (IE2) protein activities.
Collapse
|
23
|
Inhibition of herpesvirus and influenza virus replication by blocking polymerase subunit interactions. Antiviral Res 2013; 99:318-27. [DOI: 10.1016/j.antiviral.2013.05.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Revised: 05/24/2013] [Accepted: 05/29/2013] [Indexed: 11/18/2022]
|
24
|
Novikov MS, Babkov DA, Paramonova MP, Khandazhinskaya AL, Ozerov AA, Chizhov AO, Andrei G, Snoeck R, Balzarini J, Seley-Radtke KL. Synthesis and anti-HCMV activity of 1-[ω-(phenoxy)alkyl]uracil derivatives and analogues thereof. Bioorg Med Chem 2013; 21:4151-7. [PMID: 23743443 PMCID: PMC7127185 DOI: 10.1016/j.bmc.2013.05.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 04/27/2013] [Accepted: 05/07/2013] [Indexed: 01/31/2023]
Abstract
HCMV infection represents a life-threatening condition for immunocompromised patients and newborn infants and novel anti-HCMV agents are clearly needed. In this regard, a series of 1-[ω-(phenoxy)alkyl]uracil derivatives were synthesized and examined for antiviral properties. Compounds 17, 20, 24 and 28 were found to exhibit highly specific and promising inhibitory activity against HCMV replication in HEL cell cultures with EC50 values within 5.5-12μM range. Further studies should be undertaken to elucidate the mechanism of action of these compounds and the structure-activity relationship for the linker region.
Collapse
Affiliation(s)
- Mikhail S. Novikov
- Department of Pharmaceutical & Toxicological Chemistry, Volgograd State Medical University, Pavshikh Bortsov Sq., 1, Volgograd 400131, Russia
| | - Denis A. Babkov
- Department of Pharmaceutical & Toxicological Chemistry, Volgograd State Medical University, Pavshikh Bortsov Sq., 1, Volgograd 400131, Russia
| | - Maria P. Paramonova
- Department of Pharmaceutical & Toxicological Chemistry, Volgograd State Medical University, Pavshikh Bortsov Sq., 1, Volgograd 400131, Russia
| | | | - Alexander A. Ozerov
- Department of Pharmaceutical & Toxicological Chemistry, Volgograd State Medical University, Pavshikh Bortsov Sq., 1, Volgograd 400131, Russia
| | - Alexander O. Chizhov
- Zelinsky Institute of Organic Chemistry, Russian Academy of Science, Leninsky pr., 47, Moscow 119991, Russia
| | - Graciela Andrei
- Rega Institute for Medical Research, KU Leuven, Minderbroedersstraat 10, Leuven B-3000, Belgium
| | - Robert Snoeck
- Rega Institute for Medical Research, KU Leuven, Minderbroedersstraat 10, Leuven B-3000, Belgium
| | - Jan Balzarini
- Rega Institute for Medical Research, KU Leuven, Minderbroedersstraat 10, Leuven B-3000, Belgium
| | - Katherine L. Seley-Radtke
- Department of Chemistry & Biochemistry, University of Maryland, Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, USA
| |
Collapse
|
25
|
Massari S, Mercorelli B, Sancineto L, Sabatini S, Cecchetti V, Gribaudo G, Palù G, Pannecouque C, Loregian A, Tabarrini O. Design, synthesis, and evaluation of WC5 analogues as inhibitors of human cytomegalovirus Immediate-Early 2 protein, a promising target for anti-HCMV treatment. ChemMedChem 2013; 8:1403-14. [PMID: 23757191 DOI: 10.1002/cmdc.201300106] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 05/16/2013] [Indexed: 12/13/2022]
Abstract
Although human cytomegalovirus (HCMV) infection is mostly asymptomatic for immunocompetent individuals, it remains a serious threat for those who are immunocompromised, in whom it is associated with various clinical manifestations. The therapeutic utility of the few available anti-HCMV drugs is limited by several drawbacks, including cross-resistance due to their common mechanism of action, i.e., inhibition of viral DNA polymerase. Therefore, compounds that target other essential viral events could overcome this problem. One example of this is the 6-aminoquinolone WC5, which acts by directly blocking the transactivation of essential viral Early genes by the Immediate-Early 2 (IE2) protein. In this study, the quinolone scaffold of the lead compound WC5 was investigated in depth, defining more suitable substituents for each of the scaffold positions explored and identifying novel, potent and nontoxic compounds. Some compounds showed potent anti-HCMV activity by interfering with IE2-dependent viral E gene expression. Among them, naphthyridone 1 was also endowed with potent anti-HIV activity in latently infected cells. Their antiviral profile along with their innovative mechanism of action make these anti-HCMV quinolones a very promising class of compounds to be exploited for more effective antiviral therapeutic treatment.
Collapse
Affiliation(s)
- Serena Massari
- Department of Chemistry and Technology of Drugs, University of Perugia, 06123 Perugia, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Ibrišimović M, Nagl U, Kneidinger D, Rauch M, Lion T, Klein R. Targeted expression of herpes simplex virus thymidine kinase in adenovirus-infected cells reduces virus titers upon treatment with ganciclovir in vitro. J Gene Med 2012; 14:3-19. [PMID: 22190534 DOI: 10.1002/jgm.1638] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
Abstract
BACKGROUND Adenoviruses are a frequent cause of life-threatening infections in immunocompromised patients. Available therapeutics still cannot completely prevent fatal outcomes. By contrast, herpes viruses are well treatable with prodrugs such as ganciclovir (GCV), which are selectively activated in virus-infected cells by virus-encoded thymidine kinases. This effective group of prodrugs is not applicable to adenoviruses and other DNA viruses because they lack those kinases. METHODS To render adenoviruses amenable to GCV treatment, we generated an adenoviral vector-based delivery system for targeted expression of herpes simplex virus thymidine kinase (HSV-TK) in wild-type adenovirus 5 (wt Ad5)-infected cells. HSV-TK expression was largely restricted to wt virus-infected cells by transcription of the gene from the Ad5 E4 promoter. Its activity is dependent on the adenoviral E1A gene product which is not produced by the vector but is only provided in cells infected with wt adenovirus. The anti-adenoviral effect of HSV-TK expression and concomitant treatment with GCV was assessed in vitro in four different cell lines or primary cells. RESULTS E4 promoter-mediated HSV-TK background expression was sufficiently low to prevent cytotoxicity in the presence of low-levels GCV in cells not infected with wt Ad5. However, expression was several-fold increased in wt Ad5-infected cells and treatment with low levels of GCV efficiently inhibited wt Ad5 DNA replication. Genome copy numbers and output of infectious particles were reduced by up to > 99.99% and cell viability was greatly increased. CONCLUSIONS We extended the concept of enzyme/prodrug therapy to adenovirus infections by selectively sensitizing adenovirus-infected cells to treatment with GCV.
Collapse
Affiliation(s)
- Mirza Ibrišimović
- Children's Cancer Research Institute, St. Anna Kinderkrebsforschung, Department of Pediatrics, Medical University of Vienna, Austria
| | | | | | | | | | | |
Collapse
|
27
|
Frenzel K, Ganepola S, Michel D, Thiel E, Krüger DH, Uharek L, Hofmann J. Antiviral function and efficacy of polyvalent immunoglobulin products against CMV isolates in different human cell lines. Med Microbiol Immunol 2012; 201:277-86. [DOI: 10.1007/s00430-012-0229-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Accepted: 01/19/2012] [Indexed: 11/28/2022]
|
28
|
Estudios de resistencia. ¿Cuándo están indicados? Enferm Infecc Microbiol Clin 2011; 29 Suppl 6:24-7. [DOI: 10.1016/s0213-005x(11)70053-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
29
|
Chelmoński A, Jabłecki J, Szajerka T. Insidious Course of Cytomegalovirus Infection in Hand Transplant Recipient: Case Report, Diagnostics, and Treatment. Transplant Proc 2011; 43:2827-30. [DOI: 10.1016/j.transproceed.2011.06.031] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Revised: 05/15/2011] [Accepted: 06/03/2011] [Indexed: 11/16/2022]
|
30
|
Sites and roles of phosphorylation of the human cytomegalovirus DNA polymerase subunit UL44. Virology 2011; 417:268-80. [PMID: 21784501 DOI: 10.1016/j.virol.2011.06.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Revised: 04/12/2011] [Accepted: 06/11/2011] [Indexed: 12/27/2022]
Abstract
The human cytomegalovirus DNA polymerase subunit UL44 is a phosphoprotein, but its sites and roles of phosphorylation have not been investigated. We compared sites of phosphorylation of UL44 in vitro by the viral protein kinase UL97 and cyclin-dependent kinase 1 with those in infected cells. Transient treatment of infected cells with a UL97 inhibitor greatly reduced labeling of two minor UL44 phosphopeptides. Viruses containing alanine substitutions of most UL44 residues that are phosphorylated in infected cells exhibited at most modest effects on viral DNA synthesis and yield. However, substitution of highly phosphorylated sites adjacent to the nuclear localization signal abolished viral replication. The results taken together are consistent with UL44 being phosphorylated directly by UL97 during infection, and a crucial role for phosphorylation-mediated nuclear localization of UL44 for viral replication, but lend little support to the widely held hypothesis that UL97-mediated phosphorylation of UL44 is crucial for viral DNA synthesis.
Collapse
|
31
|
Alvisi G, Marin O, Pari G, Mancini M, Avanzi S, Loregian A, Jans DA, Ripalti A. Multiple phosphorylation sites at the C-terminus regulate nuclear import of HCMV DNA polymerase processivity factor ppUL44. Virology 2011; 417:259-67. [PMID: 21741668 DOI: 10.1016/j.virol.2011.06.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Revised: 06/13/2011] [Accepted: 06/14/2011] [Indexed: 12/31/2022]
Abstract
The processivity factor of human cytomegalovirus DNA polymerase, phosphoprotein ppUL44, is essential for viral replication. During viral infection ppUL44 is phosphorylated by the viral kinase pUL97, but neither the target residues on ppUL44 nor the effect of phosphorylation on ppUL44's activity are known. We report here that ppUL44 is phosphorylated when transiently expressed in mammalian cells and coimmunoprecipitates with cellular kinases. Of three potential phosphorylation sites (S413, S415, S418) located upstream of ppUL44's nuclear localization signal (NLS) and one (T427) within the NLS itself, protein kinase CK2 (CK2) specifically phosphorylates S413, to trigger a cascade of phosphorylation of S418 and S415 by CK1 and CK2, respectively. Negative charge at the CK2/CK1 target serine residues facilitates optimal nuclear accumulation of ppUL44, whereas negative charge on T427, a potential cyclin-dependent 1 phosphorylation site, strongly decreases nuclear accumulation. Thus, nuclear transport of ppUL44 is finely tuned during viral infection through complex phosphorylation events.
Collapse
Affiliation(s)
- Gualtiero Alvisi
- Department of Hematology and Oncology Lorenzo e Ariosto Seràgnoli, University of Bologna, Medical School, Bologna, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Zahn KE, Tchesnokov EP, Götte M, Doublié S. Phosphonoformic acid inhibits viral replication by trapping the closed form of the DNA polymerase. J Biol Chem 2011; 286:25246-55. [PMID: 21566148 DOI: 10.1074/jbc.m111.248864] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Phosphonoformic acid (PFA, foscarnet) belongs to a class of antiviral drugs that inhibit the human cytomegalovirus DNA polymerase (UL54) by mimicking the pyrophosphate leaving group of the nucleotide transfer reaction. Difficulties expressing UL54 have hampered investigation of the precise structural requirements rendering inhibition by this drug. However, a previously engineered chimeric DNA polymerase, constructed by mutating the homologous polymerase from bacteriophage RB69 (gp43) to express several variable elements from UL54, can bypass this obstacle because of its favorable expression and acquired sensitivity to PFA (Tchesnokov, E. P., Obikhod, A., Schinazi, R. F., and Götte, M. (2008) J. Biol. Chem. 283, 34218-34228). Here, we compare two crystal structures that depict the chimeric DNA polymerase with and without PFA bound. PFA is visualized for the first time in the active site of a DNA polymerase, where interactions are resolved between the PP(i) mimic and two basic residues absolutely conserved in the fingers domain of family B polymerases. PFA also chelates metal ion B, the cation that contacts the triphosphate tail of the incoming nucleotide. These DNA complexes utilize a primer-template pair enzymatically chain-terminated by incorporation of acyclo-GMP, the phosphorylated form of the anti-herpes drug acyclovir. We postulate that the V478W mutation present in the chimera is critical in that it pushes the fingers domain to more readily adopt the closed conformation whether or not the drug is bound. The closed state of the fingers domain traps the variant polymerase in the untranslocated state and increases affinity for PFA. This finding provides a model for the mechanism of UL54 stalling by PFA.
Collapse
Affiliation(s)
- Karl E Zahn
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, Vermont 05405, USA
| | | | | | | |
Collapse
|
33
|
Mercorelli B, Lembo D, Palù G, Loregian A. Early inhibitors of human cytomegalovirus: state-of-art and therapeutic perspectives. Pharmacol Ther 2011; 131:309-29. [PMID: 21570424 PMCID: PMC7112563 DOI: 10.1016/j.pharmthera.2011.04.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Accepted: 04/05/2011] [Indexed: 12/31/2022]
Abstract
Human cytomegalovirus (HCMV) infection is associated with severe morbidity and mortality in immunocompromised individuals, mainly transplant recipients and AIDS patients, and is the most frequent cause of congenital malformations in newborn children. To date, few drugs are licensed for the treatment of HCMV infections, most of which target the viral DNA polymerase and suffer from many drawbacks, including long-term toxicity, low potency, and poor bioavailability. In addition, the emergence of drug-resistant viral strains is becoming an increasing problem for disease management. Finally, none of the current anti-HCMV drugs have been approved for the treatment of congenital infections. For all these reasons, there is still a strong need for new anti-HCMV drugs with novel mechanisms of action. The first events of the virus replication cycle, including attachment, entry, immediate-early gene expression, and immediate-early functions—in particular that of Immediate-Early 2 protein—represent attractive targets for the development of novel antiviral compounds. Such inhibitors would block not only the expression of viral immediate-early proteins, which play a key role in the pathogenesis of HCMV infection, but also the host immunomodulation and the changes to cell physiology induced by the first events of virus infection. This review describes the current knowledge on the initial phases of HCMV replication, their validation as potential novel antiviral targets, and the development of compounds that block such processes.
Collapse
Affiliation(s)
- Beatrice Mercorelli
- Department of Histology, Microbiology and Medical Biotechnologies, University of Padova, 35121 Padova, Italy
| | | | | | | |
Collapse
|
34
|
Lee SB, Lee CF, Ou DSC, Dulal K, Chang LH, Ma CH, Huang CF, Zhu H, Lin YS, Juan LJ. Host-viral effects of chromatin assembly factor 1 interaction with HCMV IE2. Cell Res 2011; 21:1230-47. [PMID: 21445097 DOI: 10.1038/cr.2011.53] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Chromatin assembly factor 1 (CAF1) consisting of p150, p60 and p48 is known to assemble histones onto newly synthesized DNA and thus maintain the chromatin structure. Here, we show that CAF1 expression was induced in human cytomegalovirus (HCMV)-infected cells, concomitantly with global chromatin decondensation. This apparent conflict was thought to result, in part, from CAF1 mislocalization to compartments of HCMV DNA synthesis through binding of its largest subunit p150 to viral immediate-early protein 2 (IE2). p150 interaction with p60 and IE2 facilitated HCMV DNA synthesis. The IE2Q548R mutation, previously reported to result in impaired HCMV growth with unknown mechanism, disrupted IE2/p150 and IE2/histones association in our study. Moreover, IE2 interaction with histones partly depends on p150, and the HCMV-induced chromatin decondensation was reduced in cells ectopically expressing the p150 mutant defective in IE2 binding. These results not only indicate that CAF1 was hijacked by IE2 to facilitate the replication of the HCMV genome, suggesting chromatin assembly plays an important role in herpesviral DNA synthesis, but also provide a model of the virus-induced chromatin instability through CAF1.
Collapse
Affiliation(s)
- Sung-Bau Lee
- Genomics Research Center, Academia Sinica, Taipei 115
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Prichard MN, Kern ER. The search for new therapies for human cytomegalovirus infections. Virus Res 2010; 157:212-21. [PMID: 21095209 DOI: 10.1016/j.virusres.2010.11.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Revised: 11/11/2010] [Accepted: 11/12/2010] [Indexed: 12/19/2022]
Abstract
Ganciclovir (GCV), the therapy of choice for human cytomegalovirus (CMV) infections and foscarnet, a drug used to treat GCV-resistant CMV infections was approved more than twenty years ago. Although cidofovir and a prodrug of GCV have since been added to the armamentarium, a highly effective drug without significant toxicities has yet to be approved. Such a therapeutic agent is required for treatment of immunocompromised hosts and infants, which bear the greatest burden of disease. The modest antiviral activity of existing drugs is insufficient to completely suppress viral replication, which results in the selection of drug-resistant variants that remain pathogenic, continue to replicate, and contribute to disease. Sustained efforts, largely in the biotech industry and academia, have identified highly active lead compounds that have progressed into clinical studies with varying levels of success. A few of these compounds inhibit new molecular targets, remain effective against isolates that have developed resistance to existing therapies, and promise to augment existing therapies. Some of the more promising drugs will be discussed with an emphasis on those progressing to clinical studies. Their antiviral activity both in vitro and in vivo, spectrum of antiviral activity, and mechanism of action will be reviewed to provide an update on the progress of potential new therapies for CMV infections.
Collapse
Affiliation(s)
- Mark N Prichard
- Department of Pediatrics, University of Alabama School of Medicine, Birmingham, AL 35233-1711, USA.
| | | |
Collapse
|
36
|
Modifications of C-2 on the pyrroloquinoline template aimed at the development of potent herpesvirus antivirals with improved aqueous solubility. Bioorg Med Chem Lett 2010; 20:3039-42. [DOI: 10.1016/j.bmcl.2010.03.115] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2010] [Revised: 03/27/2010] [Accepted: 03/31/2010] [Indexed: 11/21/2022]
|
37
|
The 6-aminoquinolone WC5 inhibits human cytomegalovirus replication at an early stage by interfering with the transactivating activity of viral immediate-early 2 protein. Antimicrob Agents Chemother 2010; 54:1930-40. [PMID: 20194695 DOI: 10.1128/aac.01730-09] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
WC5 is a 6-aminoquinolone that potently inhibits the replication of human cytomegalovirus (HCMV) but has no activity, or significantly less activity, against other herpesviruses. Here we investigated the nature of its specific anti-HCMV activity. Structure-activity relationship studies on a small series of analogues showed that WC5 possesses the most suitable pattern of substitutions around the quinolone scaffold to give potent and selective anti-HCMV activity. Studies performed to identify the possible target of WC5 indicated that it prevents viral DNA synthesis but does not significantly affect DNA polymerase activity. In yield reduction experiments with different multiplicities of infection, the anti-HCMV activity of WC5 appeared to be highly dependent on the viral inoculum, suggesting that WC5 may act at an initial stage of virus replication. Consistently, time-of-addition and time-of-removal studies demonstrated that WC5 affects a phase of the HCMV replicative cycle that precedes viral DNA synthesis. Experiments to monitor the effects of the compound on virus attachment and entry showed that it does not inhibit either process. Evaluation of viral mRNA and protein expression revealed that WC5 targets an event of the HCMV replicative cycle that follows the transcription and translation of immediate-early genes and precedes those of early and late genes. In cell-based assays to test the effects of WC5 on the transactivating activity of the HCMV immediate-early 2 (IE2) protein, WC5 markedly interfered with IE2-mediated transactivation of viral early promoters. Finally, WC5 combined with ganciclovir in checkerboard experiments exhibited highly synergistic activity. These findings suggest that WC5 deserves further investigation as a candidate anti-HCMV drug with a novel mechanism of action.
Collapse
|
38
|
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246. Antimicrob Agents Chemother 2010; 54:1290-7. [PMID: 20047911 DOI: 10.1128/aac.01596-09] [Citation(s) in RCA: 195] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Human cytomegalovirus (HCMV) remains a serious threat for immunocompromised individuals, including transplant recipients and newborns. To date, all drugs licensed for the treatment of HCMV infection and disease target the viral DNA polymerase. Although these drugs are effective, several drawbacks are associated with their use, including toxicity and emergence of drug resistance. Hence, new and improved antivirals with novel molecular targets are urgently needed. Here we report on the antiviral properties of AIC246, a representative of a novel class of low-molecular-weight compounds that is currently undergoing clinical phase II studies. The anti-HCMV activity of AIC246 was evaluated in vitro and in vivo using various cell culture assays and an engineered mouse xenograft model. In addition, antiviral properties of the drug were characterized in comparison to the current gold standard ganciclovir. We demonstrate that AIC246 exhibits excellent in vitro inhibitory activity against HCMV laboratory strains and clinical isolates, retains activity against ganciclovir-resistant viruses, is well tolerated in different cell types (median selectivity index, 18,000), and exerts a potent in vivo efficacy in a mouse xenograft model. Moreover, we show that the antiviral block induced by AIC246 is reversible and the efficacy of the drug is not significantly affected by cell culture variations such as cell type or multiplicity of infection. Finally, initial mode-of-action analyses reveal that AIC246 targets a process in the viral replication cycle that occurs later than DNA synthesis. Thus, AIC246 acts via a mode of action that differs from that of polymerase inhibitors like ganciclovir.
Collapse
|
39
|
Antiviral prevention of sepsis induced cytomegalovirus reactivation in immunocompetent mice. Antiviral Res 2009; 85:496-503. [PMID: 20004216 DOI: 10.1016/j.antiviral.2009.12.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2009] [Revised: 11/11/2009] [Accepted: 12/02/2009] [Indexed: 01/04/2023]
Abstract
INTRODUCTION Immunocompetent patients can reactivate latent cytomegalovirus (CMV) during critical illness and reactivation is associated with significantly worse outcomes. Prior to clinical trials in humans to prove causality, we sought to determine an optimal antiviral treatment strategy. METHODS Mice latently infected with murine CMV (MCMV) received a septic reactivation trigger and were randomized to receive one of four ganciclovir regimens or saline. Lungs were evaluated for viral transcriptional reactivation and fibrosis after each regimen. Influences of ganciclovir on early sepsis-induced pulmonary inflammation and T-cell activation were studied after sepsis induction. RESULTS All ganciclovir regimens reduced measurable MCMV transcriptional reactivation, and 10mg/day for 7 or 21 days was most effective. Lower dose (5mg/kg/day) or delayed therapy was associated with significant breakthrough reactivation. Higher doses of ganciclovir given early were associated with the lowest incidence of pulmonary fibrosis, and delay of therapy for 1 week was associated with significantly worse pulmonary fibrosis. Although bacterial sepsis induced activation of MCMV-specific pulmonary T-cells, this activation was not influenced by ganciclovir. CONCLUSION These results suggest that antiviral treatment trials in humans should use 10mg/kg/day ganciclovir administered as early as possible in at-risk patients to minimize reactivation events and associated pulmonary injury.
Collapse
|
40
|
Paulus C, Nevels M. The human cytomegalovirus major immediate-early proteins as antagonists of intrinsic and innate antiviral host responses. Viruses 2009; 1:760-79. [PMID: 21994568 PMCID: PMC3185523 DOI: 10.3390/v1030760] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2009] [Revised: 11/04/2009] [Accepted: 11/05/2009] [Indexed: 12/21/2022] Open
Abstract
The major immediate-early (IE) gene of human cytomegalovirus (CMV) is believed to have a decisive role in acute infection and its activity is an important indicator of viral reactivation from latency. Although a variety of gene products are expressed from this region, the 72-kDa IE1 and the 86-kDa IE2 nuclear phosphoproteins are the most abundant and important. Both proteins have long been recognized as promiscuous transcriptional regulators. More recently, a critical role of the IE1 and IE2 proteins in counteracting non-adaptive host cell defense mechanisms has been revealed. In this review we will briefly summarize the available literature on IE1- and IE2-dependent mechanisms contributing to CMV evasion from intrinsic and innate immune responses.
Collapse
Affiliation(s)
- Christina Paulus
- Institute for Medical Microbiology and Hygiene, University of Regensburg, Franz-Josef-Strauss-Allee 11, D-93053 Regensburg, Germany; E-Mail:
| | | |
Collapse
|
41
|
Giest S, Grace S, Senegaglia AC, Pasquini R, Gonzalo-Daganzo RM, Fernández MN, Mackinnon S, Madrigal JA, Travers PJ. Cytomegalovirus-specific CD8(+) T cells targeting different HLA/peptide combinations correlate with protection but at different threshold frequencies. Br J Haematol 2009; 148:311-322. [PMID: 19895611 DOI: 10.1111/j.1365-2141.2009.07969.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cytomegalovirus (CMV) causes significant morbidity and mortality in patients after haematopoietic stem cell transplantation (HSCT). Due to limitations of current antiviral therapies, alternative approaches, involving transfer of donor-derived CMV-specific CD8(+) T cells, have been considered. Levels of such cells correlating with protection against CMV infection and disease have only been reported in patients expressing HLA-A*0201 and HLA-B*0702. This is despite an increasing number of reports describing cells targeting CMV peptides presented by other human leucocyte antigens (HLAs). Considering several frequent HLA alleles, our findings suggest that HLA-A*2402/pp65 (341-349)- and HLA-B*3501/pp65 (123-131)-specific CD8(+) T cells correlate with protection from CMV reactivation at significantly lower cell levels than HLA-A*0101/pp50 (245-253)- and HLA-A*0201/pp65 (495-503)-specific CD8(+) T cells, both in HSCT recipients post-transplant and in healthy CMV seropositive volunteers. This may result from a differing efficiency of the responses restricted by the two sets of HLA alleles. These findings add to the knowledge of immunodominance and differences in antigen processing that are coordinated in individuals with different HLA alleles and have direct implications for therapy and monitoring in patients.
Collapse
|
42
|
Marschall M, Stamminger T. Molecular targets for antiviral therapy of cytomegalovirus infections. Future Microbiol 2009; 4:731-42. [PMID: 19659428 DOI: 10.2217/fmb.09.40] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Human cytomegalovirus infections are still associated with severe morbidity and mortality in immunocompromised individuals, despite the availability of five drugs that are currently licensed for antiviral therapy. Furthermore, human cytomegalovirus is the most frequent cause of congenital infections for which antiviral treatment options are very limited. Thus, the need for a potent, safe and well-tolerated antiviral drug remains. This review focuses on target molecules that are implicated in the development of innovative anticytomegaloviral approaches, such as viral immediate-early and DNA replication proteins, as well as regulatory protein kinases. Special emphasis is given to promising host factors, in particular the receptor tyrosine kinase PDGF and cyclin-dependent protein kinases, since a combined targeting of viral and cellular factors that are critical for viral replication may alleviate the emergence of drug-resistant virus variants.
Collapse
Affiliation(s)
- Manfred Marschall
- Institute for Clinical & Molecular Virology, University of Erlangen-Nuremberg, Schlossgarten 4, 91054 Erlangen, Germany.
| | | |
Collapse
|
43
|
Tchesnokov EP, Obikhod A, Schinazi RF, Götte M. Engineering of a chimeric RB69 DNA polymerase sensitive to drugs targeting the cytomegalovirus enzyme. J Biol Chem 2009; 284:26439-46. [PMID: 19622750 PMCID: PMC2785332 DOI: 10.1074/jbc.m109.012500] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2009] [Revised: 06/19/2009] [Indexed: 11/06/2022] Open
Abstract
Detailed structural and biochemical studies with the human cytomegalovirus (HCMV UL54) DNA polymerase are hampered by difficulties to obtain this enzyme in large quantities. The crystal structure of the related RB69 DNA polymerase (gp43) is often used as a model system to explain mechanisms of inhibition of DNA synthesis and drug resistance. However, here we demonstrate that gp43 is approximately 400-fold less sensitive to the pyrophosphate analog foscarnet, when compared with UL54. The RB69 enzyme is also able to discriminate against the nucleotide analog inhibitor acyclovir. In contrast, the HCMV polymerase is able to incorporate this compound with similar efficiency as observed with its natural counterpart. In an attempt to identify major determinants for drug activity, we replaced critical regions of the nucleotide-binding site of gp43 with equivalent regions of the HCMV enzyme. We show that chimeric gp43-UL54 enzymes that contain residues of helix N and helix P of UL54 are resensitized against foscarnet and acyclovir. Changing a region of three amino acids of helix N showed the strongest effects, and changes of two segments of three amino acids in helix P further contributed to the reversal of the phenotype. The engineered chimeric enzyme can be produced in large quantities and may therefore be a valuable surrogate system in drug development efforts. This system may likewise be used for detailed structural and biochemical studies on mechanisms associated with drug action and resistance.
Collapse
Affiliation(s)
- Egor P. Tchesnokov
- From the Department of Microbiology and Immunology, McGill University, Montreal, Quebec H3A 2B4, Canada and
| | - Aleksandr Obikhod
- the Center for AIDS Research, Department of Pediatrics, Emory University School of Medicine and Veterans Affairs Medical Research, Atlanta, Georgia 30322
| | - Raymond F. Schinazi
- the Center for AIDS Research, Department of Pediatrics, Emory University School of Medicine and Veterans Affairs Medical Research, Atlanta, Georgia 30322
| | - Matthias Götte
- From the Department of Microbiology and Immunology, McGill University, Montreal, Quebec H3A 2B4, Canada and
| |
Collapse
|
44
|
Prichard MN. Function of human cytomegalovirus UL97 kinase in viral infection and its inhibition by maribavir. Rev Med Virol 2009; 19:215-29. [PMID: 19434630 DOI: 10.1002/rmv.615] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The serine/threonine kinase expressed by human cytomegalovirus from gene UL97 phosphorylates the antiviral drug ganciclovir, but its biological function is the phosphorylation of its natural viral and cellular protein substrates which affect viral replication at many levels. The UL97 kinase null phenotype is therefore complex, as is the mechanism of action of maribavir, a highly specific inhibitor of its enzymatic activity. Studies that utilise the drug corroborate results from genetic approaches and together have elucidated many functions of the UL97 kinase that are critical for viral replication. The kinase phosphorylates eukaryotic elongation factor 1delta, the carboxyl terminal domain of the large subunit of RNA polymerase II, the retinoblastoma tumour suppressor and lamins A and C. Each of these is also phosphorylated and regulated by cdc2/cyclin-dependent kinase 1, suggesting that the viral kinase may perform a similar function. These and other activities of the UL97 kinase appear to stimulate the cell cycle to support viral DNA synthesis, enhance the expression of viral genes, promote virion morphogenesis and facilitate the egress of mature capsids from the nucleus. In the absence of UL97 kinase activity, viral DNA synthesis is inefficient and structural proteins are sequestered in nuclear aggresomes, reducing the efficiency of virion morphogenesis. Mature capsids that do form fail to egress the nucleus as the nuclear lamina are not dispersed by the kinase. The critical functions performed by the UL97 kinase illustrate its importance in viral replication and confirm that the kinase is a target for the development of antiviral therapies.
Collapse
Affiliation(s)
- Mark N Prichard
- Department of Pediatrics, University of Alabama School of Medicine, Birmingham, Alabama 35233, USA.
| |
Collapse
|
45
|
Inhibition of human cytomegalovirus replication via peptide aptamers directed against the nonconventional nuclear localization signal of the essential viral replication factor pUL84. J Virol 2009; 83:11902-13. [PMID: 19740994 DOI: 10.1128/jvi.01378-09] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The UL84 open reading frame of human cytomegalovirus encodes an essential multifunctional regulatory protein that is thought to act in the nucleus as an initiator of lytic viral replication. Nuclear trafficking of pUL84 is facilitated by a complex nonconventional nuclear localization signal (NLS) that mediates its interaction with the cellular importin-alpha/beta pathway. Since binding of pUL84 to importin-alpha proteins mechanistically differs from that of cellular proteins containing a classical NLS, we assumed that specific interference with the nuclear import of pUL84 might be possible and that this could constitute a novel principle for antiviral therapy. In order to test this hypothesis, we employed peptide aptamer technology and isolated several peptide aptamers from a randomized peptide expression library that specifically bind with high affinity to the unconventional pUL84 NLS under intracellular conditions. Coimmunoprecipitation experiments confirmed these interactions in mammalian cells, and the antiviral potential of the identified peptide aptamers was determined using three independent experimental approaches. (i) Infection experiments with a recombinant human cytomegalovirus expressing green fluorescent protein demonstrated 50 to 60% decreased viral replication in primary human fibroblasts stably expressing pUL84-specific aptamers. (ii) A 50 to 70% reduction of viral plaque formation, as well as a 70 to 90% inhibition of virus release in the presence of pUL84-specific aptamers, was observed. (iii) Immunofluorescence analyses revealed a shift from an almost exclusively nuclear pUL84 staining pattern to a nucleocytoplasmic distribution upon coexpression of the identified molecules, indicating that interference with the nuclear import of pUL84 contributes to the observed antiviral activity of the identified pUL84-binding aptamer molecules.
Collapse
|
46
|
Tuthill M, Chen F, Paston S, De La Peña H, Rusakiewicz S, Madrigal A. The prevention and treatment of cytomegalovirus infection in haematopoietic stem cell transplantation. Cancer Immunol Immunother 2009; 58:1481-8. [PMID: 19466407 PMCID: PMC11030108 DOI: 10.1007/s00262-009-0722-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2009] [Accepted: 05/04/2009] [Indexed: 12/01/2022]
Abstract
Allogeneic haematopoietic stem cell transplantation (HSCT) is an intensive medical treatment involving myeloablative chemo-radiotherapy followed by stem cell rescue using allogeneic haematopoietic stem cells harvested from HLA-matched donors, which is primarily used for the treatment of haematological malignancies. Cytomegalovirus (CMV) infection is one of the major causes of morbidity and death after HSCT. This focused research review highlights the advances made with research into CMV in the HSCT setting. It provides the reader with an overview of current CMV research into the prevention and management of CMV infection.
Collapse
Affiliation(s)
- Mark Tuthill
- The Anthony Nolan Research Institute, Royal Free Hospital, London, UK.
| | | | | | | | | | | |
Collapse
|
47
|
Wait S, Musingarimi P, Briggs A, Tillotson G. Assessing the economic merits of managing cytomegalovirus infection in organ and stem cell transplantation. J Med Econ 2009; 12:68-76. [PMID: 19450067 DOI: 10.3111/13696990902855019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Two preventative approaches exist to manage cytomegalovirus (CMV), a common infection in recipients of organ and stem cell transplants: prophylaxis--the prevention of viraemia--and pre-emptive therapy--the prevention of manifestation of disease in patients who have viraemia. Economic evaluation may provide a helpful framework to inform the choice between these two approaches. However, several issues arise. Direct comparisons of prophylaxis and pre-emptive therapy are rare and there are few epidemiological data that depict the full natural history of CMV infection and disease. There is a need for large, prospective randomised trials that directly compare these two strategies and are of sufficient duration to assess their overall impact on direct and indirect effects of CMV as well as patient quality of life. These methodological issues are relevant to the economic evaluation of preventative measures in other clinical settings and highlight the need for a rigorous evaluative framework to best inform decision making about the optimal strategy for patients.
Collapse
|
48
|
Yin MT, Brust JCM, Tieu HV, Hammer SM. Antiherpesvirus, Anti?Hepatitis Virus, and Anti?Respiratory Virus Agents. CLINICAL VIROLOGY 2009:217-264. [DOI: 10.1128/9781555815981.ch12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
49
|
Calistri A, Salata C, Parolin C, Palù G. Role of multivesicular bodies and their components in the egress of enveloped RNA viruses. Rev Med Virol 2009; 19:31-45. [PMID: 18618839 DOI: 10.1002/rmv.588] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
As an enveloped virus buds, the nascent viral capsid becomes wrapped in a plasma membrane-derived lipid envelope, and a membrane fission event is thus necessary to separate the virion from the host cell. This membrane fission event is well characterised in the case of enveloped RNA viruses, where it is promoted by late assembly domains (L-domains) present at the level of specific viral structural proteins. Research conducted over the past 10 years has demonstrated that L-domains represent docking sites for cellular proteins essential for the biogenesis of a cellular organelle, the multivesicular body (MVB). In this way, enveloped RNA viruses hijack the MVB components to the cellular site where the budding is executed. This review will focus on the cellular machinery exploited by enveloped RNA viruses in order to be released from infected cells. The role of ubiquitin and lipids in viral budding will also be discussed.
Collapse
Affiliation(s)
- A Calistri
- Department of Histology, Microbiology and Medical Biotechnologies, University of Padova, Padova, Italy
| | | | | | | |
Collapse
|
50
|
Shen W, Kim JS, Mitchell S, Kish P, Kijek P, Hilfinger J. 5'-O-D-valyl ara A, a potential prodrug for improving oral bioavailability of the antiviral agent vidarabine. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2009; 28:43-55. [PMID: 19116869 DOI: 10.1080/15257770802581757] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
In order to improve the oral bioavailability of Adenine 9-beta-D-arabinofuranoside (Vidarabine, also called ara A), an antiviral drug which is active against herpes simplex and varicella zoster viruses and the first agent to be licensed for the treatment of systematic herpes virus infection in man, the corresponding 5'-O-D-valyl ester derivative has been synthesized. Based on their physicochemical properties, 5'-O-valyl ara A has emerged as a potential prodrug candidate to improve the oral bioavailability of vidarabine. We describe in this paper a facile synthesis route for the prodrug and its physicochemical properties.
Collapse
Affiliation(s)
- Wei Shen
- TSRL Inc., Ann Arbor, Michigan 48108, USA.
| | | | | | | | | | | |
Collapse
|