1
|
Venturini C, Colston JM, Charles O, Lankina A, Best T, Atkinson C, Forrest C, Williams CA, Rao K, Worth A, Thorburn D, Harber M, Griffiths P, Breuer J. Persistent Low-Level Variants in a Subset of Viral Genes Are Highly Predictive of Poor Outcome in Immunocompromised Patients With Cytomegalovirus Infection. J Infect Dis 2024; 230:e427-e436. [PMID: 38181168 PMCID: PMC11326829 DOI: 10.1093/infdis/jiae001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 12/15/2023] [Accepted: 01/02/2024] [Indexed: 01/07/2024] Open
Abstract
BACKGROUND Human cytomegalovirus (HCMV) is the most common and serious opportunistic infection after solid organ and hematopoietic stem cell transplantation. In this study, we used whole-genome HCMV data to investigate viral factors associated with the clinical outcome. METHODS We sequenced HCMV samples from 16 immunocompromised pediatric patients with persistent viremia. Eight of the 16 patients died of complications due to HCMV infection. We also sequenced samples from 35 infected solid organ adult recipients, of whom 1 died with HCMV infection. RESULTS We showed that samples from both groups have fixed variants at resistance sites and mixed infections. Next-generation sequencing also revealed nonfixed variants at resistance sites in most of the patients who died (6/9). A machine learning approach identified 10 genes with nonfixed variants in these patients. These genes formed a viral signature that discriminated patients with HCMV infection who died from those who survived with high accuracy (area under the curve = 0.96). Lymphocyte numbers for a subset of patients showed no recovery posttransplant in the patients who died. CONCLUSIONS We hypothesize that the viral signature identified in this study may be a useful biomarker for poor response to antiviral drug treatment and indirectly for poor T-cell function, potentially identifying early those patients requiring nonpharmacological interventions.
Collapse
Affiliation(s)
- Cristina Venturini
- Infection, Immunity and Inflammation, Institute of Child Health, University College London (UCL)
| | - Julia M Colston
- North Bristol National Health Service (NHS) Trust, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol
| | - Oscar Charles
- Infection, Immunity and Inflammation, Institute of Child Health, University College London (UCL)
| | - Anastasia Lankina
- Infection, Immunity and Inflammation, Institute of Child Health, University College London (UCL)
| | - Timothy Best
- Virology Department, Great Ormond Street Hospital for Children NHS Foundation Trust
| | - Claire Atkinson
- Applied Science, London South Bank University
- Division of Infection and Immunity, Institute for Immunity and Transplantation, UCL
| | - Calum Forrest
- Division of Infection and Immunity, Institute for Immunity and Transplantation, UCL
| | - Charlotte A Williams
- Department of Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, UCL
| | | | - Austen Worth
- Department of Immunology, Great Ormond Street Hospital for Children NHS Foundation Trust, London
| | - Doug Thorburn
- Department of Nephrology, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Mark Harber
- Department of Nephrology, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Paul Griffiths
- Division of Infection and Immunity, Institute for Immunity and Transplantation, UCL
| | - Judith Breuer
- Infection, Immunity and Inflammation, Institute of Child Health, University College London (UCL)
- Virology Department, Great Ormond Street Hospital for Children NHS Foundation Trust
| |
Collapse
|
2
|
Külekci B, Mollik M, Schwarz S, Perkmann-Nagele N, Geleff S, Jaksch P, Hoetzenecker K, Lambers C, Puchhammer-Stöckl E, Goerzer I. Bidirectional transfer of human cytomegalovirus strains in donor and recipient seropositive lung transplant patients. J Med Virol 2024; 96:e29770. [PMID: 38949200 DOI: 10.1002/jmv.29770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/29/2024] [Accepted: 06/11/2024] [Indexed: 07/02/2024]
Abstract
Donor and recipient human cytomegalovirus (HCMV) seropositive (D+R+) lung transplant recipients (LTRs) often harbor multiple strains of HCMV, likely due to transmitted donor (D) strains and reactivated recipient (R) strains. To date, the extent and timely occurrence of each likely source in shaping the post-transplantation (post-Tx) strain population is unknown. Here, we deciphered the D and R origin of the post-Tx HCMV strain composition in blood, bronchoalveolar lavage (BAL), and CD45+ BAL cell subsets. We investigated either D and/or R formalin-fixed paraffin-embedded blocks or fresh D lung tissue from four D+R+ LTRs obtained before transplantation. HCMV strains were characterized by short amplicon deep sequencing. In two LTRs, we show that the transplanted lung is reseeded by R strains within the first 6 months after transplantation, likely by infiltrating CD14+ CD163+/- alveolar macrophages. In three LTRs, we demonstrate both rapid D-strain dissemination and persistence in the transplanted lung for >1 year post-Tx. Broad inter-host diversity contrasts with intra-host genotype sequence stability upon transmission, during follow-up and across compartments. In D+R+ LTRs, HCMV strains of both, D and R origin can emerge first and dominate long-term in subsequent episodes of infection, indicating replication of both sources despite pre-existing immunity.
Collapse
Affiliation(s)
- Büsra Külekci
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Madlen Mollik
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Stefan Schwarz
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Nicole Perkmann-Nagele
- Divison of Clinical Virology, Department of Laboratory Medicine, University of Vienna, Vienna, Austria
| | - Silvana Geleff
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Peter Jaksch
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Konrad Hoetzenecker
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Christopher Lambers
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | | | - Irene Goerzer
- Center for Virology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
3
|
Vankova O, Brusnigina N, Novikova N. NGS Technology in Monitoring the Genetic Diversity of Cytomegalovirus Strains. Sovrem Tekhnologii Med 2023; 15:41-46. [PMID: 37389021 PMCID: PMC10306971 DOI: 10.17691/stm2023.15.2.04] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Indexed: 07/01/2023] Open
Abstract
Modern molecular genetic methods, massive parallel sequencing in particular, allow for genotyping of various pathogens with the aim of their epidemiological marking and improvement of molecular epidemiological surveillance of actual infections, including cytomegalovirus infection. The aim of the study is to evaluate the next-generation sequencing (NGS) technology for genotyping clinical isolates of cytomegalovirus (CMV). Materials and Methods The object of the study were samples of biological substrates (leukocyte mass, saliva, urine) taken from patients who underwent liver and kidney transplantation. Detection of CMV DNA was carried out by a real-time PCR using commercial diagnostic AmpliSense CMV-FL test systems (Central Research Institute for Epidemiology, Moscow, Russia). DNA extraction was performed using DNA-sorb AM and DNA-sorb V kits (Central Research Institute for Epidemiology) in accordance with manufacturer's manual. The quality of the prepared DNA library for sequencing was assessed by means of the QIAxcel Advanced System capillary gel electrophoresis system (QIAGEN, Germany). Alignment and assembly of nucleotide sequences were carried out using CLC Genomics Workbench 5.5 software (CLC bio, USA). The sequencing results were analyzed using BLAST of NCBI server. Results CMV DNA samples were selected for genotyping. The two variable genes, UL55(gB) and UL73(gN), were used for CMV genotype determination, which was performed using NGS technology MiSeq sequencer (Illumina, USA). Based on the exploratory studies and analysis of literature sources, primers for genotyping on the UL55(gB) and UL73(gN) genes have been selected and the optimal conditions for the PCR reaction have been defined. The results of sequencing the UL55(gB) and UL73(gN) gene fragments of CMV clinical isolates from recipients of solid organs made it possible to determine the virus genotypes, among which gB2, gN4c, and gN4b were dominant. In some cases, association of two and three CMV genotypes has been revealed. Conclusion The application of the NGS technology for genotyping cytomegalovirus strains can become one of the main methods of CMV infection molecular epidemiology, as it allows for obtaining reliable results with a significant reduction in research time.
Collapse
Affiliation(s)
- O.E. Vankova
- Head Researcher, Laboratory of Metagenomics and Molecular Indication of Pathogens; Academician I.N. Blokhina Nizhny Novgorod Scientific Research Institute of Epidemiology and Microbiology of Rospotrebnadzor (Russian Federal Consumer Rights Protection and Human Health Control Service), 71 Malaya Yamskaya St., Nizhny Novgorod, 603950, Russia
| | - N.F. Brusnigina
- Head of the Laboratory of Metagenomics and Molecular Indication of Pathogens; Academician I.N. Blokhina Nizhny Novgorod Scientific Research Institute of Epidemiology and Microbiology of Rospotrebnadzor (Russian Federal Consumer Rights Protection and Human Health Control Service), 71 Malaya Yamskaya St., Nizhny Novgorod, 603950, Russia
| | - N.A. Novikova
- Professor, Head of the Laboratory of Molecular Epidemiology of Viral Infections Academician I.N. Blokhina Nizhny Novgorod Scientific Research Institute of Epidemiology and Microbiology of Rospotrebnadzor (Russian Federal Consumer Rights Protection and Human Health Control Service), 71 Malaya Yamskaya St., Nizhny Novgorod, 603950, Russia Modern molecular genetic methods, massive parallel sequencing in particular, allow for genotyping of various pathogens with the aim of their epidemiological marking and improvement of molecular epidemiological surveillance of actual infections, including cytomegalovirus infection
| |
Collapse
|
4
|
Brait N, Külekçi B, Goerzer I. Long range PCR-based deep sequencing for haplotype determination in mixed HCMV infections. BMC Genomics 2022; 23:31. [PMID: 34991471 PMCID: PMC8735729 DOI: 10.1186/s12864-021-08272-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 12/03/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Short read sequencing has been used extensively to decipher the genome diversity of human cytomegalovirus (HCMV) strains, but falls short to reveal individual genomes in mixed HCMV strain populations. Novel third-generation sequencing platforms offer an extended read length and promise to resolve how distant polymorphic sites along individual genomes are linked. In the present study, we established a long amplicon PacBio sequencing workflow to identify the absolute and relative quantities of unique HCMV haplotypes spanning over multiple hypervariable sites in mixtures. Initial validation of this approach was performed with defined HCMV DNA templates derived from cell-culture enriched viruses and was further tested for its suitability on patient samples carrying mixed HCMV infections. RESULTS Total substitution and indel error rate of mapped reads ranged from 0.17 to 0.43% depending on the stringency of quality trimming. Artificial HCMV DNA mixtures were correctly determined down to 1% abundance of the minor DNA source when the total HCMV DNA input was 4 × 104 copies/ml. PCR products of up to 7.7 kb and a GC content < 55% were efficiently generated when DNA was directly isolated from patient samples. In a single sample, up to three distinct haplotypes were identified showing varying relative frequencies. Alignments of distinct haplotype sequences within patient samples showed uneven distribution of sequence diversity, interspersed by long identical stretches. Moreover, diversity estimation at single polymorphic regions as assessed by short amplicon sequencing may markedly underestimate the overall diversity of mixed haplotype populations. CONCLUSIONS Quantitative haplotype determination by long amplicon sequencing provides a novel approach for HCMV strain characterisation in mixed infected samples which can be scaled up to cover the majority of the genome by multi-amplicon panels. This will substantially improve our understanding of intra-host HCMV strain diversity and its dynamic behaviour.
Collapse
Affiliation(s)
- Nadja Brait
- Center for Virology, Medical University of Vienna, Vienna, Austria
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, Netherlands
| | - Büşra Külekçi
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Irene Goerzer
- Center for Virology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
5
|
Pigliasco F, Cafaro A, Simeoli R, Barco S, Magnasco A, Faraci M, Tripodi G, Goffredo BM, Cangemi G. A UHPLC-MS/MS Method for Therapeutic Drug Monitoring of Aciclovir and Ganciclovir in Plasma and Dried Plasma Spots. Biomedicines 2021; 9:biomedicines9101379. [PMID: 34680495 PMCID: PMC8533239 DOI: 10.3390/biomedicines9101379] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 01/06/2023] Open
Abstract
The role of therapeutic drug monitoring (TDM) of valaciclovir (VA)/aciclovir (A) and valganciclovir/ganciclovir (VG/G) in critically ill patients is still a matter of debate. More data on the dose–concentration relationship might therefore be useful, especially in pediatrics where clinical practice is not adequately supported by robust PK studies. We developed and validated a new liquid chromatography-tandem mass spectrometry (LC-MS/MS) micro-method to simultaneously quantify A and G from plasma and dried plasma spots (DPS). The method was based on rapid organic extraction from DPS and separation on a reversed-phase C-18 UHPLC column after addition of deuterated internal standards. Accurate analyte quantification using SRM detection was then obtained using a Thermo Fisher Quantiva triple-quadrupole MS coupled to an Ultimate 3000 UHPLC. It was validated following international (EMA) guidelines for bioanalytical method validation and was tested on samples from pediatric patients treated with A, VG, or G for cytomegalovirus infection following solid organ or hematopoietic stem cell transplantation. Concentrations obtained from plasma and DPS were compared using Passing–Bablok and Bland–Altman statistical tests. The assay was linear over wide concentration ranges (0.01–20 mg/L) in both plasma and DPS for A and G, suitable for the expected therapeutic ranges for both Cmin and Cmax, accurate, and reproducible in the absence of matrix effects. The results obtained from plasma and DPS were comparable. Using an LC-MS/MS method allowed us to obtain a very specific, sensitive, and rapid quantification of these antiviral drugs starting from very low volumes (50 μL) of plasma samples and DPS. The stability of analytes for at least 30 days allows for cost-effective shipment and storage at room temperature. Our method is suitable for TDM and could be helpful for improving knowledge on PK/PD targets of antivirals in critically ill pediatric patients.
Collapse
Affiliation(s)
- Federica Pigliasco
- Chromatography and Mass Spectrometry Section, Central Laboratory of Analysis, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (F.P.); (A.C.); (G.T.); (G.C.)
| | - Alessia Cafaro
- Chromatography and Mass Spectrometry Section, Central Laboratory of Analysis, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (F.P.); (A.C.); (G.T.); (G.C.)
| | - Raffaele Simeoli
- Department of Pediatric Specialties, Division of Metabolic Biochemistry, Children’s Hospital Bambino Gesù, IRCCS, 00165 Rome, Italy; (R.S.); (B.M.G.)
| | - Sebastiano Barco
- Chromatography and Mass Spectrometry Section, Central Laboratory of Analysis, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (F.P.); (A.C.); (G.T.); (G.C.)
- Correspondence: ; Tel.: +39-01056363289
| | - Alberto Magnasco
- Pediatric Nephrology, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy;
| | - Maura Faraci
- Hematopoietic Stem Cell Unit, Department of Pediatric Hematology and Oncology, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy;
| | - Gino Tripodi
- Chromatography and Mass Spectrometry Section, Central Laboratory of Analysis, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (F.P.); (A.C.); (G.T.); (G.C.)
| | - Bianca Maria Goffredo
- Department of Pediatric Specialties, Division of Metabolic Biochemistry, Children’s Hospital Bambino Gesù, IRCCS, 00165 Rome, Italy; (R.S.); (B.M.G.)
| | - Giuliana Cangemi
- Chromatography and Mass Spectrometry Section, Central Laboratory of Analysis, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (F.P.); (A.C.); (G.T.); (G.C.)
| |
Collapse
|
6
|
Saeed H, Thoendel M, Razonable RR. Individualized management of cytomegalovirus in solid organ transplant recipients. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2021. [DOI: 10.1080/23808993.2021.1964951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Huma Saeed
- Division of Infectious Diseases, Department of Medicine and the William J Von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, Minnesota, United States
| | - Matthew Thoendel
- Division of Infectious Diseases, Department of Medicine and the William J Von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, Minnesota, United States
| | - Raymund R Razonable
- Division of Infectious Diseases, Department of Medicine and the William J Von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, Minnesota, United States
| |
Collapse
|
7
|
Cytomegalovirus Glycoprotein Polymorphisms and Increasing Viral Load in Non-Transplant Patients with Hematological Malignancies Undergoing Chemotherapy: A Prospective Observational Study. Infect Dis Ther 2021; 10:1549-1566. [PMID: 34148225 PMCID: PMC8322362 DOI: 10.1007/s40121-021-00457-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 05/10/2021] [Indexed: 12/04/2022] Open
Abstract
Introduction Cytomegalovirus (CMV) predisposes to several clinical complications and is a major cause of morbidity and mortality in immunocompromised patients, including patients with hematological malignancies (HM). The present study was carried out to determine the distribution of CMV glycoprotein B, N, and O (gB, gN, and gO) genotypes and their potential effect on its viral load and on clinical outcomes in a cohort of Tunisian non-hematopoietic stem cell transplant (HSCT) patients with HM undergoing chemotherapy. Methods CMV viral load was evaluated by real-time quantitative PCR. The gB, gN, and gO genotypes of the CMV strains were analyzed by multiplex nested PCR and sequencing. Results This prospective study involved 60 clinical isolates obtained from 60 non-HSCT patients with HM undergoing chemotherapy. Mixed CMV gB, gN, and gO genotypes were the predominant glycoprotein genotypes in 31%, 41.4%, and 46.4% of patients, respectively. Mixed gB genotypes were associated with higher initial levels of CMV load (p = 0.001), increased rate of fever (0.025), and co-infection with other herpesviruses (HHVs) (p = 0.024) more frequently than in single gB genotype. Mixed gN genotypes were more associated with severe lymphopenia (ALC < 500/µL) (p = 0.01) and increased risk of death (p = 0.042) than single gN genotype. Single gO2b genotype had also a more unfavorable outcome (p = 0.009) than the other single gO genotype. Mixed gO genotypes were associated with female gender (p = 0.015), acute leukemia disease (p = 0.036), initial high level of CMV viral load (at least 1000 copies/mL) (p = 0.029), skin rash (p = 0.01) more frequently than in single gO genotype. The gO1a/gN3b linkage was associated with an increased initial viral load (p = 0.012). Conclusion Infection with mixed CMV genotypes was common and multiple gB, gN, and gO genotypes were associated with clinical manifestation and higher viral load. Supplementary Information The online version contains supplementary material available at 10.1007/s40121-021-00457-z.
Collapse
|
8
|
Galitska G, Coscia A, Forni D, Steinbrueck L, De Meo S, Biolatti M, De Andrea M, Cagliani R, Leone A, Bertino E, Schulz T, Santoni A, Landolfo S, Sironi M, Cerboni C, Dell'Oste V. Genetic Variability of Human Cytomegalovirus Clinical Isolates Correlates With Altered Expression of Natural Killer Cell-Activating Ligands and IFN-γ. Front Immunol 2021; 12:532484. [PMID: 33897679 PMCID: PMC8062705 DOI: 10.3389/fimmu.2021.532484] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 03/23/2021] [Indexed: 01/03/2023] Open
Abstract
Human cytomegalovirus (HCMV) infection often leads to systemic disease in immunodeficient patients and congenitally infected children. Despite its clinical significance, the exact mechanisms contributing to HCMV pathogenesis and clinical outcomes have yet to be determined. One of such mechanisms involves HCMV-mediated NK cell immune response, which favors viral immune evasion by hindering NK cell-mediated cytolysis. This process appears to be dependent on the extent of HCMV genetic variation as high levels of variability in viral genes involved in immune escape have an impact on viral pathogenesis. However, the link between viral genome variations and their functional effects has so far remained elusive. Thus, here we sought to determine whether inter-host genetic variability of HCMV influences its ability to modulate NK cell responses to infection. For this purpose, five HCMV clinical isolates from a previously characterized cohort of pediatric patients with confirmed HCMV congenital infection were evaluated by next-generation sequencing (NGS) for genetic polymorphisms, phylogenetic relationships, and multiple-strain infection. We report variable levels of genetic characteristics among the selected clinical strains, with moderate variations in genome regions associated with modulation of NK cell functions. Remarkably, we show that different HCMV clinical strains differentially modulate the expression of several ligands for the NK cell-activating receptors NKG2D, DNAM-1/CD226, and NKp30. Specifically, the DNAM-1/CD226 ligand PVR/CD155 appears to be predominantly upregulated by fast-replicating (“aggressive”) HCMV isolates. On the other hand, the NGK2D ligands ULBP2/5/6 are downregulated regardless of the strain used, while other NK cell ligands (i.e., MICA, MICB, ULBP3, Nectin-2/CD112, and B7-H6) are not significantly modulated. Furthermore, we show that IFN-γ; production by NK cells co-cultured with HCMV-infected fibroblasts is directly proportional to the aggressiveness of the HCMV clinical isolates employed. Interestingly, loss of NK cell-modulating genes directed against NK cell ligands appears to be a common feature among the “aggressive” HCMV strains, which also share several gene variants across their genomes. Overall, even though further studies based on a higher number of patients would offer a more definitive scenario, our findings provide novel mechanistic insights into the impact of HCMV genetic variability on NK cell-mediated immune responses.
Collapse
Affiliation(s)
- Ganna Galitska
- Laboratory of Pathogenesis of Viral Infections, Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy
| | - Alessandra Coscia
- Neonatal Unit, Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy
| | - Diego Forni
- Laboratory of Bioinformatics, Scientific Institute IRCCS E. Medea, Bosisio Parini, Italy
| | - Lars Steinbrueck
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Simone De Meo
- Laboratory of Molecular Immunology and Immunopathology, Department of Molecular Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Matteo Biolatti
- Laboratory of Pathogenesis of Viral Infections, Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy
| | - Marco De Andrea
- Laboratory of Pathogenesis of Viral Infections, Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy.,Center for Translational Research on Autoimmune and Allergic Disease - CAAD, University of Piemonte Orientale, Novara, Italy
| | - Rachele Cagliani
- Laboratory of Bioinformatics, Scientific Institute IRCCS E. Medea, Bosisio Parini, Italy
| | - Agata Leone
- Neonatal Unit, Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy
| | - Enrico Bertino
- Neonatal Unit, Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy
| | - Thomas Schulz
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Angela Santoni
- Laboratory of Molecular Immunology and Immunopathology, Department of Molecular Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Santo Landolfo
- Laboratory of Pathogenesis of Viral Infections, Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy
| | - Manuela Sironi
- Laboratory of Bioinformatics, Scientific Institute IRCCS E. Medea, Bosisio Parini, Italy
| | - Cristina Cerboni
- Laboratory of Molecular Immunology and Immunopathology, Department of Molecular Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Valentina Dell'Oste
- Laboratory of Pathogenesis of Viral Infections, Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy
| |
Collapse
|
9
|
Dhingra A, Götting J, Varanasi PR, Steinbrueck L, Camiolo S, Zischke J, Heim A, Schulz TF, Weissinger EM, Kay-Fedorov PC, Davison AJ, Suárez NM, Ganzenmueller T. Human cytomegalovirus multiple-strain infections and viral population diversity in haematopoietic stem cell transplant recipients analysed by high-throughput sequencing. Med Microbiol Immunol 2021; 210:291-304. [PMID: 34611744 PMCID: PMC8541999 DOI: 10.1007/s00430-021-00722-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 09/21/2021] [Indexed: 01/22/2023]
Abstract
Human cytomegalovirus (HCMV) is an important opportunistic pathogen in allogeneic haematopoietic stem cell transplant (HSCT) recipients. High-throughput sequencing of target-enriched libraries was performed to characterise the diversity of HCMV strains present in this high-risk group. Forty-four HCMV-DNA-positive plasma specimens (median viral input load 321 IU per library) collected at defined time points from 23 HSCT recipients within 80 days of transplantation were sequenced. The genotype distribution for 12 hypervariable HCMV genes and the number of HCMV strains present (i.e. single- vs. multiple-strain infection) were determined for 29 samples from 16 recipients. Multiple-strain infection was observed in seven of these 16 recipients, and five of these seven recipients had the donor (D)/recipient (R) HCMV-serostatus combination D + R + . A very broad range of genotypes was detected, with an intrahost composition that was generally stable over time. Multiple-strain infection was not associated with particular virological or clinical features, such as altered levels or duration of antigenaemia, development of acute graft-versus-host disease or increased mortality. In conclusion, despite relatively low viral plasma loads, a high frequency of multiple-strain HCMV infection and a high strain complexity were demonstrated in systematically collected clinical samples from this cohort early after HSCT. However, robust evaluation of the pathogenic role of intrahost viral diversity and multiple-strain infection will require studies enrolling larger numbers of recipients.
Collapse
Affiliation(s)
- A. Dhingra
- Hannover Medical School, Institute of Virology, Hannover, Germany ,German Center for Infection Research (DZIF), Site Hannover-Braunschweig, Hannover, Germany
| | - J. Götting
- Hannover Medical School, Institute of Virology, Hannover, Germany ,German Center for Infection Research (DZIF), Site Hannover-Braunschweig, Hannover, Germany
| | - P. R. Varanasi
- German Center for Infection Research (DZIF), Site Hannover-Braunschweig, Hannover, Germany ,Department of Haematology, Haemostasis and Oncology, Hannover Medical School, Hannover, Germany ,Present Address: National Centre for Biological Sciences, Bangalore, India
| | - L. Steinbrueck
- Hannover Medical School, Institute of Virology, Hannover, Germany ,German Center for Infection Research (DZIF), Site Hannover-Braunschweig, Hannover, Germany
| | - S. Camiolo
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - J. Zischke
- Hannover Medical School, Institute of Virology, Hannover, Germany ,German Center for Infection Research (DZIF), Site Hannover-Braunschweig, Hannover, Germany
| | - A. Heim
- Hannover Medical School, Institute of Virology, Hannover, Germany ,German Center for Infection Research (DZIF), Site Hannover-Braunschweig, Hannover, Germany
| | - T. F. Schulz
- Hannover Medical School, Institute of Virology, Hannover, Germany ,German Center for Infection Research (DZIF), Site Hannover-Braunschweig, Hannover, Germany
| | - E. M. Weissinger
- German Center for Infection Research (DZIF), Site Hannover-Braunschweig, Hannover, Germany ,Department of Haematology, Haemostasis and Oncology, Hannover Medical School, Hannover, Germany
| | - P. C. Kay-Fedorov
- Hannover Medical School, Institute of Virology, Hannover, Germany ,German Center for Infection Research (DZIF), Site Hannover-Braunschweig, Hannover, Germany
| | - A. J. Davison
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - N. M. Suárez
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - T. Ganzenmueller
- Hannover Medical School, Institute of Virology, Hannover, Germany ,German Center for Infection Research (DZIF), Site Hannover-Braunschweig, Hannover, Germany ,Institute for Medical Virology and Epidemiology, University Hospital Tuebingen, Elfriede-Aulhorn-Str. 6, 72076 Tuebingen, Germany
| |
Collapse
|
10
|
Houldcroft CJ, Jackson SE, Lim EY, Sedikides GX, Davies EL, Atkinson C, McIntosh M, Remmerswaal EBM, Okecha G, Bemelman FJ, Stanton RJ, Reeves M, Wills MR. Assessing Anti-HCMV Cell Mediated Immune Responses in Transplant Recipients and Healthy Controls Using a Novel Functional Assay. Front Cell Infect Microbiol 2020; 10:275. [PMID: 32670891 PMCID: PMC7332694 DOI: 10.3389/fcimb.2020.00275] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 05/08/2020] [Indexed: 12/14/2022] Open
Abstract
HCMV infection, reinfection or reactivation occurs in 60% of untreated solid organ transplant (SOT) recipients. Current clinical approaches to HCMV management include pre-emptive and prophylactic antiviral treatment strategies. The introduction of immune monitoring to better stratify patients at risk of viraemia and HCMV mediated disease could improve clinical management. Current approaches quantify T cell IFNγ responses specific for predominantly IE and pp65 proteins ex vivo, as a proxy for functional control of HCMV in vivo. However, these approaches have only a limited predictive ability. We measured the IFNγ T cell responses to an expanded panel of overlapping peptide pools specific for immunodominant HCMV proteins IE1/2, pp65, pp71, gB, UL144, and US3 in a cohort of D+R- kidney transplant recipients in a longitudinal analysis. Even with this increased antigen diversity, the results show that while all patients had detectable T cell responses, this did not correlate with control of HCMV replication in some. We wished to develop an assay that could directly measure anti-HCMV cell-mediated immunity. We evaluated three approaches, stimulation of PBMC with (i) whole HCMV lysate or (ii) a defined panel of immunodominant HCMV peptides, or (iii) fully autologous infected cells co-cultured with PBMC or isolated CD8+ T cells or NK cells. Stimulation with HCMV lysate often generated non-specific antiviral responses while stimulation with immunodominant HCMV peptide pools produced responses which were not necessarily antiviral despite strong IFNγ production. We demonstrated that IFNγ was only a minor component of secreted antiviral activity. Finally, we used an antiviral assay system to measure the effect of whole PBMC, and isolated CD8+ T cells and NK cells to control HCMV in infected autologous dermal fibroblasts. The results show that both PBMC and especially CD8+ T cells from HCMV seropositive donors have highly specific antiviral activity against HCMV. In addition, we were able to show that NK cells were also antiviral, but the level of this control was highly variable between donors and not dependant on HCMV seropositivity. Using this approach, we show that non-viraemic D+R+ SOT recipients had significant and specific antiviral activity against HCMV.
Collapse
Affiliation(s)
- Charlotte J. Houldcroft
- Department of Medicine, Addenbrookes Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Sarah E. Jackson
- Department of Medicine, Addenbrookes Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Eleanor Y. Lim
- Department of Medicine, Addenbrookes Hospital, University of Cambridge, Cambridge, United Kingdom
| | - George X. Sedikides
- Department of Medicine, Addenbrookes Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Emma L. Davies
- Department of Medicine, Addenbrookes Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Claire Atkinson
- Division of Infection and Immunity, Institute for Immunity and Transplantation, University College London, London, United Kingdom
| | - Megan McIntosh
- Division of Infection and Immunity, Institute for Immunity and Transplantation, University College London, London, United Kingdom
| | - Ester B. M. Remmerswaal
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Renal Transplant Unit, Division of Internal Medicine, Academic Medical Centre, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Georgina Okecha
- Department of Medicine, Addenbrookes Hospital, University of Cambridge, Cambridge, United Kingdom
| | - Frederike J. Bemelman
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
- Renal Transplant Unit, Division of Internal Medicine, Academic Medical Centre, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Richard J. Stanton
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Matthew Reeves
- Division of Infection and Immunity, Institute for Immunity and Transplantation, University College London, London, United Kingdom
| | - Mark R. Wills
- Department of Medicine, Addenbrookes Hospital, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
11
|
Suárez NM, Blyth E, Li K, Ganzenmueller T, Camiolo S, Avdic S, Withers B, Linnenweber-Held S, Gwinner W, Dhingra A, Heim A, Schulz TF, Gunson R, Gottlieb D, Slobedman B, Davison AJ. Whole-Genome Approach to Assessing Human Cytomegalovirus Dynamics in Transplant Patients Undergoing Antiviral Therapy. Front Cell Infect Microbiol 2020; 10:267. [PMID: 32612959 PMCID: PMC7308726 DOI: 10.3389/fcimb.2020.00267] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 05/05/2020] [Indexed: 12/16/2022] Open
Abstract
Human cytomegalovirus (HCMV) is the most frequent cause of opportunistic viral infection following transplantation. Viral factors of potential clinical importance include the selection of mutants resistant to antiviral drugs and the occurrence of infections involving multiple HCMV strains. These factors are typically addressed by analyzing relevant HCMV genes by PCR and Sanger sequencing, which involves independent assays of limited sensitivity. To assess the dynamics of viral populations with high sensitivity, we applied high-throughput sequencing coupled with HCMV-adapted target enrichment to samples collected longitudinally from 11 transplant recipients (solid organ, n = 9, and allogeneic hematopoietic stem cell, n = 2). Only the latter presented multiple-strain infections. Four cases presented resistance mutations (n = 6), two (A594V and L595S) at high (100%) and four (V715M, V781I, A809V, and T838A) at low (<25%) frequency. One allogeneic hematopoietic stem cell transplant recipient presented up to four resistance mutations, each at low frequency. The use of high-throughput sequencing to monitor mutations and strain composition in people at risk of HCMV disease is of potential value in helping clinicians implement the most appropriate therapy.
Collapse
Affiliation(s)
- Nicolás M Suárez
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Emily Blyth
- Westmead Institute for Medical Research, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Blood and Bone Marrow Transplant Unit, Westmead Hospital, Sydney, NSW, Australia.,Sydney Cellular Therapies Laboratory, Westmead Hospital, Sydney, NSW, Australia
| | - Kathy Li
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Tina Ganzenmueller
- Institute for Medical Virology, University Hospital Tübingen, Tübingen, Germany.,Institute of Virology, Hannover Medical School, Hanover, Germany
| | - Salvatore Camiolo
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Selmir Avdic
- Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Barbara Withers
- Westmead Institute for Medical Research, Sydney, NSW, Australia
| | - Silvia Linnenweber-Held
- Department of Nephrology, Hannover Medical School, Hanover, Germany.,Public Health Agency of Lower Saxony, Hanover, Germany
| | - Wilfried Gwinner
- Department of Nephrology, Hannover Medical School, Hanover, Germany
| | - Akshay Dhingra
- Institute of Virology, Hannover Medical School, Hanover, Germany
| | - Albert Heim
- Institute of Virology, Hannover Medical School, Hanover, Germany
| | - Thomas F Schulz
- Institute of Virology, Hannover Medical School, Hanover, Germany.,German Center for Infection Research, Hanover, Germany
| | - Rory Gunson
- West of Scotland Specialist Virology Centre, Glasgow Royal Infirmary, Glasgow, United Kingdom
| | - David Gottlieb
- Westmead Institute for Medical Research, Sydney, NSW, Australia.,Blood and Bone Marrow Transplant Unit, Westmead Hospital, Sydney, NSW, Australia
| | - Barry Slobedman
- Discipline of Infectious Diseases and Immunology, Faculty of Medicine and Health, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Andrew J Davison
- MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| |
Collapse
|
12
|
Cagliani R, Forni D, Mozzi A, Sironi M. Evolution and Genetic Diversity of Primate Cytomegaloviruses. Microorganisms 2020; 8:E624. [PMID: 32344906 PMCID: PMC7285053 DOI: 10.3390/microorganisms8050624] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/17/2020] [Accepted: 04/19/2020] [Indexed: 12/30/2022] Open
Abstract
Cytomegaloviruses (CMVs) infect many mammals, including humans and non-human primates (NHPs). Human cytomegalovirus (HCMV) is an important opportunistic pathogen among immunocompromised patients and represents the most common infectious cause of birth defects. HCMV possesses a large genome and very high genetic diversity. NHP-infecting CMVs share with HCMV a similar genomic organization and coding content, as well as the course of viral infection. Recent technological advances have allowed the sequencing of several HCMV strains from clinical samples and provided insight into the diversity of NHP-infecting CMVs. The emerging picture indicates that, with the exclusion of core genes (genes that have orthologs in all herpesviruses), CMV genomes are relatively plastic and diverse in terms of gene content, both at the inter- and at the intra-species level. Such variability most likely underlies the strict species-specificity of these viruses, as well as their ability to persist lifelong and with relatively little damage to their hosts. However, core genes, despite their strong conservation, also represented a target of adaptive evolution and subtle changes in their coding sequence contributed to CMV adaptation to different hosts. Indubitably, important knowledge gaps remain, the most relevant of which concerns the role of viral genetics in HCMV-associated human disease.
Collapse
Affiliation(s)
| | | | | | - Manuela Sironi
- Scientific Institute, IRCCS E. MEDEA, Bioinformatics, 23842 Bosisio Parini, Italy
| |
Collapse
|
13
|
White JL, Patel EU, Abraham AG, Grabowski MK, Arav-Boger R, Avery RK, Quinn TC, Tobian AAR. Prevalence, Magnitude, and Genotype Distribution of Urinary Cytomegalovirus (CMV) Shedding Among CMV-Seropositive Children and Adolescents in the United States. Open Forum Infect Dis 2019; 6:ofz272. [PMID: 31281866 PMCID: PMC6602884 DOI: 10.1093/ofid/ofz272] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 06/04/2019] [Indexed: 11/12/2022] Open
Abstract
Background There are limited nationally representative data on correlates of cytomegalovirus (CMV) shedding among children and adolescents. In addition, the genotype distribution of CMV infections has not been well characterized among general populations in the United States. Methods This study characterized urinary CMV shedding among CMV immunoglobulin G-positive 6- to 19-year-olds in the US household population using data from the 1999-2004 National Health and Nutrition Examination Survey (NHANES). Multivariable Poisson regression was used to estimate adjusted prevalence ratios (aPR) and corresponding 95% confidence intervals (CIs). Analyses were weighted and multiple imputation was performed to handle missing data (with the exception of CMV genotypes). Results Prevalence of urinary CMV shedding was significantly lower among 9- to 11-year-olds (20.6%; aPR = 0.61; 95% CI, 0.44-0.83) and 12- to 19-year-olds (7.0%; aPR = 0.21; 95% CI, 0.14-0.30) compared with 6- to 8-year-olds (34.4%). Among CMV shedders, the youngest age group also had the highest urinary CMV viral loads. The prevalence of urinary CMV shedding among obese individuals was significantly lower compared with lean individuals (aPR = 0.68; 95% CI, 0.47-0.99). Among CMV shedders, glycoprotein B (gB)1 (51%) was the most prevalent gB variant, followed by gB2 (29%), gB3 (21%), and gB4 (13%); glycoprotein H (gH)2 (60%) was more prevalent than gH1 (48%). Multiple (≥2) gB (14%) and multiple gH (7%) infections were detected among CMV shedders. Conclusions This study underscores the importance of young children even above the age of 5 years as a potential source of CMV transmission. The detection of multiple CMV strains among CMV shedders may have implications for the transmission of viral diversity as well as vaccine development.
Collapse
Affiliation(s)
- Jodie L White
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Eshan U Patel
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alison G Abraham
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mary Kate Grabowski
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ravit Arav-Boger
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee
| | - Robin K Avery
- Department Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Thomas C Quinn
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Baltimore, Maryland
| | - Aaron A R Tobian
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
14
|
Abstract
Viruses, which are the most abundant biological entities on the planet, have been regarded as the "dark matter" of biology in the sense that despite their ubiquity and frequent presence in large numbers, their detection and analysis are not always straightforward. The majority of them are very small (falling under the limit of 0.5 μm), and collectively, they are extraordinarily diverse. In fact, the majority of the genetic diversity on the planet is found in the so-called virosphere, or the world of viruses. Furthermore, the most frequent viral agents of disease in humans display an RNA genome, and frequently evolve very fast, due to the fact that most of their polymerases are devoid of proofreading activity. Therefore, their detection, genetic characterization, and epidemiological surveillance are rather challenging. This review (part of the Curated Collection on Advances in Molecular Epidemiology of Infectious Diseases) describes many of the methods that, throughout the last few decades, have been used for viral detection and analysis. Despite the challenge of having to deal with high genetic diversity, the majority of these methods still depend on the amplification of viral genomic sequences, using sequence-specific or sequence-independent approaches, exploring thermal profiles or a single nucleic acid amplification temperature. Furthermore, viral populations, and especially those with RNA genomes, are not usually genetically uniform but encompass swarms of genetically related, though distinct, viral genomes known as viral quasispecies. Therefore, sequence analysis of viral amplicons needs to take this fact into consideration, as it constitutes a potential analytic problem. Possible technical approaches to deal with it are also described here. *This article is part of a curated collection.
Collapse
|
15
|
Human cytomegalovirus haplotype reconstruction reveals high diversity due to superinfection and evidence of within-host recombination. Proc Natl Acad Sci U S A 2019; 116:5693-5698. [PMID: 30819890 PMCID: PMC6431178 DOI: 10.1073/pnas.1818130116] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Human cytomegalovirus (HCMV) is a major global cause of congenital disability and transplant-related morbidity. Excessive levels of within-host HCMV nucleotide diversity are attributed to unexpectedly high mutation rates. Here, we show that high HCMV diversity is due to the frequent presence of mixed infections with genetically distinct strains, whereas HCMV in nonmixed infections is no more diverse than other DNA viruses. Using serial patient samples, we reconstruct viral strain haplotypes to pinpoint the timing of HCMV superinfections occurring within the study sampling time frame and uncover within-host viral recombination. From these results, we identify likely sources of infection and demonstrate probable selection for recombinant viruses. These results generate new, yet testable, insights into putative viral and host drivers of HCMV evolution and pathogenesis. Recent sequencing efforts have led to estimates of human cytomegalovirus (HCMV) genome-wide intrahost diversity that rival those of persistent RNA viruses [Renzette N, Bhattacharjee B, Jensen JD, Gibson L, Kowalik TF (2011) PLoS Pathog 7:e1001344]. Here, we deep sequence HCMV genomes recovered from single and longitudinally collected blood samples from immunocompromised children to show that the observations of high within-host HCMV nucleotide diversity are explained by the frequent occurrence of mixed infections caused by genetically distant strains. To confirm this finding, we reconstructed within-host viral haplotypes from short-read sequence data. We verify that within-host HCMV nucleotide diversity in unmixed infections is no greater than that of other DNA viruses analyzed by the same sequencing and bioinformatic methods and considerably less than that of human immunodeficiency and hepatitis C viruses. By resolving individual viral haplotypes within patients, we reconstruct the timing, likely origins, and natural history of superinfecting strains. We uncover evidence for within-host recombination between genetically distinct HCMV strains, observing the loss of the parental virus containing the nonrecombinant fragment. The data suggest selection for strains containing the recombinant fragment, generating testable hypotheses about HCMV evolution and pathogenesis. These results highlight that high HCMV diversity present in some samples is caused by coinfection with multiple distinct strains and provide reassurance that within the host diversity for single-strain HCMV infections is no greater than for other herpesviruses.
Collapse
|
16
|
Gao YL, Gao Z, He M, Liao P. Infection status of human parvovirus B19, cytomegalovirus and herpes simplex Virus-1/2 in women with first-trimester spontaneous abortions in Chongqing, China. Virol J 2018; 15:74. [PMID: 29688863 PMCID: PMC5913817 DOI: 10.1186/s12985-018-0988-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 04/17/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Infection with Parvovirus B19 (B19V), Cytomegalovirus (CMV) and Herpes Simplex Virus-1/2 (HSV-1/2) may cause fetal loses including spontaneous abortion, intrauterine fetal death and non-immune hydrops fetalis. Few comprehensive studies have investigated first-trimester spontaneous abortions caused by virus infections in Chongqing, China. Our study intends to investigate the infection of B19V, CMV and HSV-1/2 in first-trimester spontaneous abortions and the corresponding immune response. METHODS 100 abortion patients aged from 17 to 47 years were included in our study. The plasma samples (100) were analyzed qualitatively for specific IgG/IgM for B19V, CMV and HSV-1/2 (Virion\Serion, Germany) according to the manufacturer's recommendations. B19V, CMV and HSV-1/2 DNA were quantification by Real-Time PCR. RESULTS No specimens were positive for B19V, CMV, and HSV-1/2 DNA. By serology, 30.0%, 95.0%, 92.0% of patients were positive for B19V, CMV and HSV-1/2 IgG respectively, while 2% and 1% for B19V and HSV-1/2 IgM. CONCLUSION The low rate of virus DNA and a high proportion of CMV and HSV-1/2 IgG for most major of abortion patients in this study suggest that B19V, CMV and HSV-1/2 may not be the common factor leading to the spontaneous abortion of early pregnancy.
Collapse
Affiliation(s)
- Ya-Ling Gao
- Clinical Medical School, Southwest Medical University, Luzhou, 646000, China
| | - Zhan Gao
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Chengdu, 610052, China.,The Sichuan Blood Safety and Blood Substitute International Science and Technology Cooperation Base, Chengdu, 610052, China
| | - Miao He
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Chengdu, 610052, China. .,The Sichuan Blood Safety and Blood Substitute International Science and Technology Cooperation Base, Chengdu, 610052, China.
| | - Pu Liao
- The People's Hospital of Chongqing, Chongqing, 400000, China.
| |
Collapse
|
17
|
Cytomegalovirus induces HLA-class-II-restricted alloreactivity in an acute myeloid leukemia cell line. PLoS One 2018; 13:e0191482. [PMID: 29377903 PMCID: PMC5788343 DOI: 10.1371/journal.pone.0191482] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Accepted: 01/06/2018] [Indexed: 02/07/2023] Open
Abstract
Cytomegalovirus (HCMV) reactivation is found frequently after allogeneic hematopoietic stem cell transplantation (alloSCT) and is associated with an increased treatment-related mortality. Recent reports suggest a link between HCMV and a reduced risk of cancer progression in patients with acute leukemia or lymphoma after alloSCT. Here we show that HCMV can inhibit the proliferation of the acute myeloid leukemia cell line Kasumi-1 and the promyeloid leukemia cell line NB4. HCMV induced a significant up-regulation of HLA-class-II-molecules, especially HLA-DR expression and an increase of apoptosis, granzyme B, perforin and IFN-γ secretion in Kasumi-1 cells cocultured with peripheral blood mononuclear cells (PBMCs). Indolamin-2,3-dioxygenase on the other hand led only to a significant dose-dependent effect on IFN-γ secretion without effects on proliferation. The addition of CpG-rich oligonucleotides and ganciclovir reversed those antiproliferative effects. We conclude that HCMV can enhance alloreactivity of PBMCs against Kasumi-1 and NB4 cells in vitro. To determine if this phenomenon may be clinically relevant further investigations will be required.
Collapse
|
18
|
Human cytomegalovirus glycoprotein polymorphisms and increasing viral load in AIDS patients. PLoS One 2017; 12:e0176160. [PMID: 28467444 PMCID: PMC5415198 DOI: 10.1371/journal.pone.0176160] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 04/06/2017] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Multiple strains infection of human cytomegalovirus (HCMV) was found to be correlated with increased viral load in immunodeficient patients. However, the pathogenic mechanism underlying this correlation remains unclear. To evaluate genetic polymorphisms of HCMV glycoprotein and their potential role in its viral load, HCMV glycoprotein B, N, and O (gB, gN and gO) genotypes was studied in the population of HCMV infected acquired immune deficiency syndrome (AIDS) patients. The association between glycoprotein polymorphisms and HCMV viral load was analyzed. METHODS The genetic polymorphisms of glycoprotein from sera of 60 HCMV infected AIDS patients was investigated by multiplex nested PCR and sequencing. HCMV viral load was evaluated by quantitative PCR. RESULTS gB1, gO1a, and gN4a were the predominant glycoprotein genotypes in HCMV infected AIDS patients and composed 86.96%, 78.8%, and 49.2%, respectively. Only gN4a genotype infection significantly increased viral load (P = 0.048). 71% (43/60) of HCMV infected AIDS patients were found to carry multiple HCMV strains infection. A novel potential linkage of gO1a/gN4a was identified from multiple HCMV infected patients. It was the most frequent occurrence, accounted for 51.5% in 33 patients with gO and gN genotypes infection. Furthermore, the gO1a/gN4a linkage was correlated to an increased viral load (P = 0.020). CONCLUSION The gN4a correlates to higher level HCMV load in AIDS patients. Interestingly, a novel gO1a/gN4a linkage is identified from the patients with multiple HCMV strains infection and is also associated with an increased viral load. Therefore, the pathogenic mechanism underlying glycoprotein polymorphisms and interaction of variants should be analyzed further.
Collapse
|
19
|
Fishman JA. Infection in Organ Transplantation. Am J Transplant 2017; 17:856-879. [PMID: 28117944 DOI: 10.1111/ajt.14208] [Citation(s) in RCA: 465] [Impact Index Per Article: 66.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 01/09/2017] [Indexed: 01/25/2023]
Abstract
The prevention, diagnosis, and management of infectious disease in transplantation are major contributors to improved outcomes in organ transplantation. The risk of serious infections in organ recipients is determined by interactions between the patient's epidemiological exposures and net state of immune suppression. In organ recipients, there is a significant incidence of drug toxicity and a propensity for drug interactions with immunosuppressive agents used to maintain graft function. Thus, every effort must be made to establish specific microbiologic diagnoses to optimize therapy. A timeline can be created to develop a differential diagnosis of infection in transplantation based on common patterns of infectious exposures, immunosuppressive management, and antimicrobial prophylaxis. Application of quantitative molecular microbial assays and advanced antimicrobial therapies have advanced care. Pathogen-specific immunity, genetic polymorphisms in immune responses, and dynamic interactions between the microbiome and the risk of infection are beginning to be explored. The role of infection in the stimulation of alloimmune responses awaits further definition. Major hurdles include the shifting worldwide epidemiology of infections, increasing antimicrobial resistance, suboptimal assays for the microbiologic screening of organ donors, and virus-associated malignancies. Transplant infectious disease remains a key to the clinical and scientific investigation of organ transplantation.
Collapse
Affiliation(s)
- J A Fishman
- Transplant Infectious Disease and Immunocompromised Host Program and MGH Transplant Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
20
|
Current Perspectives on Cytomegalovirus in Heart Transplantation. CURRENT TRANSPLANTATION REPORTS 2016. [DOI: 10.1007/s40472-016-0121-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
21
|
Kaminski H, Fishman JA. The Cell Biology of Cytomegalovirus: Implications for Transplantation. Am J Transplant 2016; 16:2254-69. [PMID: 26991039 DOI: 10.1111/ajt.13791] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 02/17/2016] [Accepted: 03/07/2016] [Indexed: 01/25/2023]
Abstract
Interpretation of clinical data regarding the impact of cytomegalovirus (CMV) infection on allograft function is complicated by the diversity of viral strains and substantial variability of cellular receptors and viral gene expression in different tissues. Variation also exists in nonspecific (monocytes and dendritic cells) and specific (NK cells, antibodies) responses that augment T cell antiviral activities. Innate immune signaling pathways and expanded pools of memory NK cells and γδ T cells also serve to amplify host responses to infection. The clinical impact of specific memory T cell anti-CMV responses that cross-react with graft antigens and alloantigens is uncertain but appears to contribute to graft injury and to the abrogation of allograft tolerance. These responses are modified by diverse immunosuppressive regimens and by underlying host immune deficits. The impact of CMV infection on the transplant recipient reflects cellular changes and corresponding host responses, the convergence of which has been termed the "indirect effects" of CMV infection. Future studies will clarify interactions between CMV infection and allograft injury and will guide interventions that may enhance clinical outcomes in transplantation.
Collapse
Affiliation(s)
- H Kaminski
- Kidney Transplant Unit, CHU Bordeaux Pellegrin, Place Raba Léon, Bordeaux, France
| | - J A Fishman
- Transplant Infectious Disease and Immunocompromised Host Program and MGH Transplant Center, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
22
|
Mujtaba G, Khurshid A, Sharif S, Alam MM, Aamir UB, Shaukat S, Angez M, Rana MS, Umair M, Shah AA, Zaidi SSZ. Distribution of Cytomegalovirus Genotypes among Neonates Born to Infected Mothers in Islamabad, Pakistan. PLoS One 2016; 11:e0156049. [PMID: 27367049 PMCID: PMC4930188 DOI: 10.1371/journal.pone.0156049] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 05/09/2016] [Indexed: 01/27/2023] Open
Abstract
Background Congenital cytomegalovirus (cCMV) infection contributes to considerable long-term sequelae in neonates and children all over the world. The association between viral genotypes and severity of clinical cytomegalovirus (CMV) infection is yet to be defined. The objective of this study was to find the impact of active CMV infection during pregnancy and the clinical significance of genotypes in neonates with congenital cytomegalovirus infections in Pakistan. Methods A total of 409 blood samples from pregnant women seeking health care services at the two antenatal hospitals of Islamabad during January to December 2012 were tested by ELISA and nested-PCR. Pregnant women with active infection (detected as IgM positive, PCR positive or positive on both assays) were followed until delivery, to detect the outcome of overt cCMV infection in neonates. Genetic characterization of CMV strains was performed by sequence analysis of envelope glycoproteins: gB, gN and gH to detect the contributing CMV genotypes. Results The seroprevalence of anti-CMV IgG and IgM was 97.5% (399 out of 409) and 12.7% (52 out of 409), respectively, while 20% (82/409) pregnant women were found positive for CMV DNA by PCR. Logistic regression analysis showed a significant association of active infection with parity [OR = 2.56, 95% CI = 1.82–2.62, p = 0.04], febrile illness [OR = 1.84, 95% CI = 1.76–3.65, p = 0.01] and jaundice [OR = 22.5, 95% CI = 4.53–85.02, p = 0.002]. We were able to isolate virus in 41 out of 70 neonates; 36.6% (15 out of 41) of them were symptomatic at birth while 63.4% (26 out of 41) were asymptomatic. The most prominent clinical feature observed in symptomatic neonates was hepatosplenomegaly (26.6%; 4 out of 15). All three genotypes gB, gN and gH were found with the highest frequency of gB1 genotype, found in 75% infants with hepatic damage. Phylogenetic analysis of Pakistani strains showed 96%-100% homology to their prototype strains. Conclusions Active CMV infection during pregnancy is a major cause of congenital CMV infection with comparable distribution of all three genotypes: gB, gN and gH in symptomatic and asymptomatic neonates. Our findings emphasize to conduct a comprehensive large scale survey and introduction of country wide routine screening at maternity clinics for early diagnosis of CMV to reduce its associated devastating outcomes.
Collapse
Affiliation(s)
- Ghulam Mujtaba
- Department of Microbiology, Quaid-i-Azam University, Islamabad, Pakistan
| | - Adnan Khurshid
- Department of Virology, National Institute of Health, Islamabad, Pakistan
| | - Salmaan Sharif
- Department of Virology, National Institute of Health, Islamabad, Pakistan
| | | | - Uzma Bashir Aamir
- Department of Virology, National Institute of Health, Islamabad, Pakistan
| | - Shahzad Shaukat
- Department of Virology, National Institute of Health, Islamabad, Pakistan
| | - Mehar Angez
- Department of Virology, National Institute of Health, Islamabad, Pakistan
| | | | - Massab Umair
- Department of Virology, National Institute of Health, Islamabad, Pakistan
| | - Aamer Ali Shah
- Department of Microbiology, Quaid-i-Azam University, Islamabad, Pakistan
| | | |
Collapse
|
23
|
Åsberg A, Humar A, Rollag H, Jardine AG, Kumar D, Aukrust P, Ueland T, Bignamini AA, Hartmann A. Lessons Learned From a Randomized Study of Oral Valganciclovir Versus Parenteral Ganciclovir Treatment of Cytomegalovirus Disease in Solid Organ Transplant Recipients: The VICTOR Trial. Clin Infect Dis 2016; 62:1154-60. [DOI: 10.1093/cid/ciw084] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 02/09/2016] [Indexed: 12/28/2022] Open
|
24
|
Görzer I, Trajanoski S, Popow-Kraupp T, Puchhammer-Stöckl E. Analysis of human cytomegalovirus strain populations in urine samples of newborns by ultra deep sequencing. J Clin Virol 2015; 73:101-104. [DOI: 10.1016/j.jcv.2015.11.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 10/30/2015] [Accepted: 11/01/2015] [Indexed: 11/26/2022]
|
25
|
Lisboa LF, Egli A, O'Shea D, Åsberg A, Hartmann A, Rollag H, Pang XL, Tyrrell DL, Kumar D, Humar A. Hcmv-miR-UL22A-5p: A Biomarker in Transplantation With Broad Impact on Host Gene Expression and Potential Immunological Implications. Am J Transplant 2015; 15:1893-902. [PMID: 25833298 DOI: 10.1111/ajt.13222] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Revised: 12/28/2014] [Accepted: 01/14/2015] [Indexed: 01/25/2023]
Abstract
Cytomegalovirus (CMV) encodes multiple microRNAs. While these have been partially characterized in vitro, their relevance to clinical CMV infection has not been evaluated. We analyzed samples from a cohort of solid organ transplant patients with CMV disease (n = 245) for viral microRNA expression. Several CMV microRNAs were readily detectable in patients with CMV disease in variable relative abundance. Expression level generally correlated with DNA viral load and the absence of viral microRNA was associated with faster viral clearance. Detection of hcmv-miR-UL22A-5p at baseline independently predicted the recurrence of CMV viremia upon discontinuation of antiviral therapy (OR 3.024, 95% CI: 1.35-6.8; p = 0.007). A combination of direct mRNA targeting by the microRNA and indirect modulation of gene expression involving isoforms of the transcriptional regulator C-MYC may be responsible for the broad effects seen in the association of gene transcripts with the RNA-induced silencing complex and in global protein expression upon hcmv-miR-UL22A-5p transfection. This novel study of in vivo viral microRNA expression profiles provides unique insight into the complexity of clinical CMV infection following transplantation. We provide evidence that viral microRNAs may have complex effects on gene expression and be associated with specific virologic and clinical outcomes, and thus could be further evaluated as biomarkers.
Collapse
Affiliation(s)
- L F Lisboa
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - A Egli
- Infection Biology Lab, Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - D O'Shea
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - A Åsberg
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway.,Department of Transplant Medicine, Section for Nephrology, Oslo University Hospital Rikshospitalet, University of Oslo, Oslo, Norway
| | - A Hartmann
- Department of Transplant Medicine, Section for Nephrology, Oslo University Hospital Rikshospitalet, University of Oslo, Oslo, Norway
| | - H Rollag
- Department of Microbiology, Oslo University Hospital Rikshospitalet , Oslo, and Faculty of Medicine, University of Oslo, Oslo, Norway
| | - X L Pang
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - D L Tyrrell
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - D Kumar
- Multi-organ Transplant Program, University Health Network, Toronto, Ontario, Canada
| | - A Humar
- Multi-organ Transplant Program, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
26
|
Zhou Y, Bian G, Zhou Q, Gao Z, Liao P, Liu Y, He M. Detection of cytomegalovirus, human parvovirus B19, and herpes simplex virus-1/2 in women with first-trimester spontaneous abortions. J Med Virol 2015; 87:1749-53. [DOI: 10.1002/jmv.24218] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2015] [Indexed: 11/11/2022]
Affiliation(s)
- Ya Zhou
- Institute of Blood Transfusion; Chinese Academy of Medical Sciences; Chengdu China
| | - Guohui Bian
- Institute of Blood Transfusion; Chinese Academy of Medical Sciences; Chengdu China
| | - Qiongxiu Zhou
- Institute of Blood Transfusion; Chinese Academy of Medical Sciences; Chengdu China
| | - Zhan Gao
- Institute of Blood Transfusion; Chinese Academy of Medical Sciences; Chengdu China
| | - Pu Liao
- The Third People's Hospital of Chongqing; Chongqing China
| | - Yu Liu
- Institute of Blood Transfusion; Chinese Academy of Medical Sciences; Chengdu China
| | - Miao He
- Institute of Blood Transfusion; Chinese Academy of Medical Sciences; Chengdu China
| |
Collapse
|
27
|
Taherkhani R, Farshadpour F, Makvandi M, Hamidifard M, Esmailizadeh M, Ahmadi B, Heidari H. Determination of cytomegalovirus prevalence and glycoprotein B genotypes among ulcerative colitis patients in ahvaz, iran. Jundishapur J Microbiol 2015; 8:e17458. [PMID: 25793098 PMCID: PMC4353060 DOI: 10.5812/jjm.17458] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 03/03/2014] [Accepted: 03/12/2014] [Indexed: 01/07/2023] Open
Abstract
Background: The human cytomegalovirus (HCMV) is a common pathogen which usually remains asymptomatic in the healthy adults; however, it can cause a symptomatic disease in the immunocompromised patients. The risk of infection with HCMV increases in ulcerative colitis (UC) patients as a result of receiving immunosuppressive agents. Objectives: This study aimed to determine the prevalence and the glycoprotein B genotypes of HCMV among the patients with HCMV disease superimposed on an UC flare that required hospitalization in Imam Khomeini Hospital in Ahvaz, Iran, during 2010- 2012. Patients and Methods: In this case-control study, formalin-fixed paraffin-embedded intestinal tissue samples were taken from 98 patients with UC disease including 53 males and 45 females (mean age ± standard deviation, 38.95 ± 17.93) and 67 control patients with noninflammatory disease who were referred to Imam Khomeini Hospital during 2010-2012. Detection of HCMV genome in intestinal samples was carried out by seminested polymerase chain reaction. Glycoprotein B genotypes were determined by sequencing. Results: Among 98 patients with UC, only 12 (12.2%) patients were positive for HCMV genome, while the HCMV genome was not detected in any of the controls. (P = 0.002). The distribution of HCMV gB genotypes in 12 CMV-positive UC patients was as follow: gB1, 11 (91.7%) and gB3, 1 (8.3%). The most prevalent genotype in CMV-positive UC patients was gB1. Conclusions: In this study, high prevalence of 91.7% HCMV gB1 genotype was predominant among HCMV-positive UC patients, which suggests that there might be an association between HCMV gB genotype 1 and UC disease.
Collapse
Affiliation(s)
- Reza Taherkhani
- Infectious and Tropical Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, IR Iran
- Department of Microbiology and Parasitology, Faculty of Medicine, Bushehr University of Medical Sciences, Bushehr, IR Iran
| | - Fatemeh Farshadpour
- Department of Microbiology and Parasitology, Faculty of Medicine, Bushehr University of Medical Sciences, Bushehr, IR Iran
| | - Manoochehr Makvandi
- Infectious and Tropical Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, IR Iran
- Corresponding author: Manoochehr Makvandi, Infectious and Tropical Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, IR Iran. Tel: +98-9166181683, Fax: +98-6113738313, E-mail:
| | - Mojtaba Hamidifard
- Infectious and Tropical Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, IR Iran
| | - Mahdi Esmailizadeh
- Department of Medical Physiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, IR Iran
| | - Bijan Ahmadi
- Internal Medicine Department, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, IR Iran
| | - Hamid Heidari
- Department of Medical Physiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, IR Iran
| |
Collapse
|
28
|
Paradowska E, Jabłońska A, Studzińska M, Kasztelewicz B, Zawilińska B, Wiśniewska-Ligier M, Dzierżanowska-Fangrat K, Woźniakowska-Gęsicka T, Kosz-Vnenchak M, Leśnikowski ZJ. Cytomegalovirus glycoprotein H genotype distribution and the relationship with hearing loss in children. J Med Virol 2014; 86:1421-7. [PMID: 24615599 DOI: 10.1002/jmv.23906] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2014] [Indexed: 01/29/2023]
Abstract
Cytomegalovirus (CMV) is a leading cause of congenital infection and a leading infectious cause of hearing loss in children. The ORF UL75 gene encodes envelope glycoprotein H (gH), which is essential for CMV entry into host cells and the target of the immune response in humans. However, the distribution of gH variants and the relationship between the viral genotype, viral load, and sequelae in children infected with CMV is debated. The UL75 genetic variation of CMV isolates from 42 newborns infected congenitally with CMV and 93 infants with postnatal or unproven congenital CMV infection was analyzed. Genotyping was performed by analysis of PCR-amplified fragments, and the viral load was measured by quantitative real-time PCR. There were no differences in the distribution of gH genotypes in the children infected congenitally and postnatally. Mixed-genotype infections with both gH1 and gH2 variants were detected in approximately 25% of the examined patients. No relationship between UL75 gene polymorphisms and the symptoms at birth was observed. The results suggest that the infection with gH2 genotype diminishes the risk of hearing loss in children (P = 0.010). In addition, sensorineural hearing loss was associated with CMV gH1 genotype infection in infants (P = 0.032) and a high viral load in urine (P = 0.005). In conclusion, it was found that the gH genotype does not predict clinical sequelae in newborn infants following congenital CMV infection. However, these results suggest that the gH genotype might be associated with hearing loss in children.
Collapse
Affiliation(s)
- Edyta Paradowska
- Laboratory of Molecular Virology and Biological Chemistry, Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Pillet S, Roblin X, Cornillon J, Mariat C, Pozzetto B. Quantification of cytomegalovirus viral load. Expert Rev Anti Infect Ther 2013; 12:193-210. [PMID: 24341395 DOI: 10.1586/14787210.2014.870887] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cytomegalovirus (CMV), a member of the Herpesviridae family, is worldwide distributed. After the primary infection, CMV induces a latent infection with possible reactivation(s). It is responsible for severe to life-threatening diseases in immunocompromised patients and in foetuses and newborns of infected mothers. For monitoring CMV load, classical techniques based on rapid culture or pp65 antigenemia are progressively replaced by quantitative nuclear acid tests (QNAT), easier to implement and standardize. A large variety of QNAT are available from laboratory-developed assays to fully-automated commercial tests. The indications of CMV quantification include CMV infection during pregnancy and in newborns, and viral surveillance of grafted and non-grafted immunocompromised patients, patients with bowel inflammatory diseases and those hospitalised in intensive care unit. A close cooperation between virologists and clinicians is essential for optimizing the benefit of CMV DNA monitoring.
Collapse
Affiliation(s)
- Sylvie Pillet
- Faculty of Medicine of Saint-Etienne, University of Lyon, Groupe Immunité des Muqueuses et Agents Pathogènes (GIMAP)-EA3064, 42023 Saint-Etienne, France
| | | | | | | | | |
Collapse
|
30
|
Copy numbers of telomeric repeat sequences of human herpesvirus 6B in clinical isolates: possibility of mixed infections. J Clin Microbiol 2013; 52:419-24. [PMID: 24478470 DOI: 10.1128/jcm.02192-13] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In order to determine whether mixed infections of human herpesvirus 6B (HHV-6B) occur in immunocompetent and immunocompromised individuals, we examined the copy numbers of telomeric repeat sequences (TRS) of clinical isolates. In clinical isolates obtained from patients with exanthem subitum caused by primary HHV-6B infection, PCR products with HHV-6B TRS ranging between 400 and 800 bp were amplified. PCR products of various sizes were amplified in four clinical isolates from drug-induced hypersensitivity syndrome (DIHS) patients and 15 isolates from hematopoietic stem cell transplant (HSCT) recipients with HHV-6B reactivation. Based on the sequence analysis of the PCR products, the copy numbers of TRS in DIHS and HSCT patients were between 42 and 82 and 22 and >90, respectively. For two of the HSCT recipients, HHV-6B TRS PCR products of different sizes were detected in several isolates from each patient, which suggests mixed HHV-6B infections. In two of the posttransplant HHV-6B encephalitis patients, the sizes of the TRS nested PCR products amplified from the reactivated virus detected in the central nervous system differed from those of the virus detected in initial isolates from peripheral blood mononuclear cells. Taken together, these results suggest that PCR analysis of TRS copy number is a reliable tool for the discrimination of HHV-6B clinical isolates. Additionally, mixed HHV-6B infections occurred in HSCT recipients, and in some cases, compartmentalization of the HHV-6B strains to the central nervous system versus the blood compartment occurred in posttransplant HHV-6B encephalitis patients.
Collapse
|
31
|
Genotypic diversity and mixed infection in newborn disease and hearing loss in congenital cytomegalovirus infection. Pediatr Infect Dis J 2013; 32:1050-4. [PMID: 23694837 PMCID: PMC3785554 DOI: 10.1097/inf.0b013e31829bb0b9] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Congenital cytomegalovirus (cCMV) is a common congenital infection and a leading nongenetic cause of sensorineural hearing loss (SNHL). CMV exhibits extensive genetic variability, and infection with multiple CMV strains (mixed infection) was shown to be common in congenital CMV. The role of mixed infections in disease and outcome remains to be defined. METHODS Genotyping of envelope glycoproteins, UL55 (gB), UL73 (gN) and UL75 (gH), was performed on saliva specimens of 79 infants from the ongoing CMV and Hearing Multicenter Screening (CHIMES) Study and on blood and urine specimens of 52 infants who participated in natural history studies at the University of Alabama at Birmingham. Genotyping of UL144 and US28 was also performed in the CHIMES cohort. The association of individual genotypes and mixed infection with clinical findings at birth and SNHL was examined. RESULTS Thirty-seven of 131 infants (28%) were symptomatic at birth and 26 (20%) had SNHL at birth. All known genotypes of UL55, UL75, UL73 and US28 were represented, and no particular genotype was associated with symptomatic infection or SNHL. UL144 subtype C was more common in symptomatic infants but not associated with SNHL. Mixed infection was observed in 59 infants (45%) and not associated with symptoms (P = 0.43) or SNHL at birth (P = 0.82). In the cohort of 52 infants with long-term hearing outcome, mixed infection at birth was not predictive of SNHL. CONCLUSIONS Mixed infection is common in infants with congenital CMV but is neither associated with symptomatic infection nor associated with SNHL.
Collapse
|