201
|
Jones RP. Roles for Cytomegalovirus in Infection, Inflammation, and Autoimmunity. INFECTION AND AUTOIMMUNITY 2015:319-357. [DOI: 10.1016/b978-0-444-63269-2.00068-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
202
|
Kharfan-Dabaja MA, Nishihori T. Vaccine therapy for cytomegalovirus in the setting of allogeneic hematopoietic cell transplantation. Expert Rev Vaccines 2014; 14:341-50. [PMID: 25468066 DOI: 10.1586/14760584.2015.989990] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Passive immunization against CMV is desirable to minimize or perhaps eliminate complications related to CMV disease. In allogeneic hematopoietic cell transplantation (allo-HCT), the major challenge facing a successful anti-CMV vaccine is inducing immunity in an immunocompromised host. To date, only one CMV vaccine, ASP0113, has been evaluated in a randomized, placebo-controlled Phase II study. ASP0113 is a bivalent product containing two plasmids that encode CMV glycoprotein B and tegument phosphoprotein 65, respectively. Although there was no significant difference in rate of initiation of anti-CMV therapy, rates of CMV viremia were lower in the ASP0113 group when measured by a central laboratory. Also, time-to-first episode of viremia was longer in subjects receiving ASP0113. These findings paved the way for an ongoing placebo-controlled Phase III study aiming at enrolling 500 subjects. Results of this Phase III trial, especially if it meets clinically meaningful endpoints, will ultimately determine the role of anti-CMV vaccine strategies in allo-HCT.
Collapse
Affiliation(s)
- Mohamed A Kharfan-Dabaja
- Department of Blood and Marrow Transplantation, H. Lee Moffitt Cancer Center, 12902 Magnolia Drive, FOB-3, Tampa, FL, USA
| | | |
Collapse
|
203
|
Landi D, Hegde M, Ahmed N. Human cytomegalovirus antigens in malignant gliomas as targets for adoptive cellular therapy. Front Oncol 2014; 4:338. [PMID: 25505736 PMCID: PMC4244608 DOI: 10.3389/fonc.2014.00338] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 11/10/2014] [Indexed: 12/13/2022] Open
Abstract
Malignant gliomas are the most common primary brain tumor in adults, with over 12,000 new cases diagnosed in the United States each year. Over the last decade, investigators have reliably identified human cytomegalovirus (HCMV) proteins, nucleic acids, and virions in most high-grade gliomas, including glioblastoma (GBM). This discovery is significant because HCMV gene products can be targeted by immune-based therapies. In this review, we describe the current level of understanding regarding the presence and role in pathogenesis of HCMV in GBM. We describe our success detecting and expanding HCMV-specific cytotoxic T lymphocytes to kill GBM cells and explain how these cells can be used as a platform for enhanced cellular therapies. We discuss alternative approaches that capitalize on HCMV infection to treat patients with HCMV-positive tumors. Adoptive cellular therapy for HCMV-positive GBM has been tried in a small number of patients with some benefit, but we reason why, to date, these approaches generally fail to generate long-term remission or cure. We conjecture how cellular therapy for GBM can be improved and describe the barriers that must be overcome to cure these patients.
Collapse
Affiliation(s)
- Daniel Landi
- Center for Cell and Gene Therapy, Baylor College of Medicine , Houston, TX , USA ; Hematology and Oncology, Texas Children's Cancer Center , Houston, TX , USA
| | - Meenakshi Hegde
- Center for Cell and Gene Therapy, Baylor College of Medicine , Houston, TX , USA ; Hematology and Oncology, Texas Children's Cancer Center , Houston, TX , USA
| | - Nabil Ahmed
- Center for Cell and Gene Therapy, Baylor College of Medicine , Houston, TX , USA ; Hematology and Oncology, Texas Children's Cancer Center , Houston, TX , USA ; Houston Methodist Hospital , Houston, TX , USA
| |
Collapse
|
204
|
Leroy S, M'Zali F, Kann M, Weber DJ, Smith DD. Impact of vaginal-rectal ultrasound examinations with covered and low-level disinfected transducers on infectious transmissions in france. Infect Control Hosp Epidemiol 2014; 35:1497-504. [PMID: 25419772 DOI: 10.1086/678604] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND The risk of cross-infection from shared ultrasound probes in endorectal and vaginal ultrasonography due to low-level disinfection (LLD) is difficult to estimate because potential infections are also sexually transmitted diseases, and route of contamination is often difficult to establish. In France, the widely used standard for prevention of infections is through the use of probe covers and LLD of the ultrasound transducer by disinfectant wipes. We performed an in silico simulation based on a systematic review to estimate the number of patients infected after endorectal or vaginal ultrasonography examination using LLD for probes. STUDY DESIGN We performed a stochastic Monte Carlo computer simulation to produce hypothetical cohorts for a population of 4 million annual ultrasound examinations performed in France, and we estimated the number of infected patients for human immunodeficiency virus (HIV), herpes simplex virus, hepatitis B virus, hepatitis C virus, human papilloma virus, cytomegalovirus, and Chlamydia trachomatis. Modeling parameters were estimated by meta-analysis when possible. RESULTS The probability of infection from a contaminated probe ranged from 1% to 6%, depending on the pathogen. For cases of HIV infection, this would result in approximately 60 infected patients per year. For other common viral infections, the number of new cases ranged from 1,600 to 15,000 per year that could be attributable directly to ultrasound and LLD procedures. CONCLUSIONS Our simulation results showed that, despite cumulative use of probe cover and LLD, there were still some cases of de novo infection that may be attributable to ultrasound procedures. These cases are preventable by reviewing the currently used LLD and/or upgrading LLD to high-level disinfection, as recommended by the US Centers for Disease Control and Prevention.
Collapse
Affiliation(s)
- Sandrine Leroy
- Laboratoire de Biostatistique, Epidémiologie, Santé Publique et Informatique Médicale, Centre Hospitalier Universitaire (CHU) de Nîmes, Nîmes, France; and EA 2415 Unit, Montpellier 1 University, Montpellier, France
| | | | | | | | | |
Collapse
|
205
|
|
206
|
Gardner TJ, Cohen T, Redmann V, Lau Z, Felsenfeld D, Tortorella D. Development of a high-content screen for the identification of inhibitors directed against the early steps of the cytomegalovirus infectious cycle. Antiviral Res 2014; 113:49-61. [PMID: 25446405 DOI: 10.1016/j.antiviral.2014.10.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 10/16/2014] [Accepted: 10/20/2014] [Indexed: 01/06/2023]
Abstract
Human cytomegalovirus (CMV) is a latent and persistent virus whose proliferation increases morbidity and mortality of immune-compromised individuals. The current anti-CMV therapeutics targeting the viral DNA polymerase or the major immediate-early (MIE) gene locus are somewhat effective at limiting CMV-associated disease. However, due to low bioavailability, severe toxicity, and the development of drug resistant CMV strains following prolonged treatment, current anti-CMV therapeutics are insufficient. To help address this shortfall, we established a high-content assay to identify inhibitors targeting CMV entry and the early steps of infection. The infection of primary human fibroblasts with a variant of the CMV laboratory strain AD169 expressing a chimeric IE2-yellow fluorescence protein (YFP) (AD169IE2-YFP) provided the basis for the high-content assay. The localization of IE2-YFP to the nucleus shortly following an AD169IE2-YFP infection induced a robust fluorescent signal that was quantified using confocal microscopy. The assay was optimized to achieve outstanding assay fitness and high Z' scores. We then screened a bioactive chemical library consisting of 2080 compounds and identified hit compounds based on the decrease of fluorescence signal from IE2-YFP nuclear expression. The hit compounds likely target various cellular processes involved in the early steps of infection including capsid transport, chromatin remodeling, and viral gene expression. Extensive secondary assays confirmed the ability of a hit compound, convallatoxin, to inhibit infection of both laboratory and clinical CMV strains and limit virus proliferation. Collectively, the data demonstrate that we have established a robust high-content screen to identify compounds that limit the early steps of the CMV life cycle, and that novel inhibitors of early infection events may serve as viable CMV therapeutics.
Collapse
Affiliation(s)
- Thomas J Gardner
- Icahn School of Medicine at Mount Sinai, Department of Microbiology, New York, NY 10029, USA
| | - Tobias Cohen
- Icahn School of Medicine at Mount Sinai, Department of Microbiology, New York, NY 10029, USA
| | - Veronika Redmann
- Icahn School of Medicine at Mount Sinai, Department of Microbiology, New York, NY 10029, USA
| | - Zerlina Lau
- Icahn School of Medicine at Mount Sinai, Integrated Screening Core, Experimental Therapeutics Institute, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Dan Felsenfeld
- Icahn School of Medicine at Mount Sinai, Integrated Screening Core, Experimental Therapeutics Institute, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Domenico Tortorella
- Icahn School of Medicine at Mount Sinai, Department of Microbiology, New York, NY 10029, USA
| |
Collapse
|
207
|
Drori A, Messerle M, Brune W, Tirosh B. Lack of XBP-1 impedes murine cytomegalovirus gene expression. PLoS One 2014; 9:e110942. [PMID: 25333725 PMCID: PMC4205010 DOI: 10.1371/journal.pone.0110942] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 09/26/2014] [Indexed: 11/17/2022] Open
Abstract
The unfolded protein response (UPR) is an endoplasmic reticulum (ER)-to-nucleus signaling cascade induced in response to ER stress. The UPR aims at restoring homeostasis, but can also induce apoptosis if stress persists. Infection by human and murine cytomegaloviruses (CMVs) provokes ER stress and induces the UPR. However, both CMVs manipulate the UPR to promote its prosurvival activity and delay apoptosis. The underlying mechanisms remain largely unknown. Recently, we demonstrated that MCMV and HCMV encode a late protein to target IRE1 for degradation. However, the importance of its downstream effector, X Box binding protein 1 (XBP-1), has not been directly studied. Here we show that deletion of XBP-1 prior to or early after infection confers a transient delay in viral propagation in fibroblasts that can be overcome by increasing the viral dose. A similar phenotype was demonstrated in peritoneal macrophages. In vivo, acute infection by MCMV is reduced in the absence of XBP-1. Our data indicate that removal of XBP-1 confers a kinetic delay in early stages of MCMV infection and suggest that the late targeting of IRE1 is aimed at inhibiting activities other than the splicing of XBP-1 mRNA.
Collapse
Affiliation(s)
- Adi Drori
- Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Martin Messerle
- Department of Virology, Hannover Medical School, Hannover, Germany
| | - Wolfram Brune
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Boaz Tirosh
- Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
208
|
Complex expression of the UL136 gene of human cytomegalovirus results in multiple protein isoforms with unique roles in replication. J Virol 2014; 88:14412-25. [PMID: 25297993 DOI: 10.1128/jvi.02711-14] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
UNLABELLED Human cytomegalovirus (HCMV) is a complex DNA virus with a 230-kb genome encoding 170 and up to 750 proteins. The upper limit of this coding capacity suggests the evolution of complex mechanisms to substantially increase the coding potential from the 230-kb genome. Our work examines the complexity of one gene, UL136, encoded within the ULb' region of the genome that is lost during serial passage of HCMV in cultured fibroblasts. UL136 is expressed as five protein isoforms. We mapped these isoforms and demonstrate that they originate from both a complex transcriptional profile and, possibly, the usage of multiple translation initiation sites. Intriguingly, the pUL136 isoforms exhibited distinct subcellular distributions with varying association with the Golgi apparatus. The subcellular localization of membrane-bound isoforms of UL136 differed between when they were expressed exogenously and when they were expressed in the context of viral infection, suggesting that the trafficking of these isoforms is mediated by infection-specific factors. While UL136, like most ULb' genes, was dispensable for replication in fibroblasts, the soluble 23- and 19-kDa isoforms suppressed virus replication. In CD34(+) hematopoietic progenitor cells (HPCs) infected in vitro, disruption of the 23- and 19-kDa isoforms resulted in increased replication and a loss of the latency phenotype, similar to the effects of the UL138 latency determinant encoded within the same genetic locus. Our work suggests a complex interplay between the UL136 isoforms which balances viral replication in multiple cell types and likely contributes to the cell type-dependent phenotypes of the UL133/8 locus and the outcome of HCMV infection. IMPORTANCE HCMV is a significant cause of morbidity in immunocompromised individuals, including transplant patients. The lifelong persistence of the virus results in a high seroprevalence worldwide and may contribute to age-related pathologies, such as atherosclerosis. The mechanisms of viral persistence are poorly understood; however, understanding the molecular basis of persistence is imperative for the development of new treatments. In this work, we characterize a complex HCMV gene, UL136, which is expressed as five protein isoforms. These isoforms arise predominantly from complex transcriptional mechanisms, which contribute to an increased coding capacity of the virus. Further, the UL136 isoforms oppose the activity of one another to balance HCMV replication in multiple cell types. We identify soluble isoforms of UL136 that function to suppress virus replication in fibroblasts and in CD34(+) HPCs for latency.
Collapse
|
209
|
Almehmadi M, Flanagan BF, Khan N, Alomar S, Christmas SE. Increased numbers and functional activity of CD56⁺ T cells in healthy cytomegalovirus positive subjects. Immunology 2014; 142:258-68. [PMID: 24433347 DOI: 10.1111/imm.12250] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 01/07/2014] [Accepted: 01/09/2014] [Indexed: 01/10/2023] Open
Abstract
Human T cells expressing CD56 are capable of tumour cell lysis following activation with interleukin-2 but their role in viral immunity has been less well studied. Proportions of CD56(+) T cells were found to be highly significantly increased in cytomegalovirus-seropositive (CMV(+) ) compared with seronegative (CMV(-) ) healthy subjects (9.1 ± 1.5% versus 3.7 ± 1.0%; P < 0.0001). Proportions of CD56(+) T cells expressing CD28, CD62L, CD127, CD161 and CCR7 were significantly lower in CMV(+) than CMV(-) subjects but those expressing CD4, CD8, CD45RO, CD57, CD58, CD94 and NKG2C were significantly increased (P < 0.05), some having the phenotype of T effector memory cells. Levels of pro-inflammatory cytokines and CD107a were significantly higher in CD56(+) T cells from CMV(+) than CMV(-) subjects following stimulation with CMV antigens. This also resulted in higher levels of proliferation in CD56(+) T cells from CMV(+) than CMV(-) subjects. Using Class I HLA pentamers, it was found that CD56(+) T cells from CMV(+) subjects contained similar proportions of antigen-specific CD8(+) T cells to CD56(-) T cells in donors of several different HLA types. These differences may reflect the expansion and enhanced functional activity of CMV-specific CD56(+) memory T cells. In view of the link between CD56 expression and T-cell cytotoxic function, this strongly implicates CD56(+) T cells as being an important component of the cytotoxic T-cell response to CMV in healthy carriers.
Collapse
Affiliation(s)
- Mazen Almehmadi
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection & Global Health, University of Liverpool, Liverpool, UK
| | | | | | | | | |
Collapse
|
210
|
Kleiboeker S, Nutt J, Schindel B, Dannehl J, Hester J. Cytomegalovirus antiviral resistance: characterization of results from clinical specimens. Transpl Infect Dis 2014; 16:561-7. [DOI: 10.1111/tid.12241] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 01/27/2014] [Accepted: 03/01/2014] [Indexed: 11/30/2022]
Affiliation(s)
| | - J. Nutt
- Viracor-IBT Laboratories; Lee's Summit Missouri USA
| | - B. Schindel
- Viracor-IBT Laboratories; Lee's Summit Missouri USA
| | - J. Dannehl
- Viracor-IBT Laboratories; Lee's Summit Missouri USA
| | - J. Hester
- Viracor-IBT Laboratories; Lee's Summit Missouri USA
| |
Collapse
|
211
|
Umashankar M, Rak M, Bughio F, Zagallo P, Caviness K, Goodrum FD. Antagonistic determinants controlling replicative and latent states of human cytomegalovirus infection. J Virol 2014; 88:5987-6002. [PMID: 24623432 PMCID: PMC4093889 DOI: 10.1128/jvi.03506-13] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 03/05/2014] [Indexed: 01/28/2023] Open
Abstract
UNLABELLED The mechanisms by which viruses persist and particularly those by which viruses actively contribute to their own latency have been elusive. Here we report the existence of opposing functions encoded by genes within a polycistronic locus of the human cytomegalovirus (HCMV) genome that regulate cell type-dependent viral fates: replication and latency. The locus, referred to as the UL133-UL138 (UL133/8) locus, encodes four proteins, pUL133, pUL135, pUL136, and pUL138. As part of the ULb' region of the genome, the UL133/8 locus is lost upon serial passage of clinical strains of HCMV in cultured fibroblasts and is therefore considered dispensable for replication in this context. Strikingly, we could not reconstitute infection in permissive fibroblasts from bacterial artificial chromosome clones of the HCMV genome where UL135 alone was disrupted. The loss of UL135 resulted in complex phenotypes and could ultimately be overcome by infection at high multiplicities. The requirement for UL135 but not the entire locus led us to hypothesize that another gene in this locus suppressed virus replication in the absence of UL135. The defect associated with the loss of UL135 was largely rescued by the additional disruption of the UL138 latency determinant, indicating a requirement for UL135 for virus replication when UL138 is expressed. In the CD34(+) hematopoietic progenitor model of latency, viruses lacking only UL135 were defective for viral genome amplification and reactivation. Taken together, these data indicate that UL135 and UL138 comprise a molecular switch whereby UL135 is required to overcome UL138-mediated suppression of virus replication to balance states of latency and reactivation. IMPORTANCE Mechanisms by which viruses persist in their host remain one of the most poorly understood phenomena in virology. Herpesviruses, including HCMV, persist in an incurable, latent state that has profound implications for immunocompromised individuals, including transplant patients. Further, the latent coexistence of HCMV may increase the risk of age-related pathologies, including vascular disease. The key to controlling or eradicating HCMV lies in understanding the molecular basis for latency. In this work, we describe the complex interplay between two viral proteins, pUL135 and pUL138, which antagonize one another in infection to promote viral replication or latency, respectively. We previously described the role of pUL138 in suppressing virus replication for latency. Here we demonstrate a role of pUL135 in overcoming pUL138-mediated suppression for viral reactivation. From this work, we propose that pUL135 and pUL138 constitute a molecular switch balancing states of latency and reactivation.
Collapse
Affiliation(s)
| | - Michael Rak
- BIO5 Institute, University of Arizona, Tucson, Arizona, USA
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| | - Farah Bughio
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| | - Patricia Zagallo
- Department of Immunobiology, University of Arizona, Tucson, Arizona, USA
| | - Katie Caviness
- Graduate Interdisciplinary Program in Genetics, University of Arizona, Tucson, Arizona, USA
| | - Felicia D. Goodrum
- BIO5 Institute, University of Arizona, Tucson, Arizona, USA
- Department of Immunobiology, University of Arizona, Tucson, Arizona, USA
- Graduate Interdisciplinary Program in Genetics, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
212
|
Lowest numbers of primary CD8(+) T cells can reconstitute protective immunity upon adoptive immunotherapy. Blood 2014; 124:628-37. [PMID: 24855206 DOI: 10.1182/blood-2013-12-547349] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT) are threatened by potentially lethal viral manifestations like cytomegalovirus (CMV) reactivation. Because the success of today's virostatic treatment is limited by side effects and resistance development, adoptive transfer of virus-specific memory T cells derived from the stem cell donor has been proposed as an alternative therapeutic strategy. In this context, dose minimization of adoptively transferred T cells might be warranted for the avoidance of graft-versus-host disease (GVHD), in particular in prophylactic settings after T-cell-depleting allo-HSCT protocols. To establish a lower limit for successful adoptive T-cell therapy, we conducted low-dose CD8(+) T-cell transfers in the well-established murine Listeria monocytogenes (L.m.) infection model. Major histocompatibility complex-Streptamer-enriched antigen-specific CD62L(hi) but not CD62L(lo) CD8(+) memory T cells proliferated, differentiated, and protected against L.m. infections after prophylactic application. Even progenies derived from a single CD62L(hi) L.m.-specific CD8(+) T cell could be protective against bacterial challenge. In analogy, low-dose transfers of Streptamer-enriched human CMV-specific CD8(+) T cells into allo-HSCT recipients led to strong pathogen-specific T-cell expansion in a compassionate-use setting. In summary, low-dose adoptive T-cell transfer (ACT) could be a promising strategy, particularly for prophylactic treatment of infectious complications after allo-HSCT.
Collapse
|
213
|
Van Damme E, Van Loock M. Functional annotation of human cytomegalovirus gene products: an update. Front Microbiol 2014; 5:218. [PMID: 24904534 PMCID: PMC4032930 DOI: 10.3389/fmicb.2014.00218] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 04/25/2014] [Indexed: 01/31/2023] Open
Abstract
Human cytomegalovirus is an opportunistic double-stranded DNA virus with one of the largest viral genomes known. The 235 kB genome is divided in a unique long (UL) and a unique short (US) region which are flanked by terminal and internal repeats. The expression of HCMV genes is highly complex and involves the production of protein coding transcripts, polyadenylated long non-coding RNAs, polyadenylated anti-sense transcripts and a variety of non-polyadenylated RNAs such as microRNAs. Although the function of many of these transcripts is unknown, they are suggested to play a direct or regulatory role in the delicately orchestrated processes that ensure HCMV replication and life-long persistence. This review focuses on annotating the complete viral genome based on three sources of information. First, previous reviews were used as a template for the functional keywords to ensure continuity; second, the Uniprot database was used to further enrich the functional database; and finally, the literature was manually curated for novel functions of HCMV gene products. Novel discoveries were discussed in light of the viral life cycle. This functional annotation highlights still poorly understood regions of the genome but more importantly it can give insight in functional clusters and/or may be helpful in the analysis of future transcriptomics and proteomics studies.
Collapse
Affiliation(s)
- Ellen Van Damme
- Janssen Infectious Diseases BVBA, Therapeutic Area of Infectious Diseases Beerse, Belgium
| | - Marnix Van Loock
- Janssen Infectious Diseases BVBA, Therapeutic Area of Infectious Diseases Beerse, Belgium
| |
Collapse
|
214
|
Sourvinos G, Morou A, Sanidas I, Codruta I, Ezell SA, Doxaki C, Kampranis SC, Kottakis F, Tsichlis PN. The downregulation of GFI1 by the EZH2-NDY1/KDM2B-JARID2 axis and by human cytomegalovirus (HCMV) associated factors allows the activation of the HCMV major IE promoter and the transition to productive infection. PLoS Pathog 2014; 10:e1004136. [PMID: 24830456 PMCID: PMC4022736 DOI: 10.1371/journal.ppat.1004136] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 04/07/2014] [Indexed: 12/26/2022] Open
Abstract
Earlier studies had suggested that epigenetic mechanisms play an important role in the control of human cytomegalovirus (HCMV) infection. Here we show that productive HCMV infection is indeed under the control of histone H3K27 trimethylation. The histone H3K27 methyltransferase EZH2, and its regulators JARID2 and NDY1/KDM2B repress GFI1, a transcriptional repressor of the major immediate-early promoter (MIEP) of HCMV. Knocking down EZH2, NDY1/KDM2B or JARID2 relieves the repression and results in the upregulation of GFI1. During infection, the incoming HCMV rapidly downregulates the GFI1 mRNA and protein in both wild-type cells and in cells in which EZH2, NDY1/KDM2B or JARID2 were knocked down. However, since the pre-infection levels of GFI1 in the latter cells are significantly higher, the virus fails to downregulate it to levels permissive for MIEP activation and viral infection. Following the EZH2-NDY1/KDM2B-JARID2-independent downregulation of GFI1 in the early stages of infection, the virus also initiates an EZH2-NDY1/ΚDM2Β-JARID2-dependent program that represses GFI1 throughout the infection cycle. The EZH2 knockdown also delays histone H3K27 trimethylation in the immediate early region of HCMV, which is accompanied by a drop in H3K4 trimethylation that may contribute to the shEZH2-mediated repression of the major immediate early HCMV promoter. These data show that HCMV uses multiple mechanisms to allow the activation of the HCMV MIEP and to prevent cellular mechanisms from blocking the HCMV replication program.
Collapse
Affiliation(s)
- George Sourvinos
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
- Laboratory of Virology, Medical School, University of Crete, Heraklion, Crete, Greece
- * E-mail: (GS); (PNT)
| | - Antigoni Morou
- Laboratory of Virology, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Ioannis Sanidas
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Ignea Codruta
- Laboratory of Biochemistry, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Scott A. Ezell
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Christina Doxaki
- Laboratory of Biochemistry, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Sotirios C. Kampranis
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
- Laboratory of Biochemistry, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Filippos Kottakis
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Philip N. Tsichlis
- Molecular Oncology Research Institute, Tufts Medical Center, Boston, Massachusetts, United States of America
- * E-mail: (GS); (PNT)
| |
Collapse
|
215
|
Ameres S, Besold K, Plachter B, Moosmann A. CD8 T cell-evasive functions of human cytomegalovirus display pervasive MHC allele specificity, complementarity, and cooperativity. THE JOURNAL OF IMMUNOLOGY 2014; 192:5894-905. [PMID: 24808364 DOI: 10.4049/jimmunol.1302281] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Immunoevasive proteins ("evasins") of human CMV (HCMV) modulate stability and localization of MHC class I (MHC I) molecules, and their supply of antigenic peptides. However, it is largely unknown to what extent these evasins interfere with recognition by virus-specific CD8 T cells. We analyzed the recognition of HCMV-infected cells by a panel of CD8 T cells restricted through one of nine different MHC I allotypes. We employed a set of HCMV mutants deleted for three or all four of the MHC I modulatory genes US2, US3, US6, and US11. We found that different HCMV evasins exhibited different allotype-specific patterns of interference with CD8 T cell recognition of infected cells. In contrast, recognition of different epitopes presented by the same given MHC I allotype was uniformly reduced. For some allotypes, single evasins largely abolished T cell recognition; for others, a concerted action of evasins was required to abrogate recognition. In infected cells whose Ag presentation efficiency had been enhanced by IFN-γ pretreatment, HCMV evasins cooperatively impared T cell recognition for several different MHC I allotypes. T cell recognition and MHC I surface expression under influence of evasins were only partially congruent, underscoring the necessity to probe HCMV immunomodulation using specific T cells. We conclude that the CD8 T cell evasins of HCMV display MHC I allotype specificity, complementarity, and cooperativity.
Collapse
Affiliation(s)
- Stefanie Ameres
- Klinische Kooperationsgruppe Immunonkologie, Medizinische Klinik III, Klinikum der Universität München, 81377 Munich, Germany; Abteilung Genvektoren, Helmholtz Zentrum München, 81377 Munich, Germany; German Center for Infection Research, 81675 Munich, Germany; and
| | - Katrin Besold
- Institut für Virologie, Universitätsmedizin der Johannes-Gutenberg-Universität Mainz, 55131 Mainz, Germany
| | - Bodo Plachter
- Institut für Virologie, Universitätsmedizin der Johannes-Gutenberg-Universität Mainz, 55131 Mainz, Germany
| | - Andreas Moosmann
- Klinische Kooperationsgruppe Immunonkologie, Medizinische Klinik III, Klinikum der Universität München, 81377 Munich, Germany; Abteilung Genvektoren, Helmholtz Zentrum München, 81377 Munich, Germany; German Center for Infection Research, 81675 Munich, Germany; and
| |
Collapse
|
216
|
Wang H, Yao Y, Huang C, Fu X, Chen Q, Zhang H, Chen J, Fang F, Xie Z, Chen Z. An adjuvanted inactivated murine cytomegalovirus (MCMV) vaccine induces potent and long-term protective immunity against a lethal challenge with virulent MCMV. BMC Infect Dis 2014; 14:195. [PMID: 24720840 PMCID: PMC4005462 DOI: 10.1186/1471-2334-14-195] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 04/04/2014] [Indexed: 11/18/2022] Open
Abstract
Background Human cytomegalovirus (HCMV) is a ubiquitous pathogen that causes serious problems in immunocompromised or immunologically immature hosts. Vaccination is the preferred approach for prevention of HCMV infection, but so far no approved HCMV vaccine is available. In this study, we assessed the immunogenicity and protective immunity of a formalin-inactivated murine cytomegalovirus vaccine (FI-MCMV) in a mouse model in combination with adjuvants MF59, alum, or chitosan. Methods Specific-pathogen-free BALB/c mice aged 6–8 weeks were immunized twice, 3 weeks apart, with various doses of FI-MCMV (0.25 μg, 1 μg, 4 μg) with or without adjuvant. Mice were challenged with a lethal dose (5 × LD50) of a more virulent mouse salivary gland-passaged MCMV 3 weeks after the second immunization. The protective immunity of the vaccine was evaluated by determining the survival rates, residual spleen and salivary gland viral loads, body weight changes, and serum anti-MCMV IgG titers. Results Immunization with FI-MCMV vaccine induced a high level of specific antibody response. Antigen sparing was achieved by the addition of an adjuvant, which significantly enhanced the humoral response to vaccine antigens with a wide range of doses. The level of live virus detected in the spleen on day 5 and in the salivary glands on day 21 after the lethal challenge was significantly lower in adjuvant-treated groups than in controls. Survival rates in adjuvant-treated groups also increased significantly. Furthermore, these protective immune responses were sustained for at least 6 months following immunization. Conclusions These results show that inactivated MCMV vaccine is effective, and that the adjuvanted FI-MCMV vaccine provides more effective and longer-term protection than the adjuvant-free vaccine.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Ze Chen
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, Hubei, China.
| |
Collapse
|
217
|
Vescovini R, Fagnoni FF, Telera AR, Bucci L, Pedrazzoni M, Magalini F, Stella A, Pasin F, Medici MC, Calderaro A, Volpi R, Monti D, Franceschi C, Nikolich-Žugich J, Sansoni P. Naïve and memory CD8 T cell pool homeostasis in advanced aging: impact of age and of antigen-specific responses to cytomegalovirus. AGE (DORDRECHT, NETHERLANDS) 2014; 36:625-40. [PMID: 24318918 PMCID: PMC4039262 DOI: 10.1007/s11357-013-9594-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 10/22/2013] [Indexed: 05/10/2023]
Abstract
Alterations in the circulating CD8+ T cell pool, with a loss of naïve and accumulation of effector/effector memory cells, are pronounced in older adults. However, homeostatic forces that dictate such changes remain incompletely understood. This observational cross-sectional study explored the basis for variability of CD8+ T cell number and composition of its main subsets: naïve, central memory and effector memory T cells, in 131 cytomegalovirus (CMV) seropositive subjects aged over 60 years. We found great heterogeneity of CD8+ T cell numbers, which was mainly due to variability of the CD8 + CD28- T cell subset regardless of age. Analysis, by multiple regression, of distinct factors revealed that age was a predictor for the loss in absolute number of naïve T cells, but was not associated with changes in central or effector memory CD8+ T cell subsets. By contrast, the size of CD8+ T cells specific to pp65 and IE-1 antigens of CMV, predicted CD28 - CD8+ T cell, antigen-experienced CD8+ T cell, and even total CD8+ T cell numbers, but not naïve CD8+ T cell loss. These results indicate a clear dichotomy between the homeostasis of naïve and antigen-experienced subsets of CD8+ T cells which are independently affected, in human later life, by age and antigen-specific responses to CMV, respectively.
Collapse
Affiliation(s)
- Rosanna Vescovini
- Department of Clinical and Experimental Medicine, University of Parma, via Gramsci 14, 43126, Parma, Italy,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
218
|
Abstract
Oral herpes virus infections (OHVIs) are among the most common mucosal disorders encountered by oral health care providers. These infections can affect individuals at any age, from infants to the elderly, and may cause significant pain and dysfunction. Immunosuppressed patients may be at increased risk for serious and potential life-threatening complications caused by OHVIs. Clinicians may have difficulty in diagnosing these infections because they can mimic other conditions of the oral mucosa. This article provides oral health care providers with clinically relevant information regarding etiopathogenesis, diagnosis, and management of OHVIs.
Collapse
Affiliation(s)
- Ramesh Balasubramaniam
- Orofacial Pain Clinic, School of Dentistry, University of Western Australia, 35 Stirling Highway, Crawley, Western Australia 6009, Australia; Perth Oral Medicine and Dental Sleep Centre, St John of God Hospital, Suite 311, 25 McCourt Street, Subiaco, Western Australia 6008, Australia
| | - Arthur S Kuperstein
- Department of Oral Medicine, University of Pennsylvania School of Dental Medicine, 240 South 40th Street, Room 207, Philadelphia, PA 19104, USA
| | - Eric T Stoopler
- Department of Oral Medicine, University of Pennsylvania School of Dental Medicine, 240 South 40th Street, Room 206, Philadelphia, PA 19104, USA.
| |
Collapse
|
219
|
Vandamme YM, Ducancelle A, Biere L, Viot N, Rouleau F, Delbos V, Abgueguen P. Myopericarditis complicated by pulmonary embolism in an immunocompetent patient with acute cytomegalovirus infection: a case report. BMC Res Notes 2014; 7:193. [PMID: 24678987 PMCID: PMC3999874 DOI: 10.1186/1756-0500-7-193] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 03/04/2014] [Indexed: 12/25/2022] Open
Abstract
Background Primary acute cytomegalovirus infection in immunocompetent patients is common worldwide. Infection is most often asymptomatic or occurs sub-clinically with a self-limited mononucleosis-like syndrome. More rarely, the infection may lead to severe organ complications with pneumonia, myocarditis, pericarditis, colitis and hemolytic anemia. Recent cases of cytomegalovirus-associated thrombosis have also been reported sporadically in the medical literature. Case presentation We report here a case of simultaneous myopericarditis and pulmonary embolism in a 30-year-old man with no medical history. The patient was not immunocompromised. We discuss the possible role of acute cytomegalovirus infection in the induction of vascular damage and review relevant cases in the literature. Conclusion Thrombosis in patients with acute cytomegalovirus infection may be more frequent than is generally thought. Physicians need to be aware of the possible association between acute cytomegalovirus and thrombosis in immunocompetent patients, especially in the presence of severe systemic infection, as our case illustrates.
Collapse
Affiliation(s)
- Yves Marie Vandamme
- Department of Infectious Diseases and Internal Medicine, Centre Hospitalier Universitaire d'Angers, 4 rue Larrey, 49933 Angers, Cedex 9, France.
| | | | | | | | | | | | | |
Collapse
|
220
|
Sijmons S, Van Ranst M, Maes P. Genomic and functional characteristics of human cytomegalovirus revealed by next-generation sequencing. Viruses 2014; 6:1049-72. [PMID: 24603756 PMCID: PMC3970138 DOI: 10.3390/v6031049] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 02/11/2014] [Accepted: 02/11/2014] [Indexed: 01/08/2023] Open
Abstract
The complete genome of human cytomegalovirus (HCMV) was elucidated almost 25 years ago using a traditional cloning and Sanger sequencing approach. Analysis of the genetic content of additional laboratory and clinical isolates has lead to a better, albeit still incomplete, definition of the coding potential and diversity of wild-type HCMV strains. The introduction of a new generation of massively parallel sequencing technologies, collectively called next-generation sequencing, has profoundly increased the throughput and resolution of the genomics field. These increased possibilities are already leading to a better understanding of the circulating diversity of HCMV clinical isolates. The higher resolution of next-generation sequencing provides new opportunities in the study of intrahost viral population structures. Furthermore, deep sequencing enables novel diagnostic applications for sensitive drug resistance mutation detection. RNA-seq applications have changed the picture of the HCMV transcriptome, which resulted in proof of a vast amount of splicing events and alternative transcripts. This review discusses the application of next-generation sequencing technologies, which has provided a clearer picture of the intricate nature of the HCMV genome. The continuing development and application of novel sequencing technologies will further augment our understanding of this ubiquitous, but elusive, herpesvirus.
Collapse
Affiliation(s)
- Steven Sijmons
- Laboratory of Clinical Virology, Rega Institute for Medical Research, K.U.Leuven, Minderbroedersstraat 10, Leuven BE-3000, Belgium.
| | - Marc Van Ranst
- Laboratory of Clinical Virology, Rega Institute for Medical Research, K.U.Leuven, Minderbroedersstraat 10, Leuven BE-3000, Belgium.
| | - Piet Maes
- Laboratory of Clinical Virology, Rega Institute for Medical Research, K.U.Leuven, Minderbroedersstraat 10, Leuven BE-3000, Belgium.
| |
Collapse
|
221
|
Watkins RR, Lemonovich TL, Razonable RR. Immune response to CMV in solid organ transplant recipients: current concepts and future directions. Expert Rev Clin Immunol 2014; 8:383-93. [DOI: 10.1586/eci.12.25] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
222
|
|
223
|
Ariza-Heredia EJ, Nesher L, Chemaly RF. Cytomegalovirus diseases after hematopoietic stem cell transplantation: a mini-review. Cancer Lett 2014; 342:1-8. [PMID: 24041869 DOI: 10.1016/j.canlet.2013.09.004] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 09/08/2013] [Indexed: 02/03/2023]
Abstract
Cytomegalovirus (CMV) infection remains a significant complication after hematopoietic stem cell transplantation (HSCT) and may have a deleterious impact on the overall outcome after transplantation. In addition to the direct effects of CMV infection, tissue-invasive CMV diseases may be associated with increased risk of graft versus host disease, myelosuppression, and invasive bacterial and fungal infections. Because of these direct and indirect adverse effects, prevention of CMV infection, mostly through pre-emptive therapy, is one of the essential strategies that may improve outcomes of HSCT recipients. Management of CMV infection relies mainly on intravenous (IV) antiviral therapy with ganciclovir and foscarnet, with or without IV polyclonal immunoglobulins. Although viral resistance remains rare, better tolerated antiviral agents with less serious side effects are needed, and a few will be evaluated in phase III clinical trials in the near future.
Collapse
Affiliation(s)
- Ella J Ariza-Heredia
- Department of Infectious Diseases, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | |
Collapse
|
224
|
Jakovljevic A, Andric M. Human Cytomegalovirus and Epstein-Barr Virus in Etiopathogenesis of Apical Periodontitis: A Systematic Review. J Endod 2014; 40:6-15. [DOI: 10.1016/j.joen.2013.10.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 09/29/2013] [Accepted: 10/01/2013] [Indexed: 02/06/2023]
|
225
|
|
226
|
Abstract
Human cytomegalovirus (HCMV) is a human pathogen that infects greater than 50 % of the human population. HCMV infection is usually asymptomatic in most individuals. That is, primary infection or reactivation of latent virus is generally clinically silent. HCMV infection, however, is associated with significant morbidity and mortality in the immunocompromised and chronic inflammatory diseases in the immunocompetent. In immunocompromised individuals (acquired immune deficiency syndrome and transplant patients, developing children (in utero), and cancer patients undergoing chemotherapy), HCMV infection increases morbidity and mortality. In those individuals with a normal immune system, HCMV infection is also associated with a risk of serious disease, as viral infection is now considered to be a strong risk factor for the development of various vascular diseases and to be associated with some types of tumor development. Intense research is currently being undertaken to better understand the mechanisms of viral pathogenesis that are briefly discussed in this chapter.
Collapse
Affiliation(s)
- Maciej T Nogalski
- Department of Microbiology & Immunology, Center for Molecular and Tumor Virology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | | | | |
Collapse
|
227
|
Schleiss MR. Developing a Vaccine against Congenital Cytomegalovirus (CMV) Infection: What Have We Learned from Animal Models? Where Should We Go Next? Future Virol 2013; 8:1161-1182. [PMID: 24523827 DOI: 10.2217/fvl.13.106] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Congenital human cytomegalovirus (HCMV) infection can lead to long-term neurodevelopmental sequelae, including mental retardation and sensorineural hearing loss. Unfortunately, CMVs are highly adapted to their specific species, precluding the evaluation of HCMV vaccines in animal models prior to clinical trials. Several species-specific CMVs have been characterized and developed in models of pathogenesis and vaccine-mediated protection against disease. These include the murine CMV (MCMV), the porcine CMV (PCMV), the rhesus macaque CMV (RhCMV), the rat CMV (RCMV), and the guinea pig CMV (GPCMV). Because of the propensity of the GPCMV to cross the placenta, infecting the fetus in utero, it has emerged as a model of particular interest in studying vaccine-mediated protection of the fetus. In this paper, a review of these various models, with particular emphasis on the value of the model in the testing and evaluation of vaccines against congenital CMV, is provided. Recent exciting developments and advances in these various models are summarized, and recommendations offered for high-priority areas for future study.
Collapse
Affiliation(s)
- Mark R Schleiss
- University of Minnesota Medical School Center for Infectious Diseases and Microbiology Translational Research Department of Pediatrics Division of Pediatric Infectious Diseases and Immunology 2001 6 Street SE Minneapolis, MN 55455-3007
| |
Collapse
|
228
|
Kekre N, Tokessy M, Mallick R, McDiarmid S, Huebsch L, Bredeson C, Allan D, Tay J, Tinmouth A, Sheppard D. Is Cytomegalovirus Testing of Blood Products Still Needed for Hematopoietic Stem Cell Transplant Recipients in the Era of Universal Leukoreduction? Biol Blood Marrow Transplant 2013; 19:1719-24. [DOI: 10.1016/j.bbmt.2013.09.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2013] [Accepted: 09/17/2013] [Indexed: 10/26/2022]
|
229
|
Hakki M, Goldman DC, Streblow DN, Hamlin KL, Krekylwich CN, Fleming WH, Nelson JA. HCMV infection of humanized mice after transplantation of G-CSF-mobilized peripheral blood stem cells from HCMV-seropositive donors. Biol Blood Marrow Transplant 2013; 20:132-5. [PMID: 24161922 DOI: 10.1016/j.bbmt.2013.10.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 10/07/2013] [Indexed: 01/21/2023]
Abstract
Human cytomegalovirus (HCMV) infection, including primary infection resulting from transmission from a seropositive donor to a seronegative recipient (D(+)/R(-)), remains a significant problem in the setting of peripheral blood stem cell transplantation (PBSCT). The lack of a suitable animal model for studying HCMV transmission after PBSCT is a major barrier to understanding this process and, consequently, developing novel interventions to prevent HCMV infection. Our previous work demonstrated that human CD34(+) progenitor cell-engrafted NOD-scid IL2Rγc(null) (NSG) mice support latent HCMV infection after direct inoculation and reactivation after treatment with granulocyte colony-stimulating factor. To more accurately recapitulate HCMV infection in the D(+)/R(-) PBSCT setting, granulocyte colony-stimulating factor-mobilized peripheral blood stem cells from seropositive donors were used to engraft NSG mice. All recipient mice demonstrated evidence of HCMV infection in liver, spleen, and bone marrow. These findings validate the NSG mouse model for studying HCMV transmission during PBSCT.
Collapse
Affiliation(s)
- Morgan Hakki
- Division of Infectious Diseases, Oregon Health and Science University, Portland, Oregon
| | - Devorah C Goldman
- Oregon Stem Cell Center, Department of Pediatrics, Oregon Health and Science University, Portland, Oregon
| | - Daniel N Streblow
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon
| | - Kimberly L Hamlin
- Oregon Stem Cell Center, Department of Pediatrics, Oregon Health and Science University, Portland, Oregon
| | - Craig N Krekylwich
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon
| | - William H Fleming
- Oregon Stem Cell Center, Department of Pediatrics, Oregon Health and Science University, Portland, Oregon
| | - Jay A Nelson
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon.
| |
Collapse
|
230
|
Ramanan P, Razonable RR. Cytomegalovirus infections in solid organ transplantation: a review. Infect Chemother 2013; 45:260-71. [PMID: 24396627 PMCID: PMC3848521 DOI: 10.3947/ic.2013.45.3.260] [Citation(s) in RCA: 231] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Indexed: 12/12/2022] Open
Abstract
Cytomegalovirus (CMV) continues to have a tremendous impact in solid organ transplantation despite remarkable advances in its diagnosis, prevention and treatment. It can affect allograft function and increase patient morbidity and mortality through a number of direct and indirect effects. Patients may develop asymptomatic viremia, CMV syndrome or tissue-invasive disease. Late-onset CMV disease continues to be a major problem in high-risk patients after completion of antiviral prophylaxis. Emerging data suggests that immunologic monitoring may be useful in predicting the risk of late onset CMV disease. There is now increasing interest in the development of an effective vaccine for prevention. Novel antiviral drugs with unique mechanisms of action and lesser toxicity are being developed. Viral load quantification is now undergoing standardization, and this will permit the generation of clinically relevant viral thresholds for the management of patients. This article provides a brief overview of the contemporary epidemiology, clinical presentation, diagnosis, prevention and treatment of CMV infection in solid organ transplant recipients.
Collapse
Affiliation(s)
- Poornima Ramanan
- Division of Infectious Diseases, Department of Medicine and the William J von Liebig Transplant Center, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Raymund R Razonable
- Division of Infectious Diseases, Department of Medicine and the William J von Liebig Transplant Center, Mayo Clinic, Rochester, Minnesota 55905, USA
| |
Collapse
|
231
|
Juranic Lisnic V, Babic Cac M, Lisnic B, Trsan T, Mefferd A, Das Mukhopadhyay C, Cook CH, Jonjic S, Trgovcich J. Dual analysis of the murine cytomegalovirus and host cell transcriptomes reveal new aspects of the virus-host cell interface. PLoS Pathog 2013; 9:e1003611. [PMID: 24086132 PMCID: PMC3784481 DOI: 10.1371/journal.ppat.1003611] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 07/26/2013] [Indexed: 11/19/2022] Open
Abstract
Major gaps in our knowledge of pathogen genes and how these gene products interact with host gene products to cause disease represent a major obstacle to progress in vaccine and antiviral drug development for the herpesviruses. To begin to bridge these gaps, we conducted a dual analysis of Murine Cytomegalovirus (MCMV) and host cell transcriptomes during lytic infection. We analyzed the MCMV transcriptome during lytic infection using both classical cDNA cloning and sequencing of viral transcripts and next generation sequencing of transcripts (RNA-Seq). We also investigated the host transcriptome using RNA-Seq combined with differential gene expression analysis, biological pathway analysis, and gene ontology analysis. We identify numerous novel spliced and unspliced transcripts of MCMV. Unexpectedly, the most abundantly transcribed viral genes are of unknown function. We found that the most abundant viral transcript, recently identified as a noncoding RNA regulating cellular microRNAs, also codes for a novel protein. To our knowledge, this is the first viral transcript that functions both as a noncoding RNA and an mRNA. We also report that lytic infection elicits a profound cellular response in fibroblasts. Highly upregulated and induced host genes included those involved in inflammation and immunity, but also many unexpected transcription factors and host genes related to development and differentiation. Many top downregulated and repressed genes are associated with functions whose roles in infection are obscure, including host long intergenic noncoding RNAs, antisense RNAs or small nucleolar RNAs. Correspondingly, many differentially expressed genes cluster in biological pathways that may shed new light on cytomegalovirus pathogenesis. Together, these findings provide new insights into the molecular warfare at the virus-host interface and suggest new areas of research to advance the understanding and treatment of cytomegalovirus-associated diseases.
Collapse
Affiliation(s)
- Vanda Juranic Lisnic
- Department of Histology and Embryology and the Center for Proteomics, University of Rijeka School of Medicine, Rijeka, Croatia
| | - Marina Babic Cac
- Department of Histology and Embryology and the Center for Proteomics, University of Rijeka School of Medicine, Rijeka, Croatia
| | - Berislav Lisnic
- Laboratory of Biology and Microbial Genetics, Faculty of Food Technology and Biotechnology, University of Zagreb, Zagreb, Croatia
| | - Tihana Trsan
- Department of Histology and Embryology and the Center for Proteomics, University of Rijeka School of Medicine, Rijeka, Croatia
| | - Adam Mefferd
- The Department of Surgery, The Ohio State University, Columbus, Ohio, United States of America
| | | | - Charles H. Cook
- The Department of Surgery, The Ohio State University, Columbus, Ohio, United States of America
| | - Stipan Jonjic
- Department of Histology and Embryology and the Center for Proteomics, University of Rijeka School of Medicine, Rijeka, Croatia
| | - Joanne Trgovcich
- The Department of Surgery, The Ohio State University, Columbus, Ohio, United States of America
| |
Collapse
|
232
|
Smith LR, Wloch MK, Chaplin JA, Gerber M, Rolland AP. Clinical Development of a Cytomegalovirus DNA Vaccine: From Product Concept to Pivotal Phase 3 Trial. Vaccines (Basel) 2013; 1:398-414. [PMID: 26344340 PMCID: PMC4494211 DOI: 10.3390/vaccines1040398] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 08/23/2013] [Accepted: 08/28/2013] [Indexed: 12/31/2022] Open
Abstract
2013 marks a milestone year for plasmid DNA vaccine development as a first-in-class cytomegalovirus (CMV) DNA vaccine enters pivotal phase 3 testing. This vaccine consists of two plasmids expressing CMV antigens glycoprotein B (gB) and phosphoprotein 65 (pp65) formulated with a CRL1005 poloxamer and benzalkonium chloride (BAK) delivery system designed to enhance plasmid expression. The vaccine’s planned initial indication under investigation is for prevention of CMV reactivation in CMV-seropositive (CMV+) recipients of an allogeneic hematopoietic stem cell transplant (HCT). A randomized, double-blind placebo-controlled phase 2 proof-of-concept study provided initial evidence of the safety of this product in CMV+ HCT recipients who underwent immune ablation conditioning regimens. This study revealed a significant reduction in viral load endpoints and increased frequencies of pp65-specific interferon-γ-producing T cells in vaccine recipients compared to placebo recipients. The results of this endpoint-defining trial provided the basis for defining the primary and secondary endpoints of a global phase 3 trial in HCT recipients. A case study is presented here describing the development history of this vaccine from product concept to initiation of the phase 3 trial.
Collapse
Affiliation(s)
- Larry R Smith
- Vical Incorporated, 10390 Pacific Center Court, San Diego, California, CA 92121, USA.
| | - Mary K Wloch
- Vical Incorporated, 10390 Pacific Center Court, San Diego, California, CA 92121, USA.
| | - Jennifer A Chaplin
- Vical Incorporated, 10390 Pacific Center Court, San Diego, California, CA 92121, USA.
| | - Michele Gerber
- Astellas Pharma Global Development, Inc., 1 Astellas Way, Northbrook, IL 60062, USA.
| | - Alain P Rolland
- Vical Incorporated, 10390 Pacific Center Court, San Diego, California, CA 92121, USA.
| |
Collapse
|
233
|
Jin N, Malcherek G, Mani J, Zurleit R, Schmitt A, Chen B, Freund M, Ho AD, Schmitt M. Suppression of cytomegalovirus-specific CD8+T cells by everolimus. Leuk Lymphoma 2013; 55:1144-50. [DOI: 10.3109/10428194.2013.822496] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
234
|
Detection of cytomegalovirus drug resistance mutations by next-generation sequencing. J Clin Microbiol 2013; 51:3700-10. [PMID: 23985916 DOI: 10.1128/jcm.01605-13] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Antiviral therapy for cytomegalovirus (CMV) plays an important role in the clinical management of solid organ and hematopoietic stem cell transplant recipients. However, CMV antiviral therapy can be complicated by drug resistance associated with mutations in the phosphotransferase UL97 and the DNA polymerase UL54. We have developed an amplicon-based high-throughput sequencing strategy for detecting CMV drug resistance mutations in clinical plasma specimens using a microfluidics PCR platform for multiplexed library preparation and a benchtop next-generation sequencing instrument. Plasmid clones of the UL97 and UL54 genes were used to demonstrate the low overall empirical error rate of the assay (0.189%) and to develop a statistical algorithm for identifying authentic low-abundance variants. The ability of the assay to detect resistance mutations was tested with mixes of wild-type and mutant plasmids, as well as clinical CMV isolates and plasma samples that were known to contain mutations that confer resistance. Finally, 48 clinical plasma specimens with a range of viral loads (394 to 2,191,011 copies/ml plasma) were sequenced using multiplexing of up to 24 specimens per run. This led to the identification of seven resistance mutations, three of which were present in <20% of the sequenced population. Thus, this assay offers more sensitive detection of minor variants and a higher multiplexing capacity than current methods for the genotypic detection of CMV drug resistance mutations.
Collapse
|
235
|
Nucleosome maps of the human cytomegalovirus genome reveal a temporal switch in chromatin organization linked to a major IE protein. Proc Natl Acad Sci U S A 2013; 110:13126-31. [PMID: 23878222 DOI: 10.1073/pnas.1305548110] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Human CMV (hCMV) establishes lifelong infections in most of us, causing developmental defects in human embryos and life-threatening disease in immunocompromised individuals. During productive infection, the viral >230,000-bp dsDNA genome is expressed widely and in a temporal cascade. The hCMV genome does not carry histones when encapsidated but has been proposed to form nucleosomes after release into the host cell nucleus. Here, we present hCMV genome-wide nucleosome occupancy and nascent transcript maps during infection of permissive human primary cells. We show that nucleosomes occupy nuclear viral DNA in a nonrandom and highly predictable fashion. At early times of infection, nucleosomes associate with the hCMV genome largely according to their intrinsic DNA sequence preferences, indicating that initial nucleosome formation is genetically encoded in the virus. However, as infection proceeds to the late phase, nucleosomes redistribute extensively to establish patterns mostly determined by nongenetic factors. We propose that these factors include key regulators of viral gene expression encoded at the hCMV major immediate-early (IE) locus. Indeed, mutant virus genomes deficient for IE1 expression exhibit globally increased nucleosome loads and reduced nucleosome dynamics compared with WT genomes. The temporal nucleosome occupancy differences between IE1-deficient and WT viruses correlate inversely with changes in the pattern of viral nascent and total transcript accumulation. These results provide a framework of spatial and temporal nucleosome organization across the genome of a major human pathogen and suggest that an hCMV major IE protein governs overall viral chromatin structure and function.
Collapse
|
236
|
Jacob CL, Lamorte L, Sepulveda E, Lorenz IC, Gauthier A, Franti M. Neutralizing antibodies are unable to inhibit direct viral cell-to-cell spread of human cytomegalovirus. Virology 2013; 444:140-7. [PMID: 23849792 DOI: 10.1016/j.virol.2013.06.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 04/03/2013] [Accepted: 06/05/2013] [Indexed: 12/17/2022]
Abstract
Infection with human cytomegalovirus (CMV) during pregnancy is the most common cause of congenital disorders, and can lead to severe life-long disabilities with associated high cost of care. Since there is no vaccine or effective treatment, current efforts are focused on identifying potent neutralizing antibodies. A panel of CMV monoclonal antibodies identified from patent applications, was synthesized and expressed in order to reproduce data from the literature showing that anti-glycoprotein B antibodies neutralized virus entry into all cell types and that anti-pentameric complex antibodies are highly potent in preventing virus entry into epithelial cells. It had not been established whether antibodies could prevent subsequent rounds of infection that are mediated primarily by direct cell-to-cell transmission. A thorough validation of a plaque reduction assay to monitor cell-to-cell spread led to the conclusion that neutralizing antibodies do not significantly inhibit plaque formation or reduce plaque size when they are added post-infection.
Collapse
Affiliation(s)
- Christian L Jacob
- Boehringer Ingelheim (Canada) Ltd., 2100 Rue Cunard, Laval, Québec, Canada
| | | | | | | | | | | |
Collapse
|
237
|
A fluorescence-based high-throughput screening assay for identifying human cytomegalovirus inhibitors. Methods Mol Biol 2013. [PMID: 23821279 DOI: 10.1007/978-1-62703-484-5_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Human cytomegalovirus (HCMV) is a common opportunistic pathogen that can cause devastating -morbidity and mortality amongst neonates and immune-compromised patients. The current standard of care for HCMV infection is limited to four antiviral compounds that have major limitations in terms of long--term use, toxicity, and use during pregnancy. To provide patients with alternative treatment options to decrease HCMV-related morbidity and mortality, new drugs with novel modes of action are warranted. Here, we describe a validated high-throughput fluorescence antiviral screening assay based on infection of fibroblast cells with a fluorescently tagged reference strain of HCMV (AD169-GFP) to screen and profile HCMV inhibitors.
Collapse
|
238
|
Wistuba-Hamprecht K, Frasca D, Blomberg B, Pawelec G, Derhovanessian E. Age-associated alterations in γδ T-cells are present predominantly in individuals infected with Cytomegalovirus. IMMUNITY & AGEING 2013; 10:26. [PMID: 23822093 PMCID: PMC3703274 DOI: 10.1186/1742-4933-10-26] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 06/29/2013] [Indexed: 01/25/2023]
Abstract
Background Despite the common perception that latent Cytomegalovirus (CMV) infection is usually symptom-free, emerging epidemiological evidence suggests that it may in fact be associated with higher mortality over extended follow-up. Mechanisms responsible for this potentially important effect are unclear. CMV infection is known to have a large impact on the distribution of T cell phenotypes, especially the accumulation of late-stage differentiated CD8+, as well as Vδ2- γδ T-cells, which are the main subset of γδ T-cells involved in anti-CMV immunity. Its impact on γδ T-cells in the aging context is less well-defined. Results Here, we investigated a group of healthy individuals aged between 21 and 89 years, in order to correlate the frequency and differentiation status of γδ T-cells with age. We found that these parameters were only marginally influenced by age, but were marked in people with a latent CMV infection. Thus, we observed a significant age-associated accumulation of late-differentiated T-cells within the Vδ2- population, but only in CMV-seropositive donors. There was also a strong trend towards reduced frequency of early-differentiated cells within the Vδ2- phenotype. Older people had significantly higher anti-CMV IgG titers, which in turn correlated significantly with a lower Vδ2+/Vδ2- ratio and a shift from early- to a late-differentiated Vδ2- T-cell phenotype. Conclusions Our findings demonstrate a strong influence of CMV on γδ T-cells during human ageing, similar to that observed for αβ T-cells. Differences between donors of different ages are more marked in CMV-infected individuals. The biological implications of this potent age-associated CMV-mediated immune-modulation require clarification.
Collapse
Affiliation(s)
- Kilian Wistuba-Hamprecht
- Department of Internal Medicine II, Centre for Medical Research, University of Tübingen, Tübingen, Germany.
| | | | | | | | | |
Collapse
|
239
|
Myeloblastic cell lines mimic some but not all aspects of human cytomegalovirus experimental latency defined in primary CD34+ cell populations. J Virol 2013; 87:9802-12. [PMID: 23824798 DOI: 10.1128/jvi.01436-13] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Human cytomegalovirus (HCMV) is a significant human pathogen that achieves lifelong persistence by establishing latent infections in undifferentiated cells of the myeloid lineage, such as CD34(+) hematopoietic progenitor cells. When latency is established, viral lytic gene expression is silenced in part by a cellular intrinsic defense consisting of Daxx and histone deacetylases (HDACs) because pp71, the tegument transactivator that travels to the nucleus and inactivates this defense at the start of a lytic infection in differentiated cells, remains in the cytoplasm. Because the current in vitro and ex vivo latency models have physiological and practical limitations, we evaluated two CD34(+) myeloblastic cell lines, KG-1 and Kasumi-3, for their ability to establish, maintain, and reactivate HCMV experimental latent infections. Tegument protein pp71 was cytoplasmic, and immediate-early (IE) genes were silenced as in primary CD34(+) cells. However, in contrast to what occurs in primary CD34(+) cells ex vivo or in NT2 and THP-1 in vitro model systems, viral IE gene expression from the laboratory-adapted AD169 genome was not induced in the presence of HDAC inhibitors in either KG-1 or Kasumi-3 cells. Furthermore, while the clinical strain FIX was able to reactivate from Kasumi-3 cells, AD169 was not, and neither strain reactivated from KG-1 cells. Thus, KG-1 and Kasumi-3 experimental latent infections differ in important parameters from those in primary CD34(+) cell populations. Aspects of latency illuminated through the use of these myeloblastoid cell lines should not be considered independently but integrated with results obtained in primary cell systems when paradigms for HCMV latency are proposed.
Collapse
|
240
|
Wang H, Yao Y, Huang C, Chen Q, Chen J, Chen Z. Immunization with cytomegalovirus envelope glycoprotein M and glycoprotein N DNA vaccines can provide mice with complete protection against a lethal murine cytomegalovirus challenge. Virol Sin 2013; 28:174-82. [PMID: 23715998 DOI: 10.1007/s12250-013-3330-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 05/20/2013] [Indexed: 01/10/2023] Open
Abstract
Human cytomegalovirus virions contain three major glycoprotein complexes (gC I, II, III), all of which are required for CMV infectivity. These complexes also represent major antigenic targets for anti-viral immune responses. The gC II complex consists of two glycoproteins, gM and gN. In the current study, DNA vaccines expressing the murine cytomegalovirus (MCMV) homologs of the gM and gN proteins were evaluated for protection against lethal MCMV infection in a mouse model. Humoral and cellular immune responses, spleen viral titers, and mice survival and body-weight changes were examined. The results showed that immunization with gM or gN DNA vaccine alone was not able to offer good protection, whereas co-immunization with both gM and gN induced an effective neutralizing antibody response and cellular immune response, and provided mice with complete protection against a lethal MCMV challenge. This study provides the first in vivo evidence that the gC II (gM-gN) complex may be able to serve as a protective subunit antigen for future HCMV vaccine development.
Collapse
Affiliation(s)
- Huadong Wang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | | | | | | | | | | |
Collapse
|
241
|
Ameres S, Mautner J, Schlott F, Neuenhahn M, Busch DH, Plachter B, Moosmann A. Presentation of an immunodominant immediate-early CD8+ T cell epitope resists human cytomegalovirus immunoevasion. PLoS Pathog 2013; 9:e1003383. [PMID: 23717207 PMCID: PMC3662661 DOI: 10.1371/journal.ppat.1003383] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 04/10/2013] [Indexed: 02/07/2023] Open
Abstract
Control of human cytomegalovirus (HCMV) depends on CD8+ T cell responses that are shaped by an individual's repertoire of MHC molecules. MHC class I presentation is modulated by a set of HCMV-encoded proteins. Here we show that HCMV immunoevasins differentially impair T cell recognition of epitopes from the same viral antigen, immediate-early 1 (IE-1), that are presented by different MHC class I allotypes. In the presence of immunoevasins, HLA-A- and HLA-B-restricted T cell clones were ineffective, but HLA-C*0702-restricted T cell clones recognized and killed infected cells. Resistance of HLA-C*0702 to viral immunoevasins US2 and US11 was mediated by the alpha3 domain and C-terminal region of the HLA heavy chain. In healthy donors, HLA-C*0702-restricted T cells dominated the T cell response to IE-1. The same HLA-C allotype specifically protected infected cells from attack by NK cells that expressed a corresponding HLA-C-specific KIR. Thus, allotype-specific viral immunoevasion allows HCMV to escape control by NK cells and HLA-A- and HLA-B-restricted T cells, while the virus becomes selectively vulnerable to an immunodominant population of HLA-C-restricted T cells. Our work identifies a T cell population that may be of particular efficiency in HCMV-specific immunotherapy.
Collapse
Affiliation(s)
- Stefanie Ameres
- Clinical Cooperation Group Immunooncology, Department of Medicine III, Klinikum der Universität München, and Department of Gene Vectors, Helmholtz Zentrum München, Munich, Germany
- DZIF – German Center for Infection Research, Munich, Germany
| | - Josef Mautner
- DZIF – German Center for Infection Research, Munich, Germany
- Clinical Cooperation Group Pediatric Tumor Immunology, Helmholtz Zentrum München, and Children's Hospital, Technische Universität München, Munich, Germany
| | - Fabian Schlott
- DZIF – German Center for Infection Research, Munich, Germany
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München, Munich, Germany
- Clinical Cooperation Group Immune Monitoring, Helmholtz Zentrum München and Technische Universität München, Munich, Germany
| | - Michael Neuenhahn
- DZIF – German Center for Infection Research, Munich, Germany
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München, Munich, Germany
- Clinical Cooperation Group Immune Monitoring, Helmholtz Zentrum München and Technische Universität München, Munich, Germany
| | - Dirk H. Busch
- DZIF – German Center for Infection Research, Munich, Germany
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München, Munich, Germany
- Clinical Cooperation Group Immune Monitoring, Helmholtz Zentrum München and Technische Universität München, Munich, Germany
| | - Bodo Plachter
- Institute for Virology, University Medical Center, Johannes-Gutenberg-Universität Mainz, Mainz, Germany
| | - Andreas Moosmann
- Clinical Cooperation Group Immunooncology, Department of Medicine III, Klinikum der Universität München, and Department of Gene Vectors, Helmholtz Zentrum München, Munich, Germany
- DZIF – German Center for Infection Research, Munich, Germany
- * E-mail:
| |
Collapse
|
242
|
An endothelial cell-specific requirement for the UL133-UL138 locus of human cytomegalovirus for efficient virus maturation. J Virol 2013; 87:3062-75. [PMID: 23283945 DOI: 10.1128/jvi.02510-12] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Human cytomegalovirus (HCMV) infects a variety of cell types in humans, resulting in a varied pathogenesis in the immunocompromised host. Endothelial cells (ECs) are considered an important target of HCMV infection that may contribute to viral pathogenesis. Although the viral determinants important for entry into ECs are well defined, the molecular determinants regulating postentry tropism in ECs are not known. We previously identified the UL133-UL138 locus encoded within the clinical strain-specific ULb' region of the HCMV genome as important for the latent infection in CD34(+) hematopoietic progenitor cells (HPCs). Interestingly, this locus, while dispensable for replication in fibroblasts, was required for efficient replication in ECs infected with the TB40E or fusion-inducing factor X (FIX) HCMV strains. ECs infected with a virus lacking the entire locus (UL133-UL138(NULL) virus) complete the immediate-early and early phases of infection but are defective for infectious progeny virus production. ECs infected with UL133-UL138(NULL) virus exhibited striking differences in the organization of intracellular membranes and in the assembly of mature virions relative to ECs infected with wild-type (WT) virus. In UL133-UL138(NULL) virus-infected ECs, Golgi stacks were disrupted, and the viral assembly compartment characteristic of HCMV infection failed to form. Further, progeny virions in UL133-UL138(NULL) virus-infected ECs inefficiently acquired the virion tegument and secondary envelope. These defects were specific to infection in ECs and not observed in fibroblasts infected with UL133-UL138(NULL) virus, suggesting an EC-specific requirement for the UL133-UL138 locus for late stages of replication. To our knowledge, the UL133-UL138 locus represents the first cell-type-dependent, postentry tropism determinant required for viral maturation.
Collapse
|
243
|
Al-Awadhi R, Al-Harmi J, AlFadhli S. Prevalence of cytomegalovirus DNA in cord blood and voided urine obtained from pregnant women at the end of pregnancy. Med Princ Pract 2013; 22:194-9. [PMID: 23075743 PMCID: PMC5586727 DOI: 10.1159/000343167] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Accepted: 09/03/2012] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE This study was undertaken to determine the prevalence of congenital cytomegalovirus (CMV) infection in pregnant women at the end of pregnancy in Kuwait using cord blood and maternal urine. SUBJECTS AND METHODS Urine samples were collected prior to childbirth, and cord blood was collected immediately after delivery from 983 women. Anti-CMV IgG and IgM antibodies were determined using ELISA; CMV DNA was detected using nested PCR, and viral load was calculated using real-time PCR. CMV concentration in samples was categorized as low when the viral load ≤10(3) copies/µl, intermediate when the viral load = 10(3)-10(4) copies/µl, and high when the viral load >10(4) copies/µl. The cord blood serology outcome was compared to cord blood PCR, cord blood viral load, maternal urine PCR and viral load analyses. RESULTS Serology showed that of the 983 cord blood samples, 89 (9%) were positive for anti-CMV IgM antibodies; PCR test showed 44 (4.5%) contained CMV DNA, and there was a high viral load in all. Maternal urine PCR showed that 9 (10.11%) women had CMV DNA, and there was a high viral load in 7 (78%). The kappa test for measures of agreement showed a reasonable agreement (0.45) between cord blood PCR and urine PCR. CONCLUSION This study showed that CMV infection in the cord blood sera of pregnant women is common in Kuwait and highlights the need for more clinically based studies to follow up newborns with congenital CMV infection.
Collapse
Affiliation(s)
- Rana Al-Awadhi
- Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, Jabriya, Kuwait
- *Dr. R. Al-Awadhi, Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, Health Sciences Centre, Kuwait University, PO Box 31470, Sulaibikhat 90805 (Kuwait), E-Mail
| | - Jehad Al-Harmi
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Kuwait University, Jabriya, Kuwait
| | - Suad AlFadhli
- Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, Jabriya, Kuwait
| |
Collapse
|
244
|
γδT cells elicited by CMV reactivation after allo-SCT cross-recognize CMV and leukemia. Leukemia 2013; 27:1328-38. [DOI: 10.1038/leu.2012.374] [Citation(s) in RCA: 152] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
245
|
Verghese PS, Schleiss MR. Letermovir Treatment of Human Cytomegalovirus Infection Antiinfective Agent. DRUG FUTURE 2013; 38:291-298. [PMID: 24163496 DOI: 10.1358/dof.2013.038.05.1946425] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Novel therapies are urgently needed for the management of cytomegalovirus (CMV) disease in high-risk patients. Currently licensed agents target the viral DNA polymerase, and although they are effective, they are fraught with toxicities to patients. Moreover, emergence of antiviral resistance is an increasing problem, particularly for patients on long-term suppressive therapy. A new agent, letermovir (AIC246), shows great promise for the management of CMV infection. Advantages include its good oral bioavailability, its lack of toxicity, and the apparent absence of drug-drug interactions. Letermovir has a novel mechanism of action, exerting its antiviral effect by interfering with the viral pUL56 gene product and in the process disrupting the viral terminase complex. This agent demonstrates substantial promise as an alternative to more toxic antivirals in patients at high risk for CMV disease, particularly in the transplantation setting.
Collapse
Affiliation(s)
- Priya S Verghese
- University of Minnesota Medical School Department of Pediatrics, Division of Pediatric Nephrology, Amplatz Children's Hospital, East Building, MB680, 2414 South 7th Street, Minneapolis, MN 55454,
| | | |
Collapse
|
246
|
Mindel A, Dwyer D, Herring B, Cunningham AL. Global Epidemiology of Sexually Transmitted Diseases. Sex Transm Dis 2013. [DOI: 10.1016/b978-0-12-391059-2.00001-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
247
|
Cytotoxic T lymphocytes for the treatment of viral infections and posttransplant lymphoproliferative disorders in transplant recipients. Curr Opin Infect Dis 2012; 25:431-7. [PMID: 22614521 DOI: 10.1097/qco.0b013e3283551dd3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW The continuous and successful expansion of organ transplants is unfortunately associated with increased incidence of severe opportunistic viral infections and Epstein-Barr virus (EBV)-related lymphomas secondary to immunosuppression. Here, we review the strengths and limitations of T-cell-based strategies used to treat viral infections in immunocompromised individuals. RECENT FINDINGS While current antiviral drugs are often suboptimal because of associated toxicities, a promising approach in the management of infections with viruses like cytomegalovirus (CMV), adenovirus (AdV) and EBV is the adoptive transfer of T cells targeting these viruses that can be directly isolated from the peripheral blood of the donor or expanded ex vivo prior to infusions in patients. SUMMARY T-cell-based immunotherapies are now being included in the clinical practice of transplant recipients to prevent and treat infections and complications associated with CMV, AdV and EBV. Improvement of current limitations will enable the extension of these approaches to all patients at risk and to other clinically relevant viruses and pathogens that are emerging as significant complications for immunocompromised patients.
Collapse
|
248
|
Penkert RR, Kalejta RF. Tale of a tegument transactivator: the past, present and future of human CMV pp71. Future Virol 2012; 7:855-869. [PMID: 23378857 DOI: 10.2217/fvl.12.86] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Herpesviruses assemble large virions capable of delivering to a newly infected cell not only the viral genome, but also viral proteins packaged within the tegument layer between the DNA-containing capsid and the lipid envelope. In this review, we describe the tegument transactivator of the β-herpesvirus human CMV, the pp71 protein. We present the known mechanistic features through which it activates viral gene expression during a lytic infection but fails to do so when the virus establishes latency, and describe how pp71 stimulates the cell cycle and may help infected cells avoid detection by the adaptive immune system. A historical overview of pp71 is extended with current perceptions of its roles during human CMV infections and suggestions for future avenues of experimentation.
Collapse
Affiliation(s)
- Rhiannon R Penkert
- Institute for Molecular Virology & McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, 1525 Linden Drive, Madison, WI 53706, USA
| | | |
Collapse
|
249
|
Petrucelli A, Umashankar M, Zagallo P, Rak M, Goodrum F. Interactions between proteins encoded within the human cytomegalovirus UL133-UL138 locus. J Virol 2012; 86:8653-62. [PMID: 22674978 PMCID: PMC3421763 DOI: 10.1128/jvi.00465-12] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Accepted: 05/22/2012] [Indexed: 12/20/2022] Open
Abstract
We previously described a novel genetic locus within the ULb' region of the human cytomegalovirus (HCMV) genome that, while dispensable for replication in fibroblasts, suppresses replication in hematopoietic progenitors and augments replication in endothelial cells. This locus, referred to as the UL133-UL138 locus, encodes four proteins, pUL133, pUL135, pUL136, and pUL138. In this work, we have mapped the interactions among these proteins. An analysis of all pairwise interactions during transient expression revealed a robust interaction between pUL133 and pUL138. Potential interactions between pUL136 and both pUL133 and pUL138 were also revealed. In addition, each of the UL133-UL138 locus proteins self-associated, suggesting a potential to form higher-order homomeric complexes. As both pUL133 and pUL138 function in promoting viral latency in CD34(+) hematopoietic progenitor cells (HPCs) infected in vitro, we further focused on this interaction. pUL133 and pUL138 are the predominant complex detected when all proteins are expressed together and require no other proteins in the locus for their association. During infection, the interaction between pUL133 and pUL138 or pUL136 can be detected. A recombinant virus that fails to express both pUL133 and pUL138 exhibited a latency phenotype similar to that of viruses that fail to express either pUL133 or pUL138, indicating that these proteins function cooperatively in latency and do not have independent functions that additively contribute to HCMV latency. These studies identify protein interactions among proteins encoded by the UL133-UL138 locus and demonstrate an important interaction impacting the outcome of HCMV infection.
Collapse
Affiliation(s)
| | | | | | - Michael Rak
- BIO5 Institute, University of Arizona, Tucson, Arizona, USA
| | - Felicia Goodrum
- Department of Immunobiology
- BIO5 Institute, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
250
|
Histone H3 lysine 4 methylation marks postreplicative human cytomegalovirus chromatin. J Virol 2012; 86:9817-27. [PMID: 22761369 DOI: 10.1128/jvi.00581-12] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
In the nuclei of permissive cells, human cytomegalovirus genomes form nucleosomal structures initially resembling heterochromatin but gradually switching to a euchromatin-like state. This switch is characterized by a decrease in histone H3 K9 methylation and a marked increase in H3 tail acetylation and H3 K4 methylation across the viral genome. We used ganciclovir and a mutant virus encoding a reversibly destabilized DNA polymerase to examine the impact of DNA replication on histone modification dynamics at the viral chromatin. The changes in H3 tail acetylation and H3 K9 methylation proceeded in a DNA replication-independent fashion. In contrast, the increase in H3 K4 methylation proved to depend widely on viral DNA synthesis. Consistently, labeling of nascent DNA using "click chemistry" revealed preferential incorporation of methylated H3 K4 into viral (but not cellular) chromatin during or following DNA replication. This study demonstrates largely selective epigenetic tagging of postreplicative human cytomegalovirus chromatin.
Collapse
|