1
|
Akingbola A, Adegbesan A, Adewole O, Adegoke K, Benson AE, Jombo PA, Uchechukwu Eboson S, Oluwasola V, Aiyenuro A. The mRNA-1647 vaccine: A promising step toward the prevention of cytomegalovirus infection (CMV). Hum Vaccin Immunother 2025; 21:2450045. [PMID: 39825496 DOI: 10.1080/21645515.2025.2450045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/14/2024] [Accepted: 01/02/2025] [Indexed: 01/30/2025] Open
Abstract
Cytomegalovirus (CMV) is a leading cause of congenital infections and significant health complications in immunocompromised individuals. With no licensed CMV vaccine available, the development of the mRNA-1647 offers promising advancements in CMV prevention. We have reviewed results from Phase 1 and 2 clinical trials of the mRNA-1647 vaccine, demonstrating robust immune responses in both seronegative and seropositive participants. Vaccines exhibited significantly elevated neutralizing antibody titers against CMV, particularly in fibroblast and epithelial cells, with sustained responses lasting up to 18 months post-vaccination. The mRNA-1647 vaccine triggered strong T-cell and memory B-cell responses, suggesting its potential for long-term protection against CMV infection. The ongoing Phase 3 CMVictory trial evaluates the safety and immunogenicity of mRNA-1647 in women of childbearing age, with preliminary data showing promise in preventing congenital CMV transmission. This vaccine could significantly reduce CMV-related morbidity and mortality, particularly in newborns and immunocompromised individuals, addressing a critical unmet medical need.
Collapse
Affiliation(s)
| | - Abiodun Adegbesan
- African Cancer Institute, Department of Global Health, Stellenbosch University, Cape Town, South Africa
| | | | - Kolade Adegoke
- Faculty of Clinical Sciences, Obafemi Awolowo University Ile-Ife, Osun State,Nigeria
| | | | - Paul Ayomide Jombo
- Internal Medicine, Basildon and Thurrock University Hospitals NHS Foundation Trust: Basildon SS165NL, England, Essex, England, UK
| | | | - Victor Oluwasola
- Babcock University Teaching Hospital, Ilishan-Remo, Ogun State, Nigeria
| | - Ademola Aiyenuro
- Division of Virology, Department of Pathology, University of Cambridge, England, UK
| |
Collapse
|
2
|
Trier NH, Zivlaei N, Ostrowski SR, Sørensen E, Larsen M, Slibinskas R, Ciplys E, Frederiksen JL, Houen G. Virus-specific antibody responses in severe acute respiratory syndrome coronavirus 2-infected and vaccinated individuals. Immunol Lett 2025; 274:107004. [PMID: 40157431 DOI: 10.1016/j.imlet.2025.107004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 03/06/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection can have a serious course with many complications, especially in immunocompromised individuals. In such persons, other latent virus infections may contribute to disease pathology, in particular viruses which infect immune cells such as Epstein-Barr virus (EBV) and cytomegalovirus (CMV). METHODS In this study, serology-based assays were conducted to analyse antibody responses to SARS-CoV-2 spike protein (SP), EBV Epstein-Barr nuclear antigen (EBNA)-1 and CMV phosphoprotein (pp)52 in naturally SARS-CoV-2-infected individuals, non-infected healthy controls (HCs) and vaccinated healthy controls (VHCs) to identify an association between SARS-CoV-2 antibodies and EBV and CMV antibodies in order to determine whether latent EBV and CMV infected individuals are more prone to become infected with SARS-CoV-2. Moreover, SARS-CoV-2, EBV, and CMV antibody responses were characterized in serum from patients with relapsing-remitting multiple sclerosis (RRMS), a chronic inflammatory disease strongly associated with EBV infections, to determine whether the serologic virus antibody profile varies in immunocompromised RRMS individuals upon SARS-CoV-2 vaccinations compared to VHCs. RESULTS Significantly elevated SP IgG, IgM and IgA levels were identified in SARS-CoV-2-infected immunocompetent individuals when compared to non-infected HCs. However, no correlation was found to serum antibodies between SARS-CoV-2, EBV, and CMV in individuals infected with SARS-CoV-2 and in VHCs, suggesting that latent infections with neither EBV nor CMV associates to SARS-CoV-2 infection. Moreover, no significant difference in SP IgG, IgA and IgM levels was observed between vaccinated RRMS patients and VHCs, indicating that the immune system of immune deficient RRMS patients and VHCs respond identical to SARS-CoV-2 vaccinations. CONCLUSION Collectively, SARS-CoV-2 SP antibody levels reflect the vaccination and infection history and do not associate with EBV and CMV serostatus.
Collapse
Affiliation(s)
- Nicole Hartwig Trier
- Department of Neurology, Rigshospitalet Glostrup, Valdemar Hansens vej 13, Glostrup, Denmark.
| | - Nadia Zivlaei
- Department of Neurology, Rigshospitalet Glostrup, Valdemar Hansens vej 13, Glostrup, Denmark.
| | - Sisse Rye Ostrowski
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen OE, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, BLegdamsvej 3B, 2200 Copenhagen N, Denmark.
| | - Erik Sørensen
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen OE, Denmark.
| | - Margit Larsen
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, 2100 Copenhagen OE, Denmark.
| | - Rimantas Slibinskas
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Sauletekio av. 7, LT-10257 Vilnius, Lithuania.
| | - Evaldas Ciplys
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Sauletekio av. 7, LT-10257 Vilnius, Lithuania.
| | - Jette Lautrup Frederiksen
- Department of Neurology, Rigshospitalet Glostrup, Valdemar Hansens vej 13, Glostrup, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, BLegdamsvej 3B, 2200 Copenhagen N, Denmark.
| | - Gunnar Houen
- Department of Neurology, Rigshospitalet Glostrup, Valdemar Hansens vej 13, Glostrup, Denmark; Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55 5230 Odense M, Denmark.
| |
Collapse
|
3
|
Karthigeyan KP, Connors M, Binuya CR, Gross M, Fuller AS, Crooks CM, Wang HY, Sponholtz MR, Byrne PO, Herbek S, Andy C, Gerber LM, Campbell JD, Williams CA, Mitchell E, van der Maas L, Miller I, Yu D, Bottomley MJ, McLellan JS, Permar SR. A human cytomegalovirus prefusion-like glycoprotein B subunit vaccine elicits humoral immunity similar to that of postfusion gB in mice. J Virol 2025; 99:e0217824. [PMID: 40338082 DOI: 10.1128/jvi.02178-24] [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: 12/09/2024] [Accepted: 04/06/2025] [Indexed: 05/09/2025] Open
Abstract
Human cytomegalovirus (HCMV) is the leading infectious cause of birth defects. Despite the global disease burden, there is no Food and Drug Administration (FDA)-approved HCMV vaccine. The most efficacious HCMV vaccine candidates to date have used glycoprotein B (gB), a class III viral fusion protein, in its postfusion form. While some viral fusion proteins have been shown to elicit stronger neutralizing responses in their prefusion conformation, HCMV prefusion-like and postfusion gB were recently shown to elicit antibodies with similar fibroblast neutralization titers in mice. We aimed to define and compare the specificity and functionality of plasma IgG elicited by distinct prefusion-like and postfusion gB constructs. Prefusion-like and postfusion gB elicited comparable IgG responses that predominantly mapped to the AD-5 antigenic domain known to elicit neutralizing antibodies. Interestingly, postfusion gB elicited significantly higher plasma IgG binding to cell-associated gB and antibody-dependent cellular phagocytosis than that of prefusion-like gB. The vaccines elicited comparable neutralization titers of heterologous HCMV strain AD169r in fibroblasts; however, neither elicited neutralizing titers against the vaccine-matched strain Towne in fibroblasts. Our data indicate that gB in this prefusion-like conformation elicits similar specificity and functional humoral immunity to that of postfusion gB, unlike certain class I viral fusion proteins that have been used as vaccine antigens. These findings deepen our understanding of the immune response elicited by class III fusion proteins and may inform further design and testing of conformationally dependent herpesvirus glycoprotein vaccine candidates.IMPORTANCEVaccines against human cytomegalovirus (HCMV) still remain elusive in spite of the high disease burden of the virus, especially in pre-term infants and immunocompromised individuals. While vaccine efforts have focused on vaccine-induced antibodies to neutralize the virus, studies have increasingly shown the importance of other antibody functions in protection against cytomegalovirus (CMV) transmission. In this study, we comprehensively evaluated immune responses elicited by the prefusion state of an important HCMV protein called glycoprotein B (gB) in mice. Our results indicate that prefusion gB elicits immune responses similar to that of postfusion gB in mice and reveals areas for further redesign and testing for prefusion vaccine antigens against CMV and other herpesviruses, which could help in furthering vaccine development against HCMV.
Collapse
Affiliation(s)
| | - Megan Connors
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Christian R Binuya
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Mackensie Gross
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Adelaide S Fuller
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Chelsea M Crooks
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Hsuan-Yuan Wang
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Madeline R Sponholtz
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA
| | - Patrick O Byrne
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA
| | - Savannah Herbek
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Caroline Andy
- Department of Population Health Sciences, Weill Cornell Medicine, New York, New York, USA
| | - Linda M Gerber
- Department of Population Health Sciences, Weill Cornell Medicine, New York, New York, USA
| | - John D Campbell
- Dynavax Technologies Corporation, Emeryville, California, USA
| | - Caitlin A Williams
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Elizabeth Mitchell
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Lara van der Maas
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Itzayana Miller
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Dong Yu
- Dynavax Technologies Corporation, Emeryville, California, USA
| | | | - Jason S McLellan
- Department of Molecular Biosciences, University of Texas at Austin, Austin, Texas, USA
| | - Sallie R Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
4
|
Hayashino K, Seike K, Masunari T, Hashida R, Oka S, Fujiwara Y, Terao T, Kitamura W, Kobayashi H, Kamoi C, Kondo T, Fujiwara H, Asada N, Ennishi D, Fujii K, Fujii N, Maeda Y. Cytomegalovirus reactivation in patients with large B-cell lymphoma treated with chimeric antigen receptor T-cell therapy. Int J Hematol 2025:10.1007/s12185-025-04023-y. [PMID: 40526215 DOI: 10.1007/s12185-025-04023-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 05/28/2025] [Accepted: 05/28/2025] [Indexed: 06/19/2025]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has improved outcomes of relapsed and/or refractory large B-cell lymphoma (r/r LBCL). However, its off-tumor effects result in severe prolonged humoral immune deficiency. Cytomegalovirus (CMV) is a latent virus that can be life-threatening in immunosuppressed patients. In the setting of CAR T-cell therapy, Asian race is a risk factor for clinically significant CMV infection. However, the effect of CAR T-cell therapy on CMV reactivation in Japanese patients remains unclear. Previous reports used polymerase chain reaction (PCR), but we used the pp65 antigenemia assay to retrospectively investigate long-term effects in patients with r/r LBCL. The study included 46 patients. Nine (19.6%) developed CMV reactivation, with a median onset of 13 days. Six of these patients received preemptive therapy, and none developed CMV end-organ disease. Primary refractory disease, grade 2-4 cytokine release syndrome, and high-dose corticosteroids were risk factors for CMV reactivation. Long-term follow-up showed that CMV reactivation rarely occurred later than 28 days post-infusion. Our study using the pp65 antigenemia assay showed a similar incidence of CMV reactivation, onset, and risk factors to those in the previous reports using PCR.
Collapse
Affiliation(s)
- Kenta Hayashino
- Department of Hematology and Oncology, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, Okayama, 700-8558, Japan
- Department of Hematology, Oncology and Respiratory Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Keisuke Seike
- Department of Hematology and Oncology, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, Okayama, 700-8558, Japan.
| | - Taro Masunari
- Department of Hematology, Chugoku Central Hospital, 148-13, Kamiiwanari, Miyuki-cho, Fukuyama, 720-0001, Japan
| | - Risa Hashida
- Division of Hematology, Ehime Prefectural Central Hospital, 83, Kasuga-machi, Matsuyama, 790-0024, Japan
| | - Satoshi Oka
- Department of Hematology and Blood Transfusion, Kochi Health Science Center, Ike, Kochi, 2125-1781-8555, Japan
| | - Yuki Fujiwara
- Department of Hematology and Oncology, Japanese Red Cross Society Himeji Hospital, 1-12-1, Shimoteno, Himeji, 670-8540, Japan
| | - Toshiki Terao
- Department of Hematology and Oncology, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, Okayama, 700-8558, Japan
- Department of Hematology, Oncology and Respiratory Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Wataru Kitamura
- Department of Hematology and Oncology, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, Okayama, 700-8558, Japan
- Department of Hematology, Oncology and Respiratory Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Division of Transfusion and Cell Therapy, Okayama University Hospital, 2-5-1 Shikata, OkayamaOkayama-shi, Japan
| | - Hiroki Kobayashi
- Department of Hematology and Oncology, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, Okayama, 700-8558, Japan
- Department of Hematology and Oncology, Japanese Red Cross Society Himeji Hospital, 1-12-1, Shimoteno, Himeji, 670-8540, Japan
| | - Chihiro Kamoi
- Department of Hematology and Oncology, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, Okayama, 700-8558, Japan
- Division of Transfusion and Cell Therapy, Okayama University Hospital, 2-5-1 Shikata, OkayamaOkayama-shi, Japan
| | - Takumi Kondo
- Department of Hematology and Oncology, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, Okayama, 700-8558, Japan
| | - Hideaki Fujiwara
- Department of Hematology and Oncology, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, Okayama, 700-8558, Japan
| | - Noboru Asada
- Department of Hematology and Oncology, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, Okayama, 700-8558, Japan
| | - Daisuke Ennishi
- Department of Hematology and Oncology, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, Okayama, 700-8558, Japan
- Center for Comprehensive Genomic Medicine, Okayama University Hospital, 2-5-1 Shikata, Okayama-shi, Okayama, Japan
| | - Keiko Fujii
- Department of Hematology and Oncology, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, Okayama, 700-8558, Japan
- Division of Clinical Laboratory, Okayama University Hospital, Okayama, 2-5-1 Shikata, Okayama-shi, Japan
| | - Nobuharu Fujii
- Department of Hematology and Oncology, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, Okayama, 700-8558, Japan
- Division of Transfusion and Cell Therapy, Okayama University Hospital, 2-5-1 Shikata, OkayamaOkayama-shi, Japan
| | - Yoshinobu Maeda
- Department of Hematology and Oncology, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, Okayama, 700-8558, Japan
- Department of Hematology, Oncology and Respiratory Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
5
|
Naeini MK, Freidin MB, Smith IG, Ward S, Williams FMK. Evidence for a Causal Association Between Human Cytomegalovirus Infection and Chronic Back Pain: A One-Sample Mendelian Randomization Study. JOR Spine 2025; 8:e70063. [PMID: 40201537 PMCID: PMC11977176 DOI: 10.1002/jsp2.70063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 03/03/2025] [Accepted: 03/23/2025] [Indexed: 04/10/2025] Open
Abstract
Background Chronic back pain (CBP) is a major cause of disability globally. While its etiology is multifactorial, specific contributing genetic and environmental factors remain to be discovered. Paraspinal muscle fat has been shown in human and preclinical studies to be related to CBP. One potential risk factor is infection by cytomegalovirus (CMV) because CMV is trophic for fat. CMV may reside in the paraspinal muscle adipose tissue. We set out to test the hypothesis that previous CMV infection is linked to CPB using a one-sample Mendelian randomization (MR). Method The sample comprised 5140 UK Biobank participants with information about CMV serology and CBP status. A one-sample MR based on independent genetic variants predicting CMV positivity was conducted in Northern European participants. To validate the association further, the MR study was repeated using a CMV polygenic risk score (PRS). As a negative control for confounding and spurious causal inference, we used Epstein-Barr virus (EBV) serology, because EBV is another common viral infection but is not trophic for adipose tissue. Results A genome-wide association study for CMV seropositivity revealed 86 independent SNPs having p-value < 2 × 10 - 4 that have been used to define genetically-predicted categories of CMV infection risk. The CMV predicted categories were found statistically significantly associated with CBP (OR = 1.150; 95% CI: 1.005-1.317, p-value = 0.043). Stronger significant results were obtained using the PRS for CMV seropositivity (OR = 1.290; 95% CI: 1.133-1.469, p-value = 12E-4). No such association was seen between EBV and CBP. Conclusion Our results provide evidence for a causal relationship between CMV infection and CBP. Further investigation is warranted to get insight into the mechanism by which CMV might contribute to the pathogenesis of CBP.
Collapse
Affiliation(s)
- Maryam Kazemi Naeini
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population SciencesKing's College LondonLondonUK
| | - Maxim B. Freidin
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population SciencesKing's College LondonLondonUK
| | - Isabelle Granville Smith
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population SciencesKing's College LondonLondonUK
| | - Stephen Ward
- Pain Management CentreGuy's and St Thomas' NHS Foundation TrustLondonUK
| | - Frances M. K. Williams
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population SciencesKing's College LondonLondonUK
| |
Collapse
|
6
|
Schultz EP, Ponsness L, Lanchy JM, Zehner M, Klein F, Ryckman BJ. Human cytomegalovirus gH/gL/gO binding to PDGFRα provides a regulatory signal activating the fusion protein gB that can be blocked by neutralizing antibodies. J Virol 2025; 99:e0003525. [PMID: 40202318 PMCID: PMC12090739 DOI: 10.1128/jvi.00035-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 03/11/2025] [Indexed: 04/10/2025] Open
Abstract
Herpesviruses require membrane fusion for entry and spread, a process facilitated by the fusion glycoprotein B (gB) and the regulatory factor gH/gL. The human cytomegalovirus (HCMV) gH/gL can be modified by the accessory protein gO, or the set of proteins UL128, UL130, and UL131. While the binding of the gH/gL/gO and gH/gL/UL128-131 complexes to cellular receptors, including PDGFRα and NRP2, has been well-characterized structurally, the specific role of receptor engagements by the gH/gL/gO and gH/gL/UL128-131 in regulation of fusion has remained unclear. We describe a cell-cell fusion assay that can quantitatively measure fusion on a timescale of minutes and demonstrate that binding of gH/gL/gO to PDGFRα dramatically enhances gB-mediated cell-cell fusion. In contrast, gH/gL/pUL128-131-regulated fusion is significantly slower, and gH/gL alone cannot promote gB fusion activity within this timescale. The genetic diversity of gO influenced the observed cell-cell fusion rates, correlating with previously reported effects on HCMV infectivity. Mutations in gL that had no effect on the formation of gH/gL/gO or binding to PDGFRa dramatically reduced the cell-cell fusion rate, suggesting that gL plays a critical role in linking the gH/gL/gO-PDGFRa receptor binding to activation of gB. Several neutralizing human monoclonal antibodies were found to potently block gH/gL/gO-PDGFRa-regulated cell-cell fusion, suggesting this mechanism as a therapeutic target. IMPORTANCE Development of vaccines and therapeutics targeting the fusion apparatus of human cytomegalovirus (HCMV) has been limited by the lack of an in vitro cell-cell fusion assay that faithfully models the receptor-dependent fusion characteristic of HCMV entry. The cell-cell fusion assay described here demonstrated that the binding of gH/gL/gO to its receptor, PDGFRα, serves to regulate the activity of the fusion protein gB, and this is specifically vulnerable to inhibition by neutralizing antibodies. Moreover, the measurement of fusion kinetics allows for mutational studies of the fusion mechanism, assessing the influence of genetic diversity among the viral glycoproteins and studying the mechanism of neutralizing antibodies.
Collapse
Affiliation(s)
- Eric P. Schultz
- Division of Biological Sciences, University of Montana, Missoula, Montana, USA
- Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana, USA
| | - Lars Ponsness
- Division of Biological Sciences, University of Montana, Missoula, Montana, USA
| | - Jean-Marc Lanchy
- Division of Biological Sciences, University of Montana, Missoula, Montana, USA
- Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana, USA
| | - Matthias Zehner
- Laboratory for Infection and Immune Biology, University of Cologne, Cologne, Germany
- Institute of Virology, University Cologne, Cologne, Germany
- Faculty of Medicine, University of Cologne, Cologne, Germany
- University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Florian Klein
- Institute of Virology, University Cologne, Cologne, Germany
- Faculty of Medicine, University of Cologne, Cologne, Germany
- University Hospital Cologne, University of Cologne, Cologne, Germany
- Laboratory of Experimental Immunology, University of Cologne, Cologne, Germany
| | - Brent J. Ryckman
- Division of Biological Sciences, University of Montana, Missoula, Montana, USA
- Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, Montana, USA
| |
Collapse
|
7
|
Taguchi A, Misumi F, Teraguchi S, Nagamatsu T, Sakakibara S, Otani T, Ichinose M, Priest D, Nakajima K, Nakamura J, Sawada R, Suzutani T, Ikeda T, Nagura Y, Iriyama T, Okuzaki D, Okazaki H, Wing JB, Hirota Y, Osuga Y. Multifaceted profiling of virus-specific CD8 T cells reveals distinct immune signatures against cytomegalovirus infection states during pregnancy. iScience 2025; 28:112416. [PMID: 40343282 PMCID: PMC12059710 DOI: 10.1016/j.isci.2025.112416] [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: 08/02/2024] [Revised: 12/12/2024] [Accepted: 04/08/2025] [Indexed: 05/11/2025] Open
Abstract
Anti-cytomegalovirus (CMV) serological testing, including the IgG avidity index (AI), is used to assess CMV infection phases during pregnancy. However, little is known about anti-CMV cellular immunity during pregnancy, particularly its relation to serological diagnosis. Herein, using MHC-dextramer single-cell RNA sequencing and flow cytometry, we characterized IE1 and pp65 CMV-antigen specific CD8 T cells from pregnant women with different anti-CMV serological patterns, including IgG+IgM+/AI-low, IgG+IgM+/AI-high, and IgG+IgM-. In IgG+IgM+/AI-low and IgG+IgM+/AI-high specimens, CMV-specific T cells consisted largely of effectors, with a minor but characteristic proportion of memory T cells, including HLA-DR-positive memory precursors and granzyme K-high memory cells reactive to IE1. Conversely, IgG+IgM- cases had a distinctive expansion of pp65-specific terminally differentiated T effector memory with a signature of convergent clonal selection. Our findings revealed that different CMV infection phases have characteristic patterns of CD8 cell phenotype and antigen recognition, potentially offering a new approach for assessing congenital infection risk.
Collapse
Affiliation(s)
- Ayumi Taguchi
- Laboratory of Human Single Cell Immunology, WPI Immunology Frontier Research Center, The University of Osaka, Osaka 565-0871, Japan
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8654, Japan
| | - Fumi Misumi
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8654, Japan
| | - Shunsuke Teraguchi
- Faculty of Data Science, Shiga University, Shiga 522-8522, Japan
- Department of Genome Informatics, Research Institute for Microbial Diseases, The University of Osaka, Osaka 565-0871, Japan
| | - Takeshi Nagamatsu
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8654, Japan
- Department of Obstetrics and Gynecology, International University of Health and Welfare, Narita 286-8686, Japan
| | - Shuhei Sakakibara
- Laboratory of Systems Immunology, WPI Immunology Frontier Research Center, The University of Osaka, Osaka 565-0871, Japan
- Graduate School of Medical Safety Management, Jikei University of Health Care Sciences, Osaka 532-0003, Japan
| | - Tomohiro Otani
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8654, Japan
| | - Mari Ichinose
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8654, Japan
| | - David Priest
- Laboratory of Human Single Cell Immunology, WPI Immunology Frontier Research Center, The University of Osaka, Osaka 565-0871, Japan
| | - Kazuki Nakajima
- Department of Transfusion Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Junko Nakamura
- Department of Transfusion Medicine, The University of Tokyo, Tokyo 113-8655, Japan
- Laboratory Sciences, Department of Health Sciences, School of Health and Social Service, Saitama Prefectural University, Saitama 343-0036, Japan
| | - Ryoko Sawada
- Department of Transfusion Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Tatsuo Suzutani
- Department of Microbiology, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Toshiyuki Ikeda
- Department of Transfusion Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Yutaka Nagura
- Department of Transfusion Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Takayuki Iriyama
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8654, Japan
| | - Daisuke Okuzaki
- Laboratory of Human Immunology (Single Cell Genomics), WPI Immunology Frontier Research Center, The University of Osaka, Osaka 565-0871, Japan
- Human Single Cell Immunology Team, Center for Infectious Disease Education and Research, The University of Osaka, Osaka 565-0871, Japan
| | - Hitoshi Okazaki
- Department of Transfusion Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - James B. Wing
- Laboratory of Human Single Cell Immunology, WPI Immunology Frontier Research Center, The University of Osaka, Osaka 565-0871, Japan
- Human Single Cell Immunology Team, Center for Infectious Disease Education and Research, The University of Osaka, Osaka 565-0871, Japan
| | - Yasushi Hirota
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8654, Japan
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8654, Japan
| |
Collapse
|
8
|
Kawamura S, Nakagawa D, Nagayama T, Katayama Y, Doki N, Takeda W, Nishida T, Matsuoka KI, Ikeda T, Ohigashi H, Sawa M, Fukushima K, Kanda J, Serizawa K, Onizuka M, Fukuda T, Atsuta Y, Kanda Y, Nakasone H. Impacts of donor age and HLA mismatch on HCT outcomes differ according to the donor CMV serostatus in unrelated allo-HCT. Blood Adv 2025; 9:2356-2365. [PMID: 39808797 DOI: 10.1182/bloodadvances.2024014408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 01/16/2025] Open
Abstract
ABSTRACT In unrelated allogeneic hematopoietic cell transplantation (allo-HCT), older and/or HLA-mismatched donors are known risk factors for survival outcomes. In healthy individuals, cytomegalovirus (CMV) seropositivity is associated with impaired adaptive immune systems. We assessed whether the adverse effects of donor risk factors are influenced by the donor CMV serostatus. We analyzed 5836 patients with CMV seropositivity who received unrelated allo-HCT. We divided the entire cohort into 2 cohorts according to the donor CMV serostatus: CMV positive (DP) and negative (DN). We also stratified each cohort into 4 groups based on donor age (aged ≥40 or <40 years) and HLA parity (8/8 or 7/8): Young88 and Old88, and Young78 and Old78, respectively. In the CMV-DP cohort, the Old88 (hazard ratio [HR], 1.20; P = .012), Young78 (HR, 1.35; P < .001), and Old78 (HR, 1.60; P < .001) groups were associated with inferior overall survival (OS) than the Young88 group. In contrast, in the CMV-DN cohort, neither donor age nor HLA disparity was associated with inferior OS. The adverse impact of donor age was different between the cohorts (CMV-DP: HR, 1.19; P = .001; CMV-DN: HR, 1.04; P = .53; P for interaction, .070), as was the impact of HLA (CMV-DP: HR, 1.34; P < .001; CMV-DN: HR, 1.08; P = .23; P for interaction, .012). The impacts of donor age and HLA mismatch on OS might differ according to the donor CMV serostatus. In unrelated allo-HCT from a CMV-seronegative donor, an HLA-mismatched older donor may be able to be selected without affecting OS.
Collapse
Affiliation(s)
- Shunto Kawamura
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
- Division of Emerging Medicine for Integrated Therapeutics, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Daishi Nakagawa
- Department of Hematology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takashi Nagayama
- Division of Cell Transplantation and Transfusion, Jichi Medical University, Shimotsuke, Japan
| | - Yuta Katayama
- Department of Hematology, Hiroshima Red Cross Hospital and Atomic-bomb Survivors Hospital, Hiroshima, Japan
| | - Noriko Doki
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Wataru Takeda
- Department of Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, Tokyo, Japan
| | - Tetsuya Nishida
- Department of Hematology, Japanese Red Cross Aichi Medical Center Nagoya Daiichi Hospital, Nagoya, Japan
| | - Ken-Ichi Matsuoka
- Department of Hematology and Oncology, Okayama University Hospital, Okayama, Japan
| | - Takashi Ikeda
- Division of Hematology and Stem Cell Transplantation, Shizuoka Cancer Center, Shizuoka, Japan
| | - Hiroyuki Ohigashi
- Department of Hematology, Hokkaido University Hospital, Sapporo, Japan
| | - Masashi Sawa
- Department of Hematology and Oncology, Anjo Kosei Hospital, Anjo, Japan
| | - Kentaro Fukushima
- Department of Hematology and Oncology, Osaka University Hospital, Osaka, Japan
| | - Junya Kanda
- Department of Hematology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kentaro Serizawa
- Division of Hematology and Rheumatology, Department of Internal Medicine, Kindai University Hospital, Osakasayama, Japan
| | - Makoto Onizuka
- Department of Hematology/Oncology, Tokai University School of Medicine, Isehara, Japan
| | - Takahiro Fukuda
- Department of Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, Tokyo, Japan
| | - Yoshiko Atsuta
- Japanese Data Center for Hematopoietic Cell Transplantation, Nagakute, Japan
- Department of Registry Science for Transplant and Cellular Therapy, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Yoshinobu Kanda
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
- Division of Hematology, Jichi Medical University, Shimotsuke, Japan
| | - Hideki Nakasone
- Division of Hematology, Jichi Medical University Saitama Medical Center, Saitama, Japan
- Division of Emerging Medicine for Integrated Therapeutics, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| |
Collapse
|
9
|
Walter V, Koch A, Hillmann D, Dannehl D, Staebler A, Pfister K, Risch L, Engler T, Brucker S, Janni W, Hartkopf A, Flatz L. Association of CMV status with response to neoadjuvant chemoimmunotherapy in early triple-negative breast cancer. Cancer Lett 2025; 626:217789. [PMID: 40354994 DOI: 10.1016/j.canlet.2025.217789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 04/15/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Affiliation(s)
- Vincent Walter
- Department of Dermatology, University Hospital Tübingen, Germany
| | - André Koch
- Department of Women's Health, University Hospital Tübingen, Germany
| | | | - Dominik Dannehl
- Department of Women's Health, University Hospital Tübingen, Germany
| | | | | | - Lorenz Risch
- Division of Clinical Biochemistry, Medical University Innsbruck, Austrlia
| | - Tobias Engler
- Department of Women's Health, University Hospital Tübingen, Germany
| | - Sara Brucker
- Department of Women's Health, University Hospital Tübingen, Germany
| | - Wolfgang Janni
- Department of Gynecology, University Hospital Ulm, Germany
| | - Andreas Hartkopf
- Department of Women's Health, University Hospital Tübingen, Germany
| | - Lukas Flatz
- Department of Dermatology, University Hospital Tübingen, Germany; Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland.
| |
Collapse
|
10
|
Yadav J, Kamboj M, Vijayakumar G, Narwal A, Devi A, Kumar A, Keerthika R. Presence of Epstein-Barr Virus in Periapical Pathoses: A Systematic Review and Meta-Analysis. J Endod 2025:S0099-2399(25)00247-X. [PMID: 40350030 DOI: 10.1016/j.joen.2025.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 04/23/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND Periapical lesions, often arising from polymicrobial infections, exhibit varied clinical manifestations. The role of herpes viruses, particularly Epstein-Barr virus (EBV), in the etiopathogenesis of these conditions remains underexplored despite evidence that suggests its involvement in inflammatory processes. OBJECTIVE This systematic review and meta-analysis aimed to evaluate the presence of EBV in periapical diseases and to assess its probable role in their etiopathogenesis. MATERIAL AND METHODS Literature searches were conducted across PubMed, Scopus, Web of Science, and Google Scholar until July 2024. Original research that examined the association of EBV in periapical diseases was included for review. The risk of bias for each study was assessed using the modified Newcastle-Ottawa scale and Review Manager 5.4 was used to generate the results. RESULTS A total of 31 studies were included for qualitative synthesis, with 15 contributing to quantitative analysis. Findings revealed higher EBV detection rates in periapical lesions, particularly in symptomatic cases and lesions with larger sizes. Meta-analysis using random effects models also affirmed that EBV-positive transcripts significantly enhanced the risk of periapical lesions and symptomatic cases. CONCLUSION This review highlights a significant association between EBV and periapical lesions, particularly in symptomatic cases and larger lesions. The findings warrant further investigation into the mechanisms by which EBV contributes to the pathogenesis of periapical diseases and the implications for clinical management, including potential antiviral therapies. PROSPERO REGISTRATION NUMBER CRD4202344796.
Collapse
Affiliation(s)
- Jagveer Yadav
- Department of Oral and Maxillofacial Pathology and Microbiology, Post Graduate Institute of Dental Sciences, Pandit BD Sharma University of Health Sciences, Rohtak, Haryana, India
| | - Mala Kamboj
- Department of Oral and Maxillofacial Pathology and Microbiology, Post Graduate Institute of Dental Sciences, Pandit BD Sharma University of Health Sciences, Rohtak, Haryana, India.
| | - Gopikrishnan Vijayakumar
- Department of Oral and Maxillofacial Pathology and Microbiology, Post Graduate Institute of Dental Sciences, Pandit BD Sharma University of Health Sciences, Rohtak, Haryana, India
| | - Anjali Narwal
- Department of Oral and Maxillofacial Pathology and Microbiology, Post Graduate Institute of Dental Sciences, Pandit BD Sharma University of Health Sciences, Rohtak, Haryana, India
| | - Anju Devi
- Department of Oral and Maxillofacial Pathology and Microbiology, Post Graduate Institute of Dental Sciences, Pandit BD Sharma University of Health Sciences, Rohtak, Haryana, India
| | - Adarsh Kumar
- Department of Public Health Dentistry, Post Graduate Institute of Dental Sciences, Pandit BD Sharma University of Health Sciences, Rohtak, Haryana, India
| | - R Keerthika
- Faculty of Dental Sciences, Department of Oral and Maxillofacial Pathology and Microbiology, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
11
|
Alhalabi M, Alshiekh HA, Alsaiad S, Zarzar M. Prevalence of opportunistic infections in Syrian inflammatory bowel disease patients on biologic therapy: a multi-center retrospective cross-sectional study. BMC Infect Dis 2025; 25:652. [PMID: 40320559 PMCID: PMC12051298 DOI: 10.1186/s12879-025-11063-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 04/30/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Hepatitis B, hepatitis C, cytomegalovirus (CMV), and tuberculosis (TB) pose significant risks to patients with inflammatory bowel disease (IBD) receiving biological therapy. However, data on the prevalence of these infections in Syria are scarce. METHODS We conducted a retrospective chart review of IBD patients receiving biologic therapy at Damascus Hospital and Ibn Al-Nafees Hospital, two major public institutions in Syria, between January 2021 and November 2024. A minimum sample size of 130 was estimated; however, all available records were reviewed. RESULTS Among 185 IBD patients (104 from Damascus and 81 from Ibn Al-Nafees), 51.4% had ulcerative colitis and 47.6% had Crohn's disease. The smoking prevalence was 9.2%, which was higher in Crohn's disease (5.9%) than in ulcerative colitis (3.2%). TST performed in 61.1% of patients, with 4.3% positivity, and interferon-gamma release assay (IGRA) in 8.7% (1.1% positive). Hepatitis B surface antigen (HBsAg) and anti-HBc antibodies were found in 2.7% and 5.4% of the patients, respectively, while hepatitis C seroprevalence was low (0.5%). CMV seropositivity was high in Damascus (50.8%), with two cases (1.1%) of CMV colitis. Biologic therapies included infliximab (42.7%), ustekinumab (24.3%), golimumab (10.8%), and adalimumab (6.5%). Data gaps, particularly in viral serology and TB screening, are notable. CONCLUSION This study identifies deficiencies in TB/hepatitis B screening (notably anti-HBs Ab) and elevated CMV seroprevalence among Syrian IBD patients receiving biologics, extending to immunosuppressed cohorts (rheumatology, dermatology, oncology). Insufficient screening heightens occult infection/reactivation risks, necessitating standardized pretreatment protocols to reduce morbidity in high-risk populations. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Marouf Alhalabi
- Gastroenterologist at Gastroenterology Department of Damascus Hospital, Almujtahed Street, Damascus, Syria.
| | | | - Shadi Alsaiad
- Gastroenterologist at Gastroenterology Department of Damascus Hospital, Almujtahed Street, Damascus, Syria
| | - Mouayad Zarzar
- Gastroenterologist at Gastroenterology Department of Damascus Hospital, Almujtahed Street, Damascus, Syria
| |
Collapse
|
12
|
Hadjiiona A, Michaelides I, Kummer P, Kappelmeyer M, Koeninger A, Reuschel E. Frequency of CMV testing during pregnancy-a retrospective study. Arch Gynecol Obstet 2025; 311:1297-1304. [PMID: 39890645 PMCID: PMC12033204 DOI: 10.1007/s00404-025-07962-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 01/13/2025] [Indexed: 02/03/2025]
Abstract
PURPOSE The cytomegalovirus (CMV) belongs to the family of human Herpesviridae and is distributed worldwide. It is the most common cause of viral congenital infections and can have serious consequences for the health of the fetus in the event of a vertical transmission. This study, taking place for the first time in Upper Palatinate, Bavaria, aims to evaluate the frequency of CMV testing among pregnant women in our region in Germany, which for some individuals can be an expensive individual health service. METHODS Retrospectively, 1000 pregnant patients aged 17-45 years who were treated in the University Clinic St. Hedwig, Regensburg, Germany, were included in the study. It was investigated whether a CMV test was carried out during pregnancy and which results were obtained. RESULTS 597 patients (59.7%) had not received a CMV test during pregnancy. Among the 403 (40.3%) patients who had undergone CMV testing, seropositivity was detected in 143 (35.5%). 257 patients (63.8%) were seronegative, while 3 (0.74%) had a primary infection. CONCLUSION Although CMV is the most common pathogen of viral congenital infections and can severely impair the health of affected newborns, CMV diagnostics during pregnancy is still not an integral part of the maternity guidelines in Germany, but rather an individual healthcare service, meaning that the patients undergoing the test must bear the full cost. An antiviral treatment with valacyclovir has shown good preventive and therapeutic success, but unfortunately, there is currently no vaccination available to prevent vertical transmission, which is why early diagnosis and hygiene measures are the most important means of preventing seroconversion of the mother and possible infection of the fetus.
Collapse
Affiliation(s)
- A Hadjiiona
- University Department of Obstetrics and Gynecology, Clinic St. Hedwig of The Order of St. John, University of Regensburg, Steinmetzstrasse 1-3, 93049, Regensburg, Germany.
| | - I Michaelides
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Regensburg, Regensburg, Germany
| | - P Kummer
- Department of Otorhinolaryngology, Department of Phoniatrics and Pediatric Audiology, University Hospital Regensburg, Regensburg, Germany
| | - M Kappelmeyer
- Chair of Obstetrics and Gynecology, Focus: Obstetrics, University of Regensburg, Biopark 1, Regensburg, Germany
| | - A Koeninger
- University Department of Obstetrics and Gynecology, Clinic St. Hedwig of The Order of St. John, University of Regensburg, Steinmetzstrasse 1-3, 93049, Regensburg, Germany
| | - E Reuschel
- University Department of Obstetrics and Gynecology, Clinic St. Hedwig of The Order of St. John, University of Regensburg, Steinmetzstrasse 1-3, 93049, Regensburg, Germany
| |
Collapse
|
13
|
Pando-Caciano A, Mamaní-Cajachagua PE, Ingunza-Tapia AA, Sánchez-García GJ, Caffo-Valentín XL, Rizo-Patrón E, Murillo-Vizcarra SA, Maita-Malpartida H. Saliva Samples as a Potential Tool for the Diagnosis and Monitoring of Cytomegalovirus Reactivation in Children Undergoing Transplantation. Transplant Proc 2025; 57:663-669. [PMID: 40069044 DOI: 10.1016/j.transproceed.2025.02.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 02/26/2025] [Accepted: 02/26/2025] [Indexed: 04/16/2025]
Abstract
BACKGROUND Regular monitoring of cytomegalovirus (CMV) reactivation in children who undergo transplantation is performed by molecular testing on blood samples. This highlights the need to explore noninvasive alternatives, such as saliva. The present study evaluated the usefulness of saliva samples for detecting and monitoring CMV reactivation in children undergoing transplantation at a referral center in Lima, Peru. METHODS A total of 85 paired samples of liquid saliva and whole blood and 91 paired samples of swab-collected saliva and whole blood from 16 children who underwent transplantation were analyzed by quantitative polymerase chain reaction (qPCR). RESULTS The positivity rates for whole blood, liquid, and swab-collected saliva were 12.59%, 15.29%, and 12.09%, respectively. No significant differences were observed in viral load among the samples, and there was no remarkable correlation or agreement between saliva and whole blood. However, 19 samples from 4 patients were positive in their saliva but negative in their respective whole blood pairs. One patient who was positive in both the saliva samples and negative in blood developed gastrointestinal CMV disease. CONCLUSIONS Saliva may serve as a promising diagnostic tool for detecting and monitoring CMV reactivation in pediatric transplant recipients. Further studies should explore the significance of the saliva viral load to guide pre-emptive therapy and predict disease development.
Collapse
Affiliation(s)
- Alejandra Pando-Caciano
- Sub Unidad de Investigación e Innovación Tecnológica, Instituto Nacional de Salud del Niño San Borja, Lima, Perú; Department of Cellular and Molecular Sciences, School of Science and Philosophy, Universidad Peruana Cayetano Heredia, Lima, Perú.
| | - Pauline E Mamaní-Cajachagua
- Department of Cellular and Molecular Sciences, School of Science and Philosophy, Universidad Peruana Cayetano Heredia, Lima, Perú
| | - Alejandra A Ingunza-Tapia
- Department of Cellular and Molecular Sciences, School of Science and Philosophy, Universidad Peruana Cayetano Heredia, Lima, Perú
| | - Gerardo J Sánchez-García
- Department of Cellular and Molecular Sciences, School of Science and Philosophy, Universidad Peruana Cayetano Heredia, Lima, Perú; Laboratory of Clinical Microbiology, KU Leuven, Department of Microbiology, Immunology and Transplantation, Leuven, Flanders, Belgium
| | - Xiomara L Caffo-Valentín
- Sub Unidad de Investigación e Innovación Tecnológica, Instituto Nacional de Salud del Niño San Borja, Lima, Perú
| | - Emiliana Rizo-Patrón
- Sub Unidad de Investigación e Innovación Tecnológica, Instituto Nacional de Salud del Niño San Borja, Lima, Perú
| | - Sergio A Murillo-Vizcarra
- Sub Unidad de Atención Integral Especializada del Paciente de Trasplante de Progenitores Hematopoyéticos, Instituto Nacional de Salud del Niño San Borja, Lima, Perú
| | - Holger Maita-Malpartida
- Sub Unidad de Investigación e Innovación Tecnológica, Instituto Nacional de Salud del Niño San Borja, Lima, Perú; Department of Cellular and Molecular Sciences, School of Science and Philosophy, Universidad Peruana Cayetano Heredia, Lima, Perú
| |
Collapse
|
14
|
Kondo Y, Higashimoto Y, Hattori F, Kawamura Y, Kozawa K, Miura H, Yoshikawa A, Ihira M, Kawada JI, Yoshikawa T. Epidemiology and Clinical Features of Epstein-Barr Virus and Cytomegalovirus Infections in Japanese Infants and Young Children. J Med Virol 2025; 97:e70383. [PMID: 40329743 DOI: 10.1002/jmv.70383] [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: 12/08/2024] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 05/08/2025]
Abstract
The recent clinical features of Epstein-Barr virus (EBV) and cytomegalovirus (CMV) infections in young children in developed countries remain unclear. This study investigated the clinical features of EBV and CMV infections and the latest seroepidemiology in Japan. Seroprevalence was analyzed 303 stored serum samples using commercial Enzyme Immunosorbent Assay kits, and viral infections were investigated in a cohort of febrile children under 5 years of age. After maternal antibody levels declined, the seroprevalences of EBV and CMV gradually increased by adolescence to 42.9% and 57.1%, respectively. Among 2,732 febrile children, serum EBV and CMV DNAs were detected in 1.76% and 1.24%, respectively. Of 25 primary EBV-infected patients, 15 (60.0%) had infectious mononucleosis (IM) with significantly higher IM frequency, WBC, atypical lymphocyte ratios, AST, ALT, LDH, and EBV DNA load compared to EBV-reactivated patients. No CMV DNA-positive patients had IM. Among primary EBV-infected patients, those with IM were older and had more atypical lymphocytes and higher EBV DNA load than those without IM. The age of primary EBV infection appears to have decreased compared to reports from Western countries in the 1990s. Even among children under 5 years of age, 60.0% of those with primary EBV infection developed IM.
Collapse
Affiliation(s)
- Yotaro Kondo
- Department of Pediatrics, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
- Department of Pediatrics, Konan Kosei Hospital, Konan, Aichi, Japan
| | - Yuki Higashimoto
- Department of Clinical Microbiology, Fujita Health University School of Medical Sciences, Toyoake, Aichi, Japan
| | - Fumihiko Hattori
- Department of Pediatrics, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
- Department of Pediatrics, Toyokawa City Hospital, Toyokawa, Aichi, Japan
| | - Yoshiki Kawamura
- Department of Pediatrics, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
- Department of Pediatrics, Fujita Health University Okazaki Medical Center, Okazaki, Aichi, Japan
| | - Kei Kozawa
- Department of Pediatrics, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Hiroki Miura
- Department of Pediatrics, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Akiko Yoshikawa
- Department of Pediatrics, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Masaru Ihira
- Department of Clinical Engineering, Fujita Health University School of Medical Sciences, Toyoake, Aichi, Japan
| | - Jun-Ichi Kawada
- Department of Pediatrics, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Tetsushi Yoshikawa
- Department of Pediatrics, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| |
Collapse
|
15
|
Quisias J, Gill MJ, Coburn SB, Krentz HB, Beckthold B, Fonseca K, Parkins MD, Lang R. Cytomegalovirus serostatus among people with HIV, characterizing the prevalence, risk factors, and association with immune recovery. HIV Med 2025. [PMID: 40295208 DOI: 10.1111/hiv.70036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/14/2025] [Indexed: 04/30/2025]
Abstract
INTRODUCTION Cytomegalovirus (CMV) infection is common among people with HIV (PWH), and may be associated with negative outcomes. We aimed to identify the seroprevalence of CMV between 01 January 1998 and 01 June 2022 among PWH accessing care at the Southern Alberta Clinic (SAC) and the associated risk factors. We also aimed to assess the impact of CMV seropositivity on CD4+ T-cells and CD4+/CD8+ ratio recovery among PWH who maintain HIV viral suppression. METHODS Poisson regression models with robust variance estimated crude and adjusted prevalence ratios and 95% confidence intervals to identify risk factors for CMV seronegativity. Among PWH maintaining viral suppression, trends in the median CD4+ T-cell count and CD4+/CD8+ ratio were visualized, and continuous time-to-event Cox proportional hazard models estimated hazards ratios (aHR) for CD4+ cell count recovery to ≥500 cells/mm3 and CD4+/CD8+ ratio of >1 at 10 years by CMV serostatus. RESULTS Among 3249 PWH, 2954 (91%) were CMV seropositive. CMV seronegativity was associated with younger ages, male sex, non-Hispanic white race and an education of ≥12 years. While CMV seronegativity did not affect CD4+ T-cell recovery following HIV viral suppression (aHR 1.15 [0.89-1.48]), it was associated with a greater likelihood of CD4+/CD8+ ratio normalization (aHR 2.38 [1.85-3.07]) at 10 years of follow-up. CONCLUSIONS CMV is a common coinfection among PWH. We found that CMV positivity among PWH maintaining HIV viral suppression, while not associated with CD4+ T-cell recovery, was associated with a reduced CD4+/CD8+ ratio recovery. This suggests an association with chronic CMV infection-mediated immune activation and inflammation among PWH.
Collapse
Affiliation(s)
- Joshua Quisias
- Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - M John Gill
- Department of Medicine, University of Calgary, Calgary, Alberta, Canada
- Southern Alberta Clinic, Alberta Health Services, Calgary, Alberta, Canada
- Department of Microbiology, Immunology & Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Sally B Coburn
- Department of Epidemiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hartmut B Krentz
- Department of Medicine, University of Calgary, Calgary, Alberta, Canada
- Southern Alberta Clinic, Alberta Health Services, Calgary, Alberta, Canada
| | - Brenda Beckthold
- Southern Alberta Clinic, Alberta Health Services, Calgary, Alberta, Canada
| | - Kevin Fonseca
- Department of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, Alberta, Canada
- Alberta Public Health Laboratory, Alberta Health Services, Calgary, Alberta, Canada
| | - Michael D Parkins
- Department of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology & Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Raynell Lang
- Department of Medicine, University of Calgary, Calgary, Alberta, Canada
- Southern Alberta Clinic, Alberta Health Services, Calgary, Alberta, Canada
- Department of Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
16
|
Gholami Shahrebabak M, Kouchaki H, Gholami Shahrebabak A, Ravankhah M, Abdollahi M, Akbari M, Lankarani KB. Systematic review and meta-analysis of cytomegalovirus-associated adverse outcomes and healthcare resource utilization in hospitalized patients with inflammatory bowel disease. Int J Colorectal Dis 2025; 40:101. [PMID: 40272527 PMCID: PMC12021708 DOI: 10.1007/s00384-025-04886-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/05/2025] [Indexed: 04/25/2025]
Abstract
PURPOSE Serious complications and unplanned healthcare utilization are reported among inflammatory bowel disease (IBD) hospitalizations with associated cytomegalovirus (CMV). The present systematic review and meta-analysis aimed to examine the in-hospital outcomes of CMV-related hospitalization in IBD patients. METHODS Electronic databases were systematically searched in PubMed, Web of Science (ISI), Scopus, Embase, and Google Scholar until February 2024. The quality of the included studies was assessed using the Newcastle-Ottawa Scale. Cochran's Q test and I2 statistics were applied to evaluate potential heterogeneity across eligible studies. The random-effects model obtained pooled odds ratio (OR) estimates and associated 95% confidence intervals (CI). RESULTS Sixteen articles were included in the meta-analysis, encompassing 5120 IBD patients diagnosed with comorbid CMV infection. Our findings indicated that compared to IBD patients without CMV, those with both CMV and IBD had a longer hospital length of stay (LOS) (8.65 days longer; 95% CI: 6.96, 10.34; P < 0.01), a greater colectomy risk (OR = 2.26; 95% CI: 1.53, 3.34; P < 0.01), and higher in-hospital mortality (OR = 2.83; 95% CI: 1.92, 4.16; P < 0.01). However, the difference in hospital charges between the two groups was not statistically significant (P = 0.78). Sensitivity analysis using the leave-one-out approach revealed significant changes in hospital costs after excluding certain studies. Additionally, subgroup analyses showed significant differences based on IBD subtypes for surgery risk and LOS. CONCLUSION Our findings suggest that CMV infection is associated with poorer outcomes in hospitalized IBD patients, highlighting the importance of early detection and appropriate management of CMV infection in this population to improve clinical outcomes and reduce healthcare resource utilization.
Collapse
Affiliation(s)
- Maryam Gholami Shahrebabak
- Department of Pediatrics, School of Medicine, Afzalipour Hospital, Kerman University of Medical Sciences, Kerman, Iran
| | - Hosein Kouchaki
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- USERN Office, Fasa University of Medical Sciences, Fasa, Iran
| | - Azam Gholami Shahrebabak
- Department of Pediatrics, Afzalipour Hospital, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mahdi Ravankhah
- Student Research Committee, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mozhan Abdollahi
- Student Research Committee, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Akbari
- Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, 8th Floor, Building No. 2, Zand Avenue, Shiraz, Iran.
| | - Kamran B Lankarani
- Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, 8th Floor, Building No. 2, Zand Avenue, Shiraz, Iran.
| |
Collapse
|
17
|
Peterson C, Miller J, Ryckman BJ, Ganusov VV. Apparent cooperativity between human CMV virions introduces errors in conventional methods of calculating multiplicity of infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.23.650360. [PMID: 40313926 PMCID: PMC12045349 DOI: 10.1101/2025.04.23.650360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Whether infection of cells by individual virions occurs randomly or if there is some form(s) of competition or cooperativity between individual virions remains largely unknown for most virus-cell associations. Here we studied cooperativity/competition for three different strains of human cytomegalovirus ( HCMV ) on two different cell types (fibroblasts and epithelial cells). By titrating viral inocula concentrations in small steps over several orders of magnitude, and by using flow cytometry to precisely measure frequency of infected cells, we found that for most virus-cell associations, the frequency of cell infection increases faster than linear with an increasing inoculum concentration, indicating cooperativity between individual infecting virions. Mathematical modeling suggests that this apparent cooperativity cannot be explained by heterogeneity in either the infectivity of the individual virions or the resistance of individual cells to infection, or by simple aggregation/clumping of viral particles. Stochastic simulations of two additional alternative models that allow for i) reduction in cell resistance to infection when exposed to multiple virions, or ii) compensation in infectivity of poorly infectious virions when coinfecting cells with more infectious virions, resulted in apparent viral cooperativity. Analysis of other published datasets suggests presence of apparent viral cooperativity for HIV and vaccinia virus, infecting CRFK or HeLa cells, respectively, but not for tobacco mosaic virus forming plaques on plant leaves. We thus 1) propose a methodology to rigorously evaluate apparent cooperativity of viruses infecting target cells, and 2) demonstrate that knowing the degree of virus cooperativity for any given virus-cell combination is important for an accurate quantification of multiplicity of infection ( MOI ). Graphical abstract Infections per cell (IU/cell) and specific infectivity (Genomes/IU) of human cytomegalovirus depend on the infecting dose.
Collapse
|
18
|
Chen L, Shang Y, Tian X, Huang Y, Sun Y, Fu C, Bai J, Liu Y. The struggles of breastfeeding mothers of preterm infants: a qualitative study. BMC Pregnancy Childbirth 2025; 25:472. [PMID: 40269858 PMCID: PMC12020087 DOI: 10.1186/s12884-025-07597-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 04/11/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Breast milk, with its rich bioactive components and numerous maternal and infant health benefits, is globally recommended as the ideal food for babies, especially premature infants. With the elevated incidence of premature births, suboptimal breastfeeding rates are detrimental, especially considering numerous efforts to promote breastfeeding. The reasons for breastfeeding cessation remain unclear. This study utilizes the social-ecological system theory to explore reasons for the cessation of breastfeeding among mothers of premature infants in China. METHODS An interpretive phenomenological research design was implemented in Wuhan. Through purposive sampling, 14 mothers of premature infants who had discontinued breastfeeding were recruited in Wuhan, China. Semi-structured interviews were conducted to collect data guided by an interview outline developed by the theoretical framework. Content analysis was applied to analyze the interview data. RESULTS The overarching theme of this study was: "On the journey of breastfeeding, who will hold up the sky for me?" This theme encapsulated the multifaceted challenges faced by mothers before discontinuing breastfeeding and their urgent need for comprehensive support. For these mothers, breastfeeding represented not only a physiological challenge but also a significant psychological and emotional burden. The analysis identified three primary categories: Microsystems-the challenges of breastfeeding and adaptation barriers; Mesosystems-dysfunctional support systems within families and hospitals; and Macrosystems-insufficient social support systems. CONCLUSIONS Mothers of premature infants experienced weakened breastfeeding support systems, highlighting the need for (1) Enhancing psychological support and targeted breastfeeding guidance for mothers of preterm infants; (2) Establishing an evidence-based, coordinated support system that integrates hospital and family networks; (3) Strengthening monitoring and enforcement mechanisms for birth policies, enhancing breastfeeding facilities in public spaces, and improving the capacity for breastfeeding education and promotion within communities.
Collapse
Affiliation(s)
- Ling Chen
- Center for Women's and Children's Health Research, Wuhan University School of Nursing, Wuhan University, 115 Donghu Road, Wuhan, 430071, China
- Department of neonatology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yanyan Shang
- Department of neonatology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xuqi Tian
- Center for Women's and Children's Health Research, Wuhan University School of Nursing, Wuhan University, 115 Donghu Road, Wuhan, 430071, China
| | - Yingjuan Huang
- Center for Women's and Children's Health Research, Wuhan University School of Nursing, Wuhan University, 115 Donghu Road, Wuhan, 430071, China
| | - Yu Sun
- Center for Women's and Children's Health Research, Wuhan University School of Nursing, Wuhan University, 115 Donghu Road, Wuhan, 430071, China
| | - Chunhua Fu
- Department of neonatology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jinbing Bai
- Nell Hodgson Woodruff School of Nursing,Emory University , 1520 Clifton Road, Atlanta, GA, 30322, USA
| | - Yanqun Liu
- Center for Women's and Children's Health Research, Research Center for Lifespan Health, Wuhan University School of Nursing, Wuhan University, 115 Donghu Road, Wuhan, 430071, China.
| |
Collapse
|
19
|
Kaur A, Manuel T, Moström M, Crooks C, Davalos A, Barfield R, Scheef E, Kendall S, Midkiff C, Sprehe L, Trexler M, Boquet F, Shroyer M, Danner V, Doyle-Meyers L, Weinbaum C, Mirza A, Lammi S, Otero C, Lee M, Rogers L, Granek J, Owzar K, Malouli D, Fruh K, Kowalik T, Chan C, Permar S, Blair R. Nonhuman primate model mirroring human congenital cytomegalovirus infection reveals a spectrum of vertical transmission outcomes. RESEARCH SQUARE 2025:rs.3.rs-6378923. [PMID: 40313746 PMCID: PMC12045369 DOI: 10.21203/rs.3.rs-6378923/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Congenital cytomegalovirus (cCMV) is the leading infectious cause of birth defects worldwide, yet immune determinants of protection to inform maternal vaccine design remain elusive due to the lack of a translational animal model. Here, we characterized the outcome of primary rhesus CMV (RhCMV) infection in pregnant, immunocompetent, CMV-naïve rhesus macaques. RhCMV DNA was detected in amniotic fluid and/or fetal tissues in six of 12 (50% placental transmission) dams following early second trimester gestation RhCMV inoculation. Widespread tissue dissemination dominated by one of two inoculated RhCMV strains was present in one fetus (8.3% cCMV disease). Placental transmission was associated with elevated fetal and maternal plasma TNF-alpha and reduced maternal brain-derived neurotrophic factor and IL-10 levels. CMV exposure during pregnancy had a broad impact on the placenta and fetus even in the absence of congenital infection, as evidenced by ubiquitous maternal-fetal interface infection, and reduced placental efficiency and small-for-gestation age fetuses compared to control pregnancies. This model recapitulates key aspects of human cCMV and provides new insights into the complexity of CMV vertical transmission.
Collapse
Affiliation(s)
- Amitinder Kaur
- Tulane National Primate Research Center, Tulane University
| | - Tabitha Manuel
- Tulane National Primate Research Center, Tulane University
| | | | | | - Angel Davalos
- Department of Biostatistics and Bioinformatics, Duke University
| | | | | | | | | | - Lesli Sprehe
- Tulane National Primate Research Center, Tulane University
| | | | | | | | - Victoria Danner
- Division of Veterinary Medicine, Tulane National Primate Research Center
| | | | | | - Anne Mirza
- University of Massachusetts Chan Medical School
| | | | - Claire Otero
- Department of Pediatrics, Weill Cornell Medicine
| | | | | | | | | | - Daniel Malouli
- Oregon Health & Science University/Vaccine & Gene Therapy Institute
| | | | | | - Cliburn Chan
- Department of Biostatistics and Bioinformatics, Duke University
| | | | | |
Collapse
|
20
|
Milotay G, Little M, Watson RA, Muldoon D, MacKay S, Kurioka A, Tong O, Taylor CA, Nassiri I, Webb LM, Akin-Adigun O, Bremke J, Ye W, Sun B, Sharma PK, Cooper R, Danielli S, Santo FM, Verge de Los Aires A, Niu G, Cohen L, Ng E, Gilchrist JJ, Chong AY, Mentzer A, Woodcock V, Coupe N, Payne MJ, Youdell M, Middleton MR, Klenerman P, Fairfax BP. CMV serostatus is associated with improved survival and delayed toxicity onset following anti-PD-1 checkpoint blockade. Nat Med 2025:10.1038/s41591-025-03647-1. [PMID: 40269332 DOI: 10.1038/s41591-025-03647-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 03/07/2025] [Indexed: 04/25/2025]
Abstract
Cytomegalovirus (CMV) is a globally endemic latent herpes virus that profoundly impacts T cell immunity. We investigated the oncological consequences of CMV infection across 341 prospectively recruited patients receiving immune checkpoint blockade (ICB) for melanoma. CMV+ patients with metastatic melanoma (MM) have higher lymphocyte counts, reduced neutrophil to lymphocyte ratio and divergent CD8+ T cell gene expression. Combination anti-CTLA-4/anti-PD-1 ICB, but not single-agent anti-PD-1 ICB, induces cytotoxicity and CMV-associated gene expression in CD8+ T cells from CMV- patients. Correspondingly, overall survival was independent of CMV serostatus in combination anti-CTLA-4/anti-PD-1 ICB recipients (CMV+ hazard ratio for death: 1.02, P = 0.92), whereas CMV+ single-agent anti-PD-1 ICB recipients had improved overall survival (CMV+ hazard ratio for death: 0.37, P < 0.01), a finding also seen in CMV+ adjuvant single-agent anti-PD-1 ICB recipients (CMV+ hazard ratio for recurrence: 0.19, P = 0.03). We identify TBX21, encoding T-bet, as a transcriptional driver of CMV-associated CD8+ T cell gene expression, finding that TBX21 expression is predictive of overall survival (hazard ratio: 0.62, P = 0.026). CMV+ patients unexpectedly show reduced cumulative incidence of grade 3+ immune-related adverse events at 6 months (0.30 versus 0.52, P = 2.2 × 10-5), with lower incidence of colitis (P = 7.8 × 10-4) and pneumonitis (P = 0.028), an effect replicated in non-melanoma ICB recipients (n = 58, P = 0.044). Finally, we find reduced CMV seropositivity rates in patients with MM compared with UK Biobank controls (odds ratio: 0.52, P = 1.8 × 10-4), indicating CMV seropositivity may protect against MM. Specifically, patients with BRAF-mutated MM are less likely to be CMV+ (odds ratio = 2.2, P = 0.0054), while CMV- patients present 9 yr earlier with BRAF wild-type MM (P = 1.3 × 10-4). This work reveals an interaction between CMV infection, MM development according to BRAF status and response to ICB, while demonstrating CMV infection is protective against severe ICB immune-related adverse events, highlighting the potential importance of previous infection history and chronic immune activation in MM development and immunotherapy outcomes.
Collapse
Affiliation(s)
- Gusztav Milotay
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - Martin Little
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
- Oxford Cancer-CRUK Oxford Centre, University of Oxford, Churchill Hospital, Oxford, UK
- Kennedy Institute for Rheumatology, University of Oxford, Oxford, UK
| | - Robert A Watson
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
- Oxford Cancer-CRUK Oxford Centre, University of Oxford, Churchill Hospital, Oxford, UK
- Kennedy Institute for Rheumatology, University of Oxford, Oxford, UK
| | - Dylan Muldoon
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - Sophie MacKay
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - Ayako Kurioka
- The Centre for Human Genetics, University of Oxford, Oxford, UK
- Translational Gastroenterology Unit, John Radcliffe Hospital, Oxford, UK
| | - Orion Tong
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - Chelsea A Taylor
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - Isar Nassiri
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - Louisa M Webb
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - Oluwafemi Akin-Adigun
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - Julia Bremke
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - Weiyu Ye
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Bo Sun
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, UK
| | - Piyush Kumar Sharma
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - Ros Cooper
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - Sara Danielli
- Oxford Cancer-CRUK Oxford Centre, University of Oxford, Churchill Hospital, Oxford, UK
| | - Flavia Matos Santo
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - Alba Verge de Los Aires
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - Guangyi Niu
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - Lea Cohen
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Oncology, University of Oxford, Oxford, UK
| | - Esther Ng
- Kennedy Institute for Rheumatology, University of Oxford, Oxford, UK
| | - James J Gilchrist
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Amanda Y Chong
- The Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Alex Mentzer
- The Centre for Human Genetics, University of Oxford, Oxford, UK
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
| | - Victoria Woodcock
- Department of Oncology, University of Oxford, Oxford, UK
- Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Nicholas Coupe
- Department of Oncology, University of Oxford, Oxford, UK
- Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Miranda J Payne
- Department of Oncology, University of Oxford, Oxford, UK
- Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Michael Youdell
- Oxford Cancer-CRUK Oxford Centre, University of Oxford, Churchill Hospital, Oxford, UK
| | - Mark R Middleton
- Department of Oncology, University of Oxford, Oxford, UK
- Oxford Cancer-CRUK Oxford Centre, University of Oxford, Churchill Hospital, Oxford, UK
- Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Paul Klenerman
- The Centre for Human Genetics, University of Oxford, Oxford, UK
- Translational Gastroenterology Unit, John Radcliffe Hospital, Oxford, UK
| | - Benjamin P Fairfax
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK.
- Department of Oncology, University of Oxford, Oxford, UK.
- Oxford Cancer-CRUK Oxford Centre, University of Oxford, Churchill Hospital, Oxford, UK.
- Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| |
Collapse
|
21
|
Lankina A, Raposo M, Hargreaves A, Atkinson C, Griffiths P, Reeves MB. Developing a Vaccine Against Human Cytomegalovirus: Identifying and Targeting HCMV's Immunological Achilles' Heel. Vaccines (Basel) 2025; 13:435. [PMID: 40432047 PMCID: PMC12115399 DOI: 10.3390/vaccines13050435] [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: 02/25/2025] [Revised: 04/10/2025] [Accepted: 04/18/2025] [Indexed: 05/29/2025] Open
Abstract
Human cytomegalovirus (HCMV) is a critical pathogen in immunocompromised populations, such as organ transplant recipients as well as congenitally infected neonates with immature immune systems. Despite decades of research and the growing financial burden associated with the management of HCMV, there is no licensed vaccine to date. In this review, we aim to outline the complexity of HCMV and the antigens it presents and the journey and challenges of developing an effective HCMV vaccine, as well as further highlight the recent analyses of the most successful vaccine candidate so far-gB/MF59.
Collapse
Affiliation(s)
- Anastasia Lankina
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PP, UK; (A.L.); (M.R.); (A.H.); (P.G.)
| | - Marta Raposo
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PP, UK; (A.L.); (M.R.); (A.H.); (P.G.)
| | - Alexander Hargreaves
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PP, UK; (A.L.); (M.R.); (A.H.); (P.G.)
| | - Claire Atkinson
- School of Applied and Health Sciences, London South Bank University, London SE1 0AA, UK;
| | - Paul Griffiths
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PP, UK; (A.L.); (M.R.); (A.H.); (P.G.)
| | - Matthew B. Reeves
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PP, UK; (A.L.); (M.R.); (A.H.); (P.G.)
| |
Collapse
|
22
|
Boquett JA, Sauter J, Schmidt AH, Maiers M, Hollenbach JA. Human leukocyte antigen variation is associated with cytomegalovirus serostatus in healthy individuals. Am J Hum Genet 2025; 112:913-926. [PMID: 40049169 DOI: 10.1016/j.ajhg.2025.02.007] [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: 09/12/2024] [Revised: 02/05/2025] [Accepted: 02/07/2025] [Indexed: 03/12/2025] Open
Abstract
Cytomegalovirus (CMV) is a common β-herpes virus worldwide with an estimated seroprevalence among the general population of 83%. Primary infection is usually benign; however, CMV can cause severe morbidity in newborns in whom it is acquired congenitally, as well as immunocompromised individuals. Understanding the role of immunogenetic variation in risk for CMV infection can provide insight into the immune control of this ubiquitous pathogen. Here, we evaluated the association of human leukocyte antigen (HLA) genetic variation with CMV seropositivity in more than 518,000 individuals from two independent cohorts. We found three HLA class II alleles (HLA-DRB1∗04:03 with risk; HLA-DRB1∗01:03 and HLA-DRB1∗07:01 with protection) to be significantly associated with CMV serostatus across both cohorts and in multiple population subgroups. Interestingly, HLA-DRB1∗04:03 and HLA-DRB1∗01:03, the alleles with the strongest observed effect, are relatively rare, while common homologous alleles show no association with CMV. We show that these differences are mediated by changes in charge and volume to two key pockets in the peptide-binding groove of the HLA molecule, providing a structural basis for the observed association. Our results provide population-scale evidence for the role of HLA in mediating infection with this ubiquitous human virus and a framework for understanding immunological conditions necessary for efficient viral control.
Collapse
Affiliation(s)
- Juliano A Boquett
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | | | | | - Martin Maiers
- CIBMTR (Center for International Blood and Marrow Transplant Research), NMDP, Minneapolis, MN, USA
| | - Jill A Hollenbach
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
23
|
Wang W, Moise PA, Wong M, Gao W, Xiang C, Zion A, Vincenti F, Beyer AP. Healthcare resource use and clinical events associated with neutropenia among adult kidney transplant recipients receiving valganciclovir or ganciclovir prophylaxis: a matched case-control cohort study. Curr Med Res Opin 2025; 41:639-646. [PMID: 40302692 DOI: 10.1080/03007995.2025.2498674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND Cytomegalovirus prophylaxis with valganciclovir and ganciclovir is associated with increased neutropenia risk in kidney transplant recipients. This study assessed the association between neutropenia and short-term healthcare resource utilization (HCRU) and clinical events for adults receiving valganciclovir and/or ganciclovir prophylaxis post-kidney transplantation in the United States. METHODS Adult kidney transplant recipients from 2012 to 2020 who received valganciclovir and/or ganciclovir prophylaxis were identified in the TriNetX database. Patients with a neutropenia event during the 1-year post-kidney transplant were identified as cases and those without were identified as controls. Cases and controls were matched 1:1 on patient characteristics in the 1-year period before the kidney transplant. HCRU and clinical event rates in the year following the first neutropenia event were compared between the cases and controls. RESULTS Of 3564 identified case-control pairs, the average age was 52 years and 59.57% were male. Cases had higher HCRU in the first year after neutropenia than controls, characterized by 1.40-, 1.28-, and 1.33-times higher rates of inpatient, outpatient, and emergency department visits (all p < 0.001). Cases also had higher incident rates for cytomegalovirus-related events, use of granulocyte colony stimulating factor, and opportunistic infections (all adjusted incident rate ratios >1 with p < 0.01), and greater risks of death, graft loss, and acute graft rejection (all adjusted hazard ratios >1 with p < 0.001). CONCLUSION Higher HCRU and clinical burden were observed within the first year following post-transplant neutropenia. New options for cytomegalovirus prophylaxis that are less likely to induce neutropenia are needed to improve patient outcomes.
Collapse
Affiliation(s)
| | | | | | - Wei Gao
- Analysis Group, Inc., Boston, MA, USA
| | | | | | | | | |
Collapse
|
24
|
Xi Y, Mokry RL, Armas ND, Kline I, Wegner M, Purdy JG. Human Cytomegalovirus Infection Reduces an Endogenous Antiviral Fatty Acid by Promoting Host Metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.31.646481. [PMID: 40235993 PMCID: PMC11996439 DOI: 10.1101/2025.03.31.646481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Some viruses, including human cytomegalovirus (HCMV), induce the synthesis of fatty acids and lipids to ensure that the lipid environment of infected cells supports virus replication. HCMV infection broadly reprograms metabolism to ensure central carbon metabolism provides the metabolites required for anabolic synthesis of nucleotides, proteins, and lipids while also meeting the energy demands placed on the infected cells. While HCMV infection increases the levels of most very long chain fatty acids (VLCFA), we found that the levels of erucic acid (EA), a C22:1 monounsaturated VLCFA, are reduced. Treating infected cells with EA disrupted a late step in virus replication, resulting in the release of virions with reduced infectivity. Moreover, we used lipidomics to determine that EA-treated cells had elevated levels of lipids containing a combination of a C22:1 tail and a VLC polyunsaturated fatty acid tail (VLC-PUFA). We demonstrate that fatty acid elongase 5 (ELOVL5) mediated production of VLC-PUFAs is stimulated by HCMV infection. ELOVL5 aided the increase in lipids with C22:1 plus VLC-PUFA tails following EA treatment and reduced the overall level of C22:1 in HCMV-infected cells. Moreover, we found that ELOVL5 mollified EA inhibition of HCMV replication, suggesting ELOVL5 plays a critical role in reducing the level of an endogenous FA with antiviral properties. Our study provides insight into how infection may increase the synthesis of an antiviral metabolite or FA and how the virus may evade their antiviral effect by promoting their metabolism.
Collapse
|
25
|
Wang J, Rabiee B, Patel C, Jafri M, Hussain H, Chaudhry A, Chaudhry I, Kamoun L, Chaudhry I, Oh L, Bobat FI, Shukla D, Farooq AV. Herpesvirus Infections of the Corneal Endothelium. Microorganisms 2025; 13:778. [PMID: 40284615 PMCID: PMC12029634 DOI: 10.3390/microorganisms13040778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/24/2025] [Accepted: 03/11/2025] [Indexed: 04/29/2025] Open
Abstract
Corneal endotheliitis is an inflammatory process, most commonly of viral etiology, that manifests clinically with features including corneal edema, keratic precipitates, and a mild anterior chamber reaction. Several studies have implicated human herpesviruses from the Herpesviridae family as primary causes of corneal endotheliitis, including cytomegalovirus (CMV), varicella zoster virus (VZV), and herpes simplex viruses 1 and 2 (HSV-1 and HSV-2). This review critically evaluates the present literature surrounding herpesvirus infections of the corneal endothelium.
Collapse
Affiliation(s)
- Jessie Wang
- Duke Eye Center, Duke University, Durham, NC 27705, USA;
| | - Behnam Rabiee
- Department of Ophthalmology, Nazareth Hospital, Trinity Health Mid-Atlantic, Philadelphia, PA 19152, USA; (B.R.); (C.P.); (A.C.); (I.C.); (L.K.); (I.C.)
- IC Laser Eye Care, Bensalem, PA 19020, USA; (M.J.); (H.H.)
| | - Chandani Patel
- Department of Ophthalmology, Nazareth Hospital, Trinity Health Mid-Atlantic, Philadelphia, PA 19152, USA; (B.R.); (C.P.); (A.C.); (I.C.); (L.K.); (I.C.)
- IC Laser Eye Care, Bensalem, PA 19020, USA; (M.J.); (H.H.)
- Rowan-Virtua School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084, USA
| | - Mansab Jafri
- IC Laser Eye Care, Bensalem, PA 19020, USA; (M.J.); (H.H.)
| | - Hamad Hussain
- IC Laser Eye Care, Bensalem, PA 19020, USA; (M.J.); (H.H.)
| | - Aaila Chaudhry
- Department of Ophthalmology, Nazareth Hospital, Trinity Health Mid-Atlantic, Philadelphia, PA 19152, USA; (B.R.); (C.P.); (A.C.); (I.C.); (L.K.); (I.C.)
- IC Laser Eye Care, Bensalem, PA 19020, USA; (M.J.); (H.H.)
| | - Imtiaz Chaudhry
- Department of Ophthalmology, Nazareth Hospital, Trinity Health Mid-Atlantic, Philadelphia, PA 19152, USA; (B.R.); (C.P.); (A.C.); (I.C.); (L.K.); (I.C.)
- IC Laser Eye Care, Bensalem, PA 19020, USA; (M.J.); (H.H.)
| | - Layla Kamoun
- Department of Ophthalmology, Nazareth Hospital, Trinity Health Mid-Atlantic, Philadelphia, PA 19152, USA; (B.R.); (C.P.); (A.C.); (I.C.); (L.K.); (I.C.)
- IC Laser Eye Care, Bensalem, PA 19020, USA; (M.J.); (H.H.)
| | - Iftikhar Chaudhry
- Department of Ophthalmology, Nazareth Hospital, Trinity Health Mid-Atlantic, Philadelphia, PA 19152, USA; (B.R.); (C.P.); (A.C.); (I.C.); (L.K.); (I.C.)
- IC Laser Eye Care, Bensalem, PA 19020, USA; (M.J.); (H.H.)
| | - Lewis Oh
- Pritzker School of Medicine, The University of Chicago, Chicago, IL 60637, USA;
| | - Fatima I. Bobat
- Department of Ophthalmology and Visual Sciences, The University of Illinois Chicago, Chicago, IL 60607, USA; (F.I.B.); (D.S.)
| | - Deepak Shukla
- Department of Ophthalmology and Visual Sciences, The University of Illinois Chicago, Chicago, IL 60607, USA; (F.I.B.); (D.S.)
| | - Asim V. Farooq
- Department of Ophthalmology and Visual Science, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
26
|
Mai J, Nazari M, Stamminger T, Schreiner S. Daxx and HIRA go viral - How chromatin remodeling complexes affect DNA virus infection. Tumour Virus Res 2025; 19:200317. [PMID: 40120981 DOI: 10.1016/j.tvr.2025.200317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/13/2025] [Accepted: 03/13/2025] [Indexed: 03/25/2025] Open
Abstract
Daxx and HIRA are key proteins in the host response to DNA virus infections. Daxx is involved in apoptosis, transcription regulation, and stress responses. During DNA virus infections, Daxx helps modulate the immune response and viral progression. Viruses like adenoviruses and herpesviruses can exploit Daxx to evade immune detection, either by targeting it for degradation or inhibiting its function. Daxx also interacts with chromatin to regulate transcription, which viruses can manipulate to enhance their own gene expression and replication. HIRA is a histone chaperone and reported to be essential for chromatin assembly and gene regulation. It plays a critical role in maintaining chromatin structure and modulating gene accessibility. During DNA virus infection, HIRA influences chromatin remodeling, affecting both viral and host DNA accessibility, which impacts viral replication and gene expression. Additionally, the histone variant H3.3 is crucial for maintaining active chromatin states. It is incorporated into chromatin independently of DNA replication and is associated with active gene regions. During viral infections, H3.3 dynamics can be altered, affecting viral genome accessibility and replication efficiency. Overall, Daxx and HIRA are integral to orchestrating viral infection programs, maintaining latency and/or persistence, and influencing virus-induced transformation by modulating chromatin dynamics and host immune responses, making them significant targets for therapeutic strategies once fully understood. Here, we summarize various DNA viruses and their crosstalk with Daxx and HIRA.
Collapse
Affiliation(s)
- Julia Mai
- Institute of Virology, Medical Center - University of Freiburg, Freiburg, Germany
| | - Masih Nazari
- Institute of Virology, Medical Center - University of Freiburg, Freiburg, Germany
| | | | - Sabrina Schreiner
- Institute of Virology, Medical Center - University of Freiburg, Freiburg, Germany; Institute of Virology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
27
|
Kim JS, Lee NR, Park KI, Hwang HS, Lee SH, Chung BH, Jung CW, Cho JH, Park WY, Kim HJ, Jeong JC, Yang J, Lee YH, Park JB, Jeon JS, Lee J, Kim YH, Choi SJN, Oh J, Yoon HE, Kim DG, Shin HS, Ban TH, Kim MS, Ko MJ, Jeong KH. Valacyclovir for the prevention of cytomegalovirus infection after kidney transplantation. BMC Infect Dis 2025; 25:314. [PMID: 40045190 PMCID: PMC11881300 DOI: 10.1186/s12879-025-10671-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 02/18/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Cytomegalovirus (CMV) infection is a frequent complication after kidney transplantation (KT) and has various effects on recipient and graft survival. Although guidelines recommend anti-viral prophylaxis with ganciclovir or valganciclovir, there is a demand for alternative regimen for CMV prevention. We investigated the effects of a 3-month valacyclovir-based prophylaxis on CMV infection and clinical outcomes in KT recipients using a nationwide cohort. METHODS Overall, 2,584 KT recipients from 20 transplant centers registered with the Korean Organ Transplantation Registry between May 2014 and December 2019 were analyzed in this study. The recipients were divided into valacyclovir prophylaxis and non-prophylaxis groups, a 1:3 propensity score matching was performed, and 1,036 recipients (291 and 745 in the prophylaxis and non-prophylaxis groups, respectively) were analyzed. The impact of valacyclovir-based prophylaxis on CMV after KT, other clinical outcomes, and the risk factors for CMV infection development were investigated. RESULTS The prophylaxis group showed a lower incidence of CMV infection and rejection compared to the non-prophylaxis group (3.64 vs. 10.25 events/100 person-years and 1.85 vs. 7.27 events/100 person-years, respectively). Valacyclovir prophylaxis, donor age, deceased donor, length of hospitalization after KT, anti-thymocyte globulin use, and CMV serological mismatch between the donor and recipient were independent risk factors for CMV infection after KT. CONCLUSIONS Valacyclovir prophylaxis after KT significantly reduced CMV infection and rejection. We suggest that valacyclovir could be considered as an alternative strategy for CMV prophylaxis after KT. However, our study has limitations, including its retrospective design, variability in valacyclovir dosing and CMV monitoring, and unassessed confounding factors. Further prospective studies with standardized protocols and larger cohorts are needed to validate our findings.
Collapse
Affiliation(s)
- Jin Sug Kim
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University College of Medicine, Kyung Hee University Medical Center, 26, Kyungheedae-Ro, Dongdaemun-Gu, Seoul, 02447, Republic of Korea
| | - Na Rae Lee
- Division of Healthcare Technology Assessment Research, National Evidence-Based Healthcare Collaborating Agency, 400, Neungdong-Ro, Gwangjin-Gu, Seoul, 04933, Republic of Korea
| | - Kyun-Ik Park
- Division of Healthcare Technology Assessment Research, National Evidence-Based Healthcare Collaborating Agency, 400, Neungdong-Ro, Gwangjin-Gu, Seoul, 04933, Republic of Korea
| | - Hyeon Seok Hwang
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University College of Medicine, Kyung Hee University Medical Center, 26, Kyungheedae-Ro, Dongdaemun-Gu, Seoul, 02447, Republic of Korea
| | - Sang Ho Lee
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University College of Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea
| | - Byung Ha Chung
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary'S Hospital, Seoul, Republic of Korea
| | - Cheol Woong Jung
- Department of Surgery, Korea University Anam Hospital, Seoul, Republic of Korea
| | - Jang-Hee Cho
- Department of Internal Medicine, School of Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Woo Yeong Park
- Division of Nephrology, Department of Internal Medicine, Keimyung University School of Medicine, Keimyung University Dongsan Hospital, Daegu, Republic of Korea
| | - Hyo Jin Kim
- Division of Nephrology, Department of Internal Medicine, Pusan National University Hospital, Pusan National University School of Medicine, Busan, Republic of Korea
| | - Jong Cheol Jeong
- Division of Nephrology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Jaeseok Yang
- Division of Nephrology, Department of Internal Medicine, Yonsei University College of Medicine, Severance Hospital, Seoul, Republic of Korea
| | - Yu Ho Lee
- Division of Nephrology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | - Jae Berm Park
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jin Seok Jeon
- Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Juhan Lee
- Department of Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yeong Hoon Kim
- Department of Internal Medicine, Inje University Busan Paik Hospital, Busan, Republic of Korea
| | - Soo Jin Na Choi
- Department of Surgery, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Jieun Oh
- Department of Internal Medicine, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Hye Eun Yoon
- Division of Nephrology, Department of Internal Medicine, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Incheon, Republic of Korea
| | - Deok Gie Kim
- Department of Surgery, Yonsei University Wonju College of Medicine, Wonju Severance Christian Hospital, Wonju, Republic of Korea
| | - Ho Sik Shin
- Renal Division, Department of Internal Medicine, Gospel Hospital, Kosin University College of Medicine, Busan, South Korea
- Transplantation Research Institute, Kosin University College of Medicine, Busan, South Korea
| | - Tae Hyun Ban
- Division of Nephrology, Department of Internal Medicine, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Myoung Soo Kim
- Department of Surgery, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Min Jung Ko
- Division of Healthcare Technology Assessment Research, National Evidence-Based Healthcare Collaborating Agency, 400, Neungdong-Ro, Gwangjin-Gu, Seoul, 04933, Republic of Korea.
| | - Kyung Hwan Jeong
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University College of Medicine, Kyung Hee University Medical Center, 26, Kyungheedae-Ro, Dongdaemun-Gu, Seoul, 02447, Republic of Korea.
| |
Collapse
|
28
|
Coskun E, Kakkar F, Riley LE, Ciaranello AL, Prabhu M. Evaluation and Management of Congenital Cytomegalovirus Infection. Obstet Gynecol 2025; 145:297-306. [PMID: 39847776 DOI: 10.1097/aog.0000000000005840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/12/2024] [Indexed: 01/25/2025]
Abstract
The purpose of this review is to serve as an update on congenital cytomegalovirus (CMV) evaluation and management for obstetrician-gynecologists and to provide a framework for counseling birthing people at risk for or diagnosed with a primary CMV infection or reactivation or reinfection during pregnancy. A DNA virus, CMV is the most common congenital viral infection and the most common cause of nongenetic childhood hearing loss in the United States. The risk of congenital CMV infection from transplacental viral transfer depends on the gestational age at the time of maternal infection and whether the infection is primary or nonprimary. Although the risk of congenital CMV infection is lower with infection at earlier gestational ages, clinical sequelae are more severe with maternal infections earlier in gestation. At present, routine screening for maternal CMV infection is not recommended by U.S. guidelines. When maternal primary infection is confirmed in early pregnancy, emerging data support consideration of maternal antiviral therapy to prevent congenital CMV infection. When congenital CMV infection is confirmed, typically after an abnormal prenatal ultrasound result, there are more limited data on the utility of maternal antiviral therapy. Universal newborn screening for congenital CMV infection is not mandatory in most U.S. states at present. Newborns diagnosed with congenital CMV infection undergo an extensive evaluation to determine whether neurologic symptoms are present, which guides postnatal evaluation and management. In this review, we discuss the diagnosis and management of maternal CMV infection, the risk and diagnosis of congenital CMV infection, prevention and potential treatment of congenital CMV infection in utero, and neonatal congenital CMV infection diagnosis and management.
Collapse
Affiliation(s)
- Elif Coskun
- Medical Practice Evaluation Center, the Division of Infectious Disease, and the Division of Maternal Fetal Medicine, Massachusetts General Hospital, Boston, Massachusetts; the Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Montreal, Quebec, Canada; and the Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, New York
| | | | | | | | | |
Collapse
|
29
|
Ozer EA, Keskin A, Berrak YH, Cankara F, Can F, Gursoy-Ozdemir Y, Keskin O, Gursoy A, Yapici-Eser H. Shared interactions of six neurotropic viruses with 38 human proteins: a computational and literature-based exploration of viral interactions and hijacking of human proteins in neuropsychiatric disorders. DISCOVER MENTAL HEALTH 2025; 5:18. [PMID: 39987419 PMCID: PMC11846830 DOI: 10.1007/s44192-025-00128-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 01/09/2025] [Indexed: 02/24/2025]
Abstract
INTRODUCTION Viral infections may disrupt the structural and functional integrity of the nervous system, leading to acute conditions such as encephalitis, and neuropsychiatric conditions as mood disorders, schizophrenia, and neurodegenerative diseases. Investigating viral interactions of human proteins may reveal mechanisms underlying these effects and offer insights for therapeutic interventions. This study explores molecular interactions of virus and human proteins that may be related to neuropsychiatric disorders. METHODS Herpes Simplex Virus-1 (HSV-1), Cytomegalovirus (CMV), Epstein-Barr Virus (EBV), Influenza A virus (IAV) (H1N1, H5N1), and Human Immunodeficiency Virus (HIV1&2) were selected as key viruses. Protein structures for each virus were accessed from the Protein Data Bank and analyzed using the HMI-Pred web server to detect interface mimicry between viral and human proteins. The PANTHER classification system was used to categorize viral-human protein interactions based on function and cellular localization. RESULTS Energetically favorable viral-human protein interactions were identified for HSV-1 (467), CMV (514), EBV (495), H1N1 (3331), H5N1 (3533), and HIV 1&2 (62425). Besides immune and apoptosis-related pathways, key neurodegenerative pathways, including those associated with Parkinson's and Huntington's diseases, were frequently interacted. A total of 38 human proteins, including calmodulin 2, Ras-related botulinum toxin substrate 1 (Rac1), PDGF-β, and vimentin, were found to interact with all six viruses. CONCLUSION The study indicates a substantial number of energetically favorable interactions between human proteins and selected viral proteins, underscoring the complexity and breadth of viral strategies to hijack host cellular mechanisms. Further in vivo and in vitro validation is required to understand the implications of these interactions.
Collapse
Affiliation(s)
| | - Aleyna Keskin
- School of Medicine, Koç University, Istanbul, Turkey
| | | | - Fatma Cankara
- Graduate School of Sciences and Engineering, Computational Sciences and Engineering, Koç University, Istanbul, Turkey
| | - Fusun Can
- Department of Microbiology, School of Medicine, Koç University, Istanbul, Turkey
| | - Yasemin Gursoy-Ozdemir
- Department of Neurology, School of Medicine, Koç University, Istanbul, Turkey
- Research Center for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey
| | - Ozlem Keskin
- Department of Chemical and Biological Engineering, College of Engineering, Koç University, Istanbul, Turkey
| | - Attila Gursoy
- Department of Computer Science and Engineering, College of Engineering, Koç University, Istanbul, Turkey.
| | - Hale Yapici-Eser
- Research Center for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey.
- Department of Psychiatry, School of Medicine, Koç University, Istanbul, Turkey.
| |
Collapse
|
30
|
Hilt ZT, Reynaldi A, Steinhilber M, Zhang S, Wesnak SP, Smith NL, Davenport MP, Rudd BD. Recent thymic emigrants are preferentially recruited into the memory pool during persistent infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.06.636722. [PMID: 39975271 PMCID: PMC11839080 DOI: 10.1101/2025.02.06.636722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Cytomegalovirus (CMV) leads to a unique phenomenon known as 'memory inflation,' where antigen-specific memory CD8+ T cells continue to accumulate in the peripheral tissues during the latent stage of infection. However, it is still not clear how the inflating pool of memory CD8+ T cells is generated and maintained. In this study, we used murine cytomegalovirus (MCMV) as a model of persistent infection and fate-mapping mice to determine the dynamics of CD8+ T cell recruitment into the memory pool. We found that neonatal exposure to CMV leads to an expansion of newly made CD8+ T cells (recent thymic emigrants, RTEs), which are maintained in the long-lived memory compartment. In contrast, CD8+ T cells made during the latent phase of infection (mature CD8+ T cells) contribute little to the memory pool. We also observed notable phenotypic differences between RTEs and mature cells. Whereas RTEs present at the time of infection gave rise to more effector memory cells, the cells produced later in infection were biased towards becoming central memory cells. Importantly, the preferential recruitment of RTEs into the effector memory pool also occurs during adult exposure to CMV. Collectively, these data demonstrate that persistent infection expands the RTE population, and timing of infection dictates whether neonatal or adult RTEs are 'locked in' to the memory pool.
Collapse
Affiliation(s)
- Zachary T. Hilt
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Arnold Reynaldi
- Kirby Institute, University of New South Wales, Kensington, NSW, Australia
| | - Megan Steinhilber
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Shide Zhang
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Samantha P. Wesnak
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Norah L. Smith
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Miles P Davenport
- Kirby Institute, University of New South Wales, Kensington, NSW, Australia
| | - Brian D. Rudd
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
31
|
Okalany NRA, Mukunya D, Olupot-Olupot P, Chebet M, Okello F, Weeks AD, Bisso F, Tylleskär T, Burgoine K, Engebretsen IMS. Postnatal cytomegalovirus infection and its effect on hearing and neurodevelopmental outcomes among infants aged 3-10 months: A cohort study in Eastern Uganda. PLoS One 2025; 20:e0318655. [PMID: 39913500 PMCID: PMC11801545 DOI: 10.1371/journal.pone.0318655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/21/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Hearing impairment and neurodevelopmental disorders pose a significant global health burden in children. The link between postnatal cytomegalovirus (CMV) infection and these outcomes remains unclear. This study explored the association of postnatal CMV infection with hearing and neurodevelopmental outcomes in term infants aged 3 to 10 months. METHODS This was a cohort sub-study within the BabyGel cluster randomised trial in Eastern Uganda. From 1265 term infants screened for CMV, 219 were negative at birth but positive at 3 months, and were age-matched with 219 CMV-negative controls. CMV status was determined by PCR screening of saliva samples, with positive results confirmed using urine samples (Chai Open qPCR, Santa Clara, CA). From the established cohort, 424 infants were successfully followed up between 3 to 10 months of age. Clinical assessments included neurodevelopmental evaluation using the Malawi Developmental Assessment Tool, the Hammersmith Infant Neurological Examination, and hearing screening using Otoacoustic Emission testing (Otoport Lite, Otodynamics Limited). Statistical analyses were performed using descriptive statistics, chi-square tests and log binomial regression models with Stata 18. RESULTS Of the 424 infants included in the study, 206 were postnatal CMV-infected and 218 were uninfected. Neurodevelopmental assessments indicated no differences between postnatal CMV-infected infants and uninfected groups (ARR 0.88, 95% CI [0.67, 1.15], p = 0.346). Hearing screening revealed a 1.99-fold increased risk of a positive result for postnatal CMV-infected infants compared to uninfected infants (67/106 vs. 39/106, 95% CI [1.27, 3.12], p = 0.003). CONCLUSION Postnatal CMV infection was associated with more positive hearing screenings, though no significant differences in neurodevelopmental outcomes were observed in early infancy. Exploration into the feasibility of incorporating hearing and CMV screening into routine care will play a vital role in early identification and intervention, improving the management of both hearing and CMV-related conditions in resource-limited settings.
Collapse
Affiliation(s)
- Noela Regina Akwi Okalany
- Department of Global Public Health and Primary Care, Centre for International Health, University of Bergen, Bergen, Norway
| | - David Mukunya
- Department of Community and Public Health, Busitema University, Mbale, Uganda
| | - Peter Olupot-Olupot
- Department of Community and Public Health, Busitema University, Mbale, Uganda
- Mbale Clinical Research Institute, Mbale, Uganda
| | - Martin Chebet
- Department of Global Public Health and Primary Care, Centre for International Health, University of Bergen, Bergen, Norway
- Department of Paediatrics and Child Health, Busitema University, Mbale, Uganda
| | - Francis Okello
- Department of Global Public Health and Primary Care, Centre for International Health, University of Bergen, Bergen, Norway
- Department of Community and Public Health, Busitema University, Mbale, Uganda
| | - Andrew D. Weeks
- Department of Women’s and Children’s Health, Sanyu Research Unit, University of Liverpool, Liverpool, United Kingdom
| | - Fred Bisso
- Mbale Regional Referral Hospital, Mbale, Uganda
| | - Thorkild Tylleskär
- Department of Global Public Health and Primary Care, Centre for International Health, University of Bergen, Bergen, Norway
| | - Kathy Burgoine
- Department of Global Public Health and Primary Care, Centre for International Health, University of Bergen, Bergen, Norway
- Mbale Clinical Research Institute, Mbale, Uganda
- Mbale Regional Referral Hospital, Mbale, Uganda
| | | |
Collapse
|
32
|
Herrero‐Sánchez A, Haroyan‐Darbinyan E. Prevalence of Epstein-Barr Virus, Cytomegalovirus, and Periodontopathic Bacteria in Patients With Periodontitis: A Case-Control Study. Clin Exp Dent Res 2025; 11:e70084. [PMID: 39916439 PMCID: PMC11803075 DOI: 10.1002/cre2.70084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 01/05/2025] [Accepted: 01/24/2025] [Indexed: 02/11/2025] Open
Abstract
OBJECTIVES Opportunistic viruses such as cytomegalovirus (CMV) and Epstein-Barr virus (EBV) have been detected in gingival crevicular fluid (GCF) and saliva of patients with periodontal disease. However, the relationship between herpesviruses and periodontitis remains obscure. The aim of this case-control study was the detection and association of CMV and EBV with periodontitis. MATERIAL AND METHODS Forty-eight adults were included in this study: 24 patients with periodontitis (CP) and 24 periodontally healthy individuals (HS). All patients underwent periodontal examination, including probing depth (PD), clinical attachment loss (CAL), plaque index (PI), and bleeding on probing (BOP). Subgingival biofilm samples were collected from all patients and real-time PCR was performed for viral and bacterial detection. The odds ratio (OR) was calculated, and the chi-squared test or Fisher's exact test was performed to analyze the significant differences. RESULTS EBV was detected only in one healthy patient meanwhile no CMV was found. No statistically significant differences were found between the periodontal clinical parameters of EBV-positive patients and the negative ones: PI (p = 0.090), PD (p = 0.857), CAL (p = 0.801), and BOP (p = 0.104). Except for Aggregatibacter actinomycetemcomitans (Aa), Porphyromonas gingivalis (Pg), Tannerella forsythia (Tf), Prevotella intermedia (Pi), and Treponema denticola (Td) showed a statistically significant association (p < 0.001) with the clinical periodontal parameters. Aa presence was not statistically associated with periodontal sites (p < 0.296). Tf and Pg were the most frequently detected periodontopathic bacteria in the CP group (91.7% sites). CONCLUSION The present case-control study showed that the prevalence of EBV and CMV did not show significant differences between the groups evaluated in the Spanish population.
Collapse
Affiliation(s)
- Alicia Herrero‐Sánchez
- Private PracticeMadridSpain
- Faculty of DentistryComplutense University of Madrid (U.C.M)MadridSpain
| | | |
Collapse
|
33
|
D'Costa J, Chibo D, Soloczynskyj K, Batty M, Sameer R, Lee E, Tran T, Mavroulis D, Gooey M, Williams E, Jackson K. Evaluation and comparison of three high throughput assays (Alinity m CMV, Aptima CMV Quant and cobas CMV) for quantifying CMV DNA in plasma samples. J Virol Methods 2025; 332:115068. [PMID: 39551443 DOI: 10.1016/j.jviromet.2024.115068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 10/23/2024] [Accepted: 11/14/2024] [Indexed: 11/19/2024]
Abstract
BACKGROUND Cytomegalovirus (CMV) can cause symptomatic CMV syndrome or tissue-invasive CMV disease in immunocompromised individuals, including solid-organ transplant and hematopoietic stem cell transplant recipients. In these populations, monitoring of CMV load is essential, assessing both risk of disease and response to antiviral therapy. High throughput commercial assays are currently available for CMV quantitation, but they are often evaluated independently, with few studies comparing these assays. This study evaluated CMV quantitative assays for use with the Roche cobas 6800, Abbott Alinity m and Hologic Panther platforms using stored patient plasma. METHODS Analytical evaluation was performed using the 1st WHO international standard for human CMV for Nucleic Acid Amplification Techniques (cobas and Alinity m) or the Hologic CMV QC Calibrator 6 (Aptima). Parallel testing of 136 clinical plasma samples was performed across the three platforms. RESULTS Linearity for each assay ranged from 98.6 % to 99.96 % and precision and limit of quantitation were as expected with little variation between platforms. 136 clinical plasma samples were evaluated with similar agreement observed between each assay. The greatest positive agreement was between the Aptima Quant and Alinity m assays (95.6 %, 95 % CI 89-98.6 %) and the lowest between the Aptima Quant and cobas assays (94.1 %, 87.4-97.5 %). CONCLUSIONS All assays were sensitive and accurate when quantifying CMV, and performance across all 3 assays was comparable for monitoring CMV viral loads in patient plasma.
Collapse
Affiliation(s)
- Jodie D'Costa
- Victorian Infectious Diseases Reference Laboratory, Peter Doherty Institute for Infection and Immunity, 792 Elizabeth St, Melbourne 3000, Australia.
| | - Doris Chibo
- Victorian Infectious Diseases Reference Laboratory, Peter Doherty Institute for Infection and Immunity, 792 Elizabeth St, Melbourne 3000, Australia
| | - Katherine Soloczynskyj
- Victorian Infectious Diseases Reference Laboratory, Peter Doherty Institute for Infection and Immunity, 792 Elizabeth St, Melbourne 3000, Australia
| | - Mitchell Batty
- Victorian Infectious Diseases Reference Laboratory, Peter Doherty Institute for Infection and Immunity, 792 Elizabeth St, Melbourne 3000, Australia
| | - Rizmina Sameer
- Victorian Infectious Diseases Reference Laboratory, Peter Doherty Institute for Infection and Immunity, 792 Elizabeth St, Melbourne 3000, Australia
| | - Elaine Lee
- Victorian Infectious Diseases Reference Laboratory, Peter Doherty Institute for Infection and Immunity, 792 Elizabeth St, Melbourne 3000, Australia
| | - Thomas Tran
- Victorian Infectious Diseases Reference Laboratory, Peter Doherty Institute for Infection and Immunity, 792 Elizabeth St, Melbourne 3000, Australia
| | - Dimi Mavroulis
- Victorian Infectious Diseases Reference Laboratory, Peter Doherty Institute for Infection and Immunity, 792 Elizabeth St, Melbourne 3000, Australia
| | - Megan Gooey
- Victorian Infectious Diseases Reference Laboratory, Peter Doherty Institute for Infection and Immunity, 792 Elizabeth St, Melbourne 3000, Australia
| | - Eloise Williams
- Victorian Infectious Diseases Reference Laboratory, Peter Doherty Institute for Infection and Immunity, 792 Elizabeth St, Melbourne 3000, Australia
| | - Kathy Jackson
- Victorian Infectious Diseases Reference Laboratory, Peter Doherty Institute for Infection and Immunity, 792 Elizabeth St, Melbourne 3000, Australia
| |
Collapse
|
34
|
Arias A, Builes N, Niño-Serna L, Diaz A, Aristizabal BH, Trujillo M. CMV Reactivation Following Allogeneic Transplantation in Children From a High-Seroprevalence Population: A Single-Center Experience in Colombia. Pediatr Transplant 2025; 29:e70033. [PMID: 39837777 DOI: 10.1111/petr.70033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/23/2025]
Abstract
INTRODUCTION Cytomegalovirus (CMV) infection is a frequent complication among hematopoietic stem cell transplant (HSCT) recipients. Data regarding CMV reactivation in children in underdeveloped countries is scarce. This is especially notable considering the increasing utilization of haploidentical-related HSCT with the post-transplant cyclophosphamide platform. This study aimed to describe the incidence, clinical characteristics, and evolution of children with CMV reactivation after HSCT and the possible impact of unmanipulated stem cells with PTCy for GvHD prophylaxis. METHODS Retrospective cohort study of children undergoing hematopoietic stem cell transplantation from January 2012 to June 2022. Baseline characteristics and the clinical course were described. Duration of treatment, initial viral load, and time to clearance of DNAemia by type of transplant were compared using the Kruskal-Wallis test. Survival analysis was performed with the Kaplan-Meier method and log-rank test. All statistical analysis was performed using SPSS software, version 20.0. RESULTS One hundred sixty-six children were included. Among them, 87% of recipients and 88% of donors were CMV positive. The cumulative incidence of cytomegalovirus DNAemia was 28% at 100 days post-transplantation. There were no differences between different donor types. Overall survival at 1 year was 60%, and non-relapse mortality was observed in 28%. CMV reactivation did not appear to negatively impact 1-year overall survival (OS). CONCLUSIONS Our study found no differences in CMV reactivation rates, treatment duration, viral clearance times, co-infections, or 1-year overall survival across different HSCT donor types. Studies are needed to establish more precise criteria for monitoring recipients, particularly in regions where unmanipulated stem cells with PTCy for GvHD prophylaxis are increasing.
Collapse
Affiliation(s)
- Andres Arias
- Hospital Universitario Erasmo Meoz, Cúcuta, Colombia
| | | | | | - Alejandro Diaz
- Hospital Pablo Tobón Uribe, Medellín, Colombia
- Hospital General de Medellín, Medellín, Colombia
| | | | | |
Collapse
|
35
|
Cabanilla MG, Dauenhauer A, St John B, Hill D, Larson J. Taming the transplant troll: Exploring racial and ethnic disparities in cytomegalovirus infection among kidney transplant patients. PLoS One 2025; 20:e0317383. [PMID: 39879165 PMCID: PMC11778782 DOI: 10.1371/journal.pone.0317383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 12/26/2024] [Indexed: 01/31/2025] Open
Abstract
BACKGROUND Cytomegalovirus (CMV) infection poses a significant risk to kidney transplant recipients. This study investigated CMV disease incidence, outcomes, and management challenges in racial and ethnic minority populations following kidney transplantation. METHODS This single-center, mixed-methods study included a retrospective cohort analysis of kidney transplant recipients (n = 58) and qualitative surveys of healthcare providers. Patients were categorized as minorities (n = 49) or non-Hispanic whites (n = 9). The primary outcome was CMV disease incidence. Secondary outcomes included graft failure, mortality, and identification of management barriers. RESULTS The cumulative incidence of CMV disease was higher in minorities than in non-Hispanic whites (12.3% vs. 0%, p = 0.58), although the difference was not statistically significant. All graft failures (8.6%, n = 5) occurred in the minority group. Although not statistically significant, all-cause mortality was higher in the minority group (24.5% vs. 11.1%, p = 0.54), with 46.2% of the deaths occurring within 90 days of CMV diagnosis. Qualitative analysis revealed challenges in diagnosis, treatment-related side effects, medication costs, and insurance barriers. The providers emphasized the importance of interdisciplinary collaboration and standardized protocols. CONCLUSION While limited by the small sample size, this study highlights potential disparities in the incidence and outcomes of CMV disease among minority kidney transplant recipients, suggesting that barriers in care and access may contribute to these differences. These hypothesis-generating findings underscore the need for larger multicenter studies to validate these patterns and to inform targeted strategies that may reduce inequities in post-transplant outcomes.
Collapse
Affiliation(s)
- M. Gabriela Cabanilla
- Division of Infectious Diseases, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, United States of America
- Department of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, United States of America
| | - Ashlee Dauenhauer
- Department of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, United States of America
- Division of Transplant Nephrology, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, United States of America
| | - Briana St John
- Department of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, United States of America
- Division of Transplant Nephrology, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, United States of America
| | - Deirdre Hill
- University of New Mexico School of Medicine, Albuquerque, NM, United States of America
| | - Joshua Larson
- Division of Transplant Nephrology, Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, United States of America
| |
Collapse
|
36
|
Manska S, Hagemann A, Magana J, Rossetto CC, Verma SC. Characterization of Human Cytomegalovirus (HCMV) Long Non-Coding RNA1.2 During Lytic Replication. Viruses 2025; 17:149. [PMID: 40006904 PMCID: PMC11860937 DOI: 10.3390/v17020149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/17/2025] [Accepted: 01/18/2025] [Indexed: 02/27/2025] Open
Abstract
During lytic replication of human cytomegalovirus (HCMV), the most abundant viral transcripts are long non-coding RNAs (lncRNAs). Viral lncRNAs can have a variety of functions, some of which are necessary for viral production and the modulation of host processes during infection. HCMV produces four lncRNAs, Beta2.7 (RNA2.7), RNA4.9, RNA5.0 and RNA1.2. While there has been research on these viral lncRNAs, many of their functions remain uncharacterized. To determine the function of RNA1.2, we explored its requirement during lytic infection by generating viral mutants containing either a full or partial deletion of the RNA1.2 locus. Within permissive fibroblasts, the RNA1.2 deletion mutants showed no defects in viral DNA synthesis, transcript expression, protein production, or generation of viral progeny. Further investigation to identify potential cellular and viral protein binding partners of RNA1.2 was performed using liquid chromatography-mass spectrometry (LC-MS). A significant number of cellular proteins were identified and associated with RNA1.2. Specifically those associated with the innate immune response, mitochondrial processes, and cell cycle regulation. While RNA1.2 is dispensable for lytic replication, these findings suggest it may play a pivotal role in modulating the host response.
Collapse
Affiliation(s)
| | | | | | | | - Subhash C. Verma
- Department of Microbiology and Immunology, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA; (S.M.); (A.H.); (J.M.); (C.C.R.)
| |
Collapse
|
37
|
Manuel T, Moström M, Crooks CM, Davalos A, Barfield R, Scheef E, Kendall S, Midkiff CC, Sprehe L, Trexler M, Boquet F, Shroyer M, Danner V, Doyle-Myers L, Weinbaum C, Mirza A, Lammi S, Otero C, Lee MR, Rogers LW, Granek J, Owzar K, Malouli D, Früh K, Kowalik T, Chan C, Permar SR, Blair RV, Kaur A. A nonhuman primate model mirrors human congenital cytomegalovirus infection and reveals a spectrum of vertical transmission outcomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.16.633406. [PMID: 40492197 PMCID: PMC12148051 DOI: 10.1101/2025.01.16.633406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2025]
Abstract
Congenital cytomegalovirus (cCMV) is the leading infectious cause of birth defects worldwide, yet immune determinants of protection to inform design of a maternal vaccine remain elusive. Here, we characterized the outcome of primary rhesus CMV (RhCMV) infection during pregnancy in an immune competent nonhuman primate (NHP) model. RhCMV DNA was detected in amniotic fluid and/or fetal tissues in six of 12 (50% placental transmission) CMV-naive rhesus macaque dams inoculated intravenously with RhCMV in early second trimester gestation. Widespread tissue dissemination dominated by one of two inoculated RhCMV strains was present in one fetus (8.3% cCMV disease). Placental RhCMV transmission was associated with elevated fetal and maternal plasma TNF-alpha and reduced maternal brain-derived neurotrophic factor and IL-10 levels. CMV exposure during pregnancy had a broad impact on the placenta and fetus even in the absence of congenital infection as evidenced by RhCMV infection at the maternal-fetal interface in all 12 dams, along with significantly reduced placental efficiency and fetal growth metrics compared to gestation-matched control pregnancies. This NHP model recapitulates key aspects of human cCMV and provides new insight into barriers and biomarkers of successful vertical transmission. One sentence summary: The nonhuman primate model mirrors the epidemiology of human congenital CMV (cCMV) after primary infection and reveals its transmission bottlenecks.
Collapse
|
38
|
Schultz EP, Ponsness L, Lanchy JM, Zehner M, Klein F, Ryckman BJ. Human cytomegalovirus gH/gL/gO binding to PDGFRα provides a regulatory signal activating the fusion protein gB that can be blocked by neutralizing antibodies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.08.631902. [PMID: 39829861 PMCID: PMC11741351 DOI: 10.1101/2025.01.08.631902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Herpesviruses require membrane fusion for entry and spread, a process facilitated by the fusion glycoprotein B (gB) and the regulatory factor gH/gL. The human cytomegalovirus (HCMV) gH/gL can be modified by the accessory protein gO, or the set of proteins UL128, UL130 and UL131. While the binding of the gH/gL/gO and gH/gL/UL128-131 complexes to cellular receptors including PDFGRα and NRP2 has been well-characterized structurally, the specific role of receptor engagements by the gH/gL/gO and gH/gL/UL128-131 in regulation of fusion has remained unclear. We describe a cell-cell fusion assay that can quantitatively measure fusion on a timescale of minutes and demonstrate that binding of gH/gL/gO to PDGFRα dramatically enhances gB-mediated cell-cell fusion. In contrast, gH/gL/pUL128-131-regulated fusion is significantly slower and gH/gL alone cannot promote gB fusion activity within this timescale. The genetic diversity of gO influenced the observed cell-cell fusion rates, correlating with previously reported effects on HCMV infectivity. Mutations in gL that had no effect on formation of gH/gL/gO or binding to PDGFRα dramatically reduced the cell-cell fusion rate, suggesting that gL plays a critical role in linking the gH/gL/gO-PDGFRα receptor-binding to activation of gB. Several neutralizing human monoclonal antibodies were found to potently block gH/gL/gO-PDGFRα regulated cell-cell fusion, suggesting this mechanism as a therapeutic target.
Collapse
Affiliation(s)
- Eric P. Schultz
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
- Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT 59812, USA
| | - Lars Ponsness
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
| | - Jean-Marc Lanchy
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
- Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT 59812, USA
| | - Matthias Zehner
- Laboratory for Infection and Immune Biology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Florian Klein
- Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Brent J. Ryckman
- Division of Biological Sciences, University of Montana, Missoula, MT 59812, USA
- Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT 59812, USA
| |
Collapse
|
39
|
Behrens M, Comabella M, Lünemann JD. EBV-specific T-cell immunity: relevance for multiple sclerosis. Front Immunol 2024; 15:1509927. [PMID: 39776919 PMCID: PMC11703957 DOI: 10.3389/fimmu.2024.1509927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Genetic and environmental factors jointly determine the susceptibility to develop multiple sclerosis (MS). Improvements in the design of epidemiological studies have helped to identify consistent environmental risk associations such as the increased susceptibility for MS following Epstein-Barr virus (EBV) infection, while biological mechanisms that drive the association between EBV and MS remain incompletely understood. An increased and broadened repertoire of antibody and T-cell immune responses to EBV-encoded antigens, especially to the dominant CD4+ T-cell EBV nuclear antigen 1 (EBNA1), is consistently observed in patients with MS, indicating that protective EBV-specific immune responses are deregulated in MS and potentially contribute to disease development. Exploitation of B-cell trajectories by EBV infection might promote survival of autoreactive B-cell species and proinflammatory B:T-cell interactions. In this review article, we illustrate evidence for a causal role of EBV infection in MS, discuss how EBV-targeting adaptive immune responses potentially modulate disease susceptibility and progression, and provide future perspectives on how novel model systems could be utilized to better define the role of EBV and viral pathogens in MS. Insights gained from these studies might facilitate the development of prevention strategies and more effective treatments for MS.
Collapse
Affiliation(s)
- Malina Behrens
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Manuel Comabella
- Servei de Neurologia-Neuroimmunologia, Centre d’Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d’Hebron (VHIR), Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Vall d’Hebron University Hospital, Barcelona, Spain
| | - Jan D. Lünemann
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| |
Collapse
|
40
|
Touhami S, Toutée A, Chung YR, Touitou V, Bodaghi B. [Viral retinitis]. J Fr Ophtalmol 2024; 47:104341. [PMID: 39515032 DOI: 10.1016/j.jfo.2024.104341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/10/2024] [Accepted: 07/12/2024] [Indexed: 11/16/2024]
Abstract
Viruses belonging to the herpes family group, including HSV (herpes simplex virus) 1 and 2, VZV (varicella zoster virus) and CMV (cytomegalovirus) are the leading causes of necrotizing retinitis. These viral retinal necroses generally manifest in three forms, depending on the patient's immune status: acute retinal necrosis (ARN), progressive outer retinal necrosis (PORN) and CMV retinitis. Although specific, effective drug treatments are available today, early treatment initiation is essential to avoid sight-threatening complications.
Collapse
Affiliation(s)
- S Touhami
- Service d'ophtalmologie, Sorbonne université, CHU Pitié-Salpêtrière, Paris, France; Inserm, CNRS, institut de la vision, Sorbonne université, Paris, France.
| | - A Toutée
- Service d'ophtalmologie, Sorbonne université, CHU Pitié-Salpêtrière, Paris, France
| | - Y-R Chung
- Service d'ophtalmologie, Sorbonne université, CHU Pitié-Salpêtrière, Paris, France; Department of Ophthalmology, Ajou University School of Medicine, Suwon, République de Corée
| | - V Touitou
- Service d'ophtalmologie, Sorbonne université, CHU Pitié-Salpêtrière, Paris, France
| | - B Bodaghi
- Service d'ophtalmologie, Sorbonne université, CHU Pitié-Salpêtrière, Paris, France
| |
Collapse
|
41
|
Hsu FM, Mohanty RP, Rubbi L, Thompson M, Pickering H, Reed EF, Greenland JR, Schaenman JM, Pellegrini M. An epigenetic human cytomegalovirus infection score predicts viremia risk in seropositive lung transplant recipients. Epigenetics 2024; 19:2408843. [PMID: 39360678 PMCID: PMC11451273 DOI: 10.1080/15592294.2024.2408843] [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: 03/21/2024] [Revised: 08/28/2024] [Accepted: 09/04/2024] [Indexed: 10/04/2024] Open
Abstract
Cytomegalovirus (CMV) infection and reactivation in solid organ transplant (SOT) recipients increases the risk of viremia, graft failure and death. Clinical studies of CMV serostatus indicate that donor positive recipient negative (D+/R-) patients have greater viremia risk than D-/R-. The majority of patients are R+ having intermediate serologic risk. To characterize the long-term impact of CMV infection and assess viremia risk, we sought to measure the effects of CMV on the recipient immune epigenome. Specifically, we profiled DNA methylation in 156 individuals before lung or kidney transplant. We found that the methylome of CMV positive SOT recipients is hyper-methylated at loci associated with neural development and Polycomb group (PcG) protein binding, and hypo-methylated at regions critical for the maturation of lymphocytes. In addition, we developed a machine learning-based model to predict the recipient CMV serostatus after correcting for cell type composition and ancestry. This CMV episcore measured at baseline in R+ individual stratifies viremia risk accurately in the lung transplant cohort, and along with serostatus the CMV episcore could be a potential biomarker for identifying R+ patients at high viremia risk.
Collapse
Affiliation(s)
- Fei-Man Hsu
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
- Institute for Quantitative and Computational Biosciences – The Collaboratory, University of California Los Angeles, Los Angeles, CA, USA
| | - Rashmi P. Mohanty
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Liudmilla Rubbi
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | - Michael Thompson
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | - Harry Pickering
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Elaine F. Reed
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - John R. Greenland
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Joanna M. Schaenman
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
- Institute for Quantitative and Computational Biosciences – The Collaboratory, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
42
|
Luo X, Quan L, Lin Q, Rong H, Liu Y, Meng J, You X. Integrating clinical data and genetic susceptibility to elucidate the relationship between systemic lupus erythematosus and human cytomegalovirus infection. Virol J 2024; 21:311. [PMID: 39614342 PMCID: PMC11607911 DOI: 10.1186/s12985-024-02578-6] [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: 01/08/2024] [Accepted: 11/12/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND Viral infections are known to induce the occurrence and pathogenesis of systemic lupus erythematosus (SLE). Previous studies have indicated a possible relationship between SLE and human cytomegalovirus (HCMV) infection and have attributed HCMV to be associated with various autoantibodies; however, these studies were constrained by variations in sample size and potential selection bias. Therefore, in the present study, we aimed to elucidate the relationship between HCMV and autoantibodies in patients with SLE by integrating clinical data and genetic susceptibility. METHODS Using various statistical methods, we conducted a retrospective analysis of the spectrum of SLE autoantibodies and HCMV infections among patients hospitalized at our center over the past 10 years. Machine learning modeling was used to predict active HCMV infections based on the antinuclear (ANA) spectrum. Moreover, Mendelian randomization (MR) was used to investigate the causal relationship between SLE and HCMV infection. RESULTS In the HCMV group, the levels of ANA, anti-dsDNA, anti-histone antibody (AHA), and anti-nucleosome antibody (ANuA) were significantly increased (P < 0.001) and were linked to the presence of CMV-pp65-antigen-positive polymorphonuclear leukocytes (P < 0.001). A weak correlation was observed between the titers of anti-CMV IgM and ANA (P < 0.001). The ANA spectrum demonstrated a strong predictive performance for active HCMV infection based on principal component analysis (Adonis and ANOSIM P < 0.001) as well as support vector machine and extreme gradient boosting modeling. MR analyses of inverse-variance weighted, weighted mean, MR-Egger, and weighted mode revealed that patients with SLE were at a higher risk of developing HCMV infection (P < 0.05). However, HCMV infection did not have a causal effect on SLE (P > 0.05). CONCLUSION The ANA spectrum in patients with SLE can be used to predict HCMV infection status. Due to the inherent susceptibility of patients with SLE to HCMV infection, we propose for the first time that if a patient with SLE exhibits high serum titers of ANA, anti-dsDNA, ANuA, and AHA, caution should be exercised for HCMV infection, which can contribute to the clinical assessment of SLE and improve patient prognosis.
Collapse
Affiliation(s)
- Xin Luo
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Key Laboratory of Rheumatology & Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Ministry of Education, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, China
| | - Liuliu Quan
- Department of Medical Oncology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qingting Lin
- Department of Emergency, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Huiteng Rong
- University of International Business and Economics, Beijing, China
| | - Yue Liu
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Jiaqi Meng
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Xin You
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Key Laboratory of Rheumatology & Clinical Immunology, Chinese Academy of Medical Sciences & Peking Union Medical College, Ministry of Education, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Beijing, China.
| |
Collapse
|
43
|
García-Masedo Fernández S, Laporta R, García Fadul C, Aguilar Pérez M, Anel Pedroche J, Sanabrias Fernández de Sevilla R, Royuela A, Sánchez Romero I, Ussetti Gil MP. CMV Infection Risk Factors and Viral Dynamics After Valganciclovir Prophylaxis: 10 Years of Experience in Lung Transplant Recipients. Microorganisms 2024; 12:2360. [PMID: 39597748 PMCID: PMC11596771 DOI: 10.3390/microorganisms12112360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/02/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024] Open
Abstract
(1) The prevention of cytomegalovirus (CMV) in lung transplant recipients (LTx) is based on the administration of VGC for a period of 6-12 months, but there is little information on the premature discontinuation of the drug. Our objective was to evaluate the reasons for early cessation of VGC and the dynamics of CMV replication after discontinuation. (2) We carried out a retrospective study of LTx on VGC prophylaxis according to guidelines, with an outpatient follow-up period of >90 days. The detection of any level of CMV-DNA in the plasma (Cobas, Roche Diagnostics®) during a period of 18 months after the discontinuation of VGC was considered positive. (3) We included 312 patients (64% male, mean age 53.50 ± 12.27; 71% D+R+, 15% D-R+, and 14% D+R-) in our study. The prescribed prophylaxis was completed by 179 patients (57.05%). The mean duration of prophylaxis was 7.17 ± 1.08 months. The recorded reasons for VGC discontinuation in 133 patients (43%) were myelotoxicity (n = 55), impaired renal function (n = 32), and gastrointestinal disturbances (n = 11). The reason for discontinuation was not recorded for 29 patients. CMV-DNA was detected in 79% (n = 246) of cases, and D+R+ and D+R- recipients showed a high risk of detection (p < 0.001). The median times to onset of CMV-DNA detection were 35 days in D+R-, 73 days in D+R+, and 96 days in D-R+ (p < 0.001). (4) Adverse effects of VGC are frequent in LTx. CMV-DNA detection is very common after the discontinuation of VGC and is related to the CMV donor and recipient serostatus.
Collapse
Affiliation(s)
| | - Rosalía Laporta
- Pneumology Department, Hospital Universitario Puerta de Hierro, 28222 Majadahonda, Spain (M.P.U.G.)
| | - Christian García Fadul
- Pneumology Department, Hospital Universitario Puerta de Hierro, 28222 Majadahonda, Spain (M.P.U.G.)
| | - Myriam Aguilar Pérez
- Pneumology Department, Hospital Universitario Puerta de Hierro, 28222 Majadahonda, Spain (M.P.U.G.)
| | - Jorge Anel Pedroche
- Microbiology Department, Hospital Universitario Puerta de Hierro, 28222 Majadahonda, Spain
| | | | - Ana Royuela
- Clinical Biostatistics Unit, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana, 28222 Madrid, Spain
| | - Isabel Sánchez Romero
- Microbiology Department, Hospital Universitario Puerta de Hierro, 28222 Majadahonda, Spain
| | - María Piedad Ussetti Gil
- Pneumology Department, Hospital Universitario Puerta de Hierro, 28222 Majadahonda, Spain (M.P.U.G.)
| |
Collapse
|
44
|
James SA, Joshua IA. Charting Peptide Shared Sequences Between 'Diabetes-Viruses' and Human Pancreatic Proteins, Their Structural and Autoimmune Implications. Bioinform Biol Insights 2024; 18:11779322241289936. [PMID: 39502449 PMCID: PMC11536397 DOI: 10.1177/11779322241289936] [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: 11/22/2023] [Accepted: 08/21/2024] [Indexed: 11/08/2024] Open
Abstract
Diabetes mellitus (DM) is a metabolic syndrome characterized by hyperglycaemia, polydipsia, polyuria, and weight loss, among others. The pathophysiology for the disorders is complex and results in pancreatic abnormal function. Viruses have also been implicated in the metabolic syndrome. This study charted peptides to investigate and predict the autoimmune potential of shared sequences between 8 viral species proteins (which we termed 'diabetes-viruses') and the human pancreatic proteins. The structure and immunological relevance of shared sequences between viruses reported in DM onset and human pancreatic proteins were analysed. At nonapeptide mapping between human pancreatic protein and 'diabetic-viruses', reveal 1064 shared sequences distributed among 454 humans and 4288 viral protein sequences. The viral results showed herpesviruses, enterovirus (EV), human endogenous retrovirus, influenza A viruses, rotavirus, and rubivirus sequences are hosted by the human pancreatic protein. The most common shared nonapeptide was AAAAAAAAA, present in 30 human nonredundant sequences. Among the viral species, the shared sequence NSLEVLFQG occurred in 18 nonredundant EVs protein, while occurring merely in 1 human protein, whereas LGLDIEIAT occurred in 8 influenza A viruses overlapping to 1 human protein and KDELSEARE occurred in 2 rotaviruses. The prediction of the location of the shared sequences in the protein structures, showed most of the shared sequences are exposed and located either on the surface or cleft relative to the entire protein structure. Besides, the peptides in the viral protein shareome were predicted computationally for binding to MHC molecules. Here analyses showed that the entire 1064 shared sequences predicted 203 to be either HLA-A or B supertype-restricted epitopes. Fifty-one of the putative epitopes matched reported HLA ligands/T-cell epitopes majorly coming from EV B supertype representative allele restrictions. These data, shared sequences, and epitope charts provide important insight into the role of viruses on the onset of DM and its implications.
Collapse
Affiliation(s)
- Stephen A James
- Department of Biochemistry, Kaduna State University, Kaduna, Nigeria
- School of Data Sciences, Centre of Bioinformatics, Perdana University, Kuala Lumpur, Malaysia
| | - Istifanus A Joshua
- Department of Community Medicine, College of Medicine, Kaduna State University, Kaduna, Nigeria
- Department of Community Medicine, College of Health Sciences, Federal University Wukari, Wukari, Nigeria
| |
Collapse
|
45
|
Supplisson O, Visseaux B, Haim-Boukobza S, Boutolleau D, Alizon S, Burrel S, Sofonea MT. Seroprevalence of human herpes viruses in France, 2018-2022: a multilevel regression and poststratification approach. Infect Dis (Lond) 2024; 56:931-945. [PMID: 38946531 DOI: 10.1080/23744235.2024.2365906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 07/02/2024] Open
Abstract
BACKGROUND Information related to herpes simplex virus 1 and 2 (HSV-1 and 2), varicella-zoster virus (VZV), Epstein-Barr virus (EBV), and cytomegalovirus (CMV) seroprevalence in France is either lacking, incomplete, or outdated, despite their public health burden. METHOD We used routinely collected serological data between 2018 and 2022 to estimate HSV-1, HSV-2, VZV, EBV, and CMV seroprevalence in France. To account for demographic differences between our analytic samples and the French population and get estimates for sparsely sampled districts and age classes, we used a multilevel regression and poststratification approach combined with Bayesian model averaging via stacking weights. RESULTS The observed seroprevalence (number of positive tests/number of tests) were 64.6% (93,294/144,424), 16.9% (24,316/144,159), 93.0% (141,419/152,084), 83.4% (63,199/75, 781), and 49.0% (23,276/47,525), respectively, for HSV-1, HSV-2, VZV, EBV, and CMV. Between 2018 and 2022, France had a model-based average (equal-tailed interval at 95%) expected seroprevalence equal to 61.1% (60.7,61.5), 14.5% (14.2,14.81), 89.5% (89.3,89.8), 85.6% (85.2,86.0), and 50.5% (49.3,51.7), respectively, for HSV-1, HSV-2, VZV, EBV, and CMV infections. We found an almost certain lower expected seroprevalence in Metropolitan France than in overseas territories for all viruses but VZV, for which it was almost certainly greater. The expected seroprevalences were likely greater among females for all viruses. LIMITATIONS Our results relied on the assumption that individuals were sampled at random conditionally to variables used to build the poststratification table. IMPLICATIONS The analysis highlights spatial and demographic patterns in seroprevalence that should be considered for designing tailored public health policies.
Collapse
Affiliation(s)
- Olivier Supplisson
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, Paris, France
- Sorbonne Université, Paris, France
| | | | | | - David Boutolleau
- AP-HP, Sorbonne Université, Centre National de Référence Herpèsvirus (laboratoire Associé), Service de Virologie, Hôpital Pitié-Salpêtrière, Paris, France
- Sorbonne Université, INSERM UMR-S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique (iPLESP), Paris, France
| | - Samuel Alizon
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, Paris, France
| | - Sonia Burrel
- CHU de Bordeaux, Service de virologie, Bordeaux, France
- CNRS UMR 5234, Fundamental Microbiology and Pathogenicity, Université de Bordeaux, Bordeaux, France
| | - Mircea T Sofonea
- Pathogenesis and Control of Chronic and Emerging Infections (PCCEI), Université de Montpellier, Inserm, EFS, Montpellier, France and CHU de Nîmes, Nîmes, France
| |
Collapse
|
46
|
Inaam N, Othman S, Fryad H, Faqi S. Seroprevalence and Molecular Detection of Cytomegalovirus UL146 and US28 Gene Expression in Women With Recurrent Pregnancy Loss. Cureus 2024; 16:e73039. [PMID: 39640126 PMCID: PMC11620713 DOI: 10.7759/cureus.73039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
Background Human cytomegalovirus (CMV) is a global herpesvirus that is highly prevalent worldwide and is able to establish lifelong latency after initial infection. The infection is highly frequent during pregnancy in human beings, which leads to preterm birth in some cases. Circulating strains of CMV carry a high number of variable or disrupted genes. Some of these like UL146, a highly diverse gene, and the US28 gene are involved in viral dissemination. This study aims to determine the seroprevalence of CMV and to investigate whether the highly variable UL146 and US28 genes, isolated from the blood of seropositive women, are associated with recurrent pregnancy loss. Material and methods This cross-sectional study was carried out in Erbil City, Iraq from October 2022 to July 2023. A total of 150 women at their reproductive age with a history of miscarriage who attended Maternity Teaching Hospital were enrolled. Anti-CMV IgG and IgM antibodies were assessed by enzyme-linked immunosorbent assay (ELISA). Highly variable UL146 and US28 genes of CMV from seropositive samples were amplified by conventional polymerase chain reaction (PCR), and the results were visualized on a UV-transilluminator. SPSS version 22 (IBM Corp., Armonk, NY, USA) was used for data entry and analysis. The p-value less than 0.05 was regarded as statistically significant. Results Anti-CMV IgG and IgM were seropositive in 103 (53.3%) and 13 (8.7%) women, respectively, and only 10 (6.7%) of them for both anti-CMV IgG and IgM. Significant associations of CMV and history of miscarriage, age, educational level, and gestational age of miscarriages were observed (p-value less than 0.05). On the other hand, no statistically significant association between CMV and socioeconomic level or residency was observed. The frequencies of genetic analysis of UL146 and US28 of the 103 seropositive tested samples of women with a history of miscarriage were 31 (30.1%) and nine (8.7%), respectively. A significant association between recurrent miscarriage and UL146 gene expression was observed. PCR targeting the UL146 demonstrated greater sensitivity for diagnosing CMV. Conclusion The seroprevalence of CMV is relatively high in Erbil, and the UL146 and US28 genes can act as factors in the initial level of CMV. Therefore, molecular detection of these genes can aid in determining the virulence of CMV strains.
Collapse
Affiliation(s)
- Niyan Inaam
- Basic Medical Science, College of Medicine of Hawler Medical University, Erbil, IRQ
| | - Samir Othman
- Community Medicine, College of Medicine of Hawler Medical University, Erbil, IRQ
| | - Hataw Fryad
- Medical Laboratory Science, College of Health Sciences, Lebanese French University, Erbil, IRQ
| | - Shler Faqi
- Basic Medical Science, College of Medicine, University of Sulaimani, Erbil, IRQ
| |
Collapse
|
47
|
Li Y, Liu Y, Bu X, Qin Y, Zhang Y. Research progress on V delta 1 + T cells and their effect on pathogen infection. PeerJ 2024; 12:e18313. [PMID: 39494290 PMCID: PMC11531252 DOI: 10.7717/peerj.18313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 09/24/2024] [Indexed: 11/05/2024] Open
Abstract
The ongoing high occurrence of harmful infectious diseases significantly threatens human health. Existing methods used to control such diseases primarily involve targeting the pathogens, usually neglecting the vital role of host factors in disease advancement. Gamma delta (γδ) T cells act as a bridge between innate and adaptive immunity, playing a crucial role in combating pathogen invasion. Among these γδT cell subsets, which are categorized based on T cell receptor delta variable expression patterns, V delta (δ) 1+ T cells possess unique recognition abilities and regulatory characteristics and actively engage in various immune responses. The differentiation, development, and immune reactivity of Vδ1+ T cells are closely associated with the initial and progressive stages of infectious diseases. This article provides an overview of the classification, distribution, differentiation, and development of Vδ1+ T cells and their mechanisms in combating pathogenic infections, offering new insights for disease diagnosis and treatment.
Collapse
Affiliation(s)
- Yuxia Li
- School of Basic Medical Sciences, Shandong Second Medical University, Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, Weifang, Shandong, China
| | - Yanfei Liu
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong, China
| | - Xiaoxiao Bu
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong, China
| | - Yuanyuan Qin
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong, China
| | - Yanyan Zhang
- Department of Rheumatology and Immunology, Weifang Second People’s Hospital, Weifang, Shandong, China
| |
Collapse
|
48
|
Gomes Torres ACMB, Mathias C, Baal SCS, Kohler AF, Cunha ML, Blanes L. Advancements in LAMP-Based Diagnostics: Emerging Techniques and Applications in Viral Detection with a Focus on Herpesviruses in Transplant Patient Management. Int J Mol Sci 2024; 25:11506. [PMID: 39519059 PMCID: PMC11546353 DOI: 10.3390/ijms252111506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Loop-mediated isothermal amplification (LAMP) is a highly effective molecular diagnostic technique, particularly advantageous for point-of-care (POC) settings. In recent years, LAMP has expanded to include various adaptations such as DARQ-LAMP, QUASR, FLOS-LAMP, displacement probes and molecular beacons. These methods enable multiplex detection of multiple targets in a single reaction, enhancing cost-effectiveness and diagnostic efficiency. Consequently, LAMP has gained significant traction in diagnosing diverse viruses, notably during the COVID-19 pandemic. However, its application for detecting Herpesviridae remains relatively unexplored. This group of viruses is of particular interest due to their latency and potential reactivation, crucial for immunocompromised patients, including organ and hematopoietic stem cell transplant recipients. This review highlights recent advancements in LAMP for virus diagnosis and explores current research trends and future prospects, emphasizing the detection challenges posed by Herpesviridae.
Collapse
Affiliation(s)
| | - Carolina Mathias
- Post-Graduation Program in Genetics, Department of Genetics, Federal University of Parana, Curitiba 81530-980, Brazil; (C.M.); (S.C.S.B.); (A.F.K.); (M.L.C.)
| | - Suelen Cristina Soares Baal
- Post-Graduation Program in Genetics, Department of Genetics, Federal University of Parana, Curitiba 81530-980, Brazil; (C.M.); (S.C.S.B.); (A.F.K.); (M.L.C.)
| | - Ana Flávia Kohler
- Post-Graduation Program in Genetics, Department of Genetics, Federal University of Parana, Curitiba 81530-980, Brazil; (C.M.); (S.C.S.B.); (A.F.K.); (M.L.C.)
| | - Mylena Lemes Cunha
- Post-Graduation Program in Genetics, Department of Genetics, Federal University of Parana, Curitiba 81530-980, Brazil; (C.M.); (S.C.S.B.); (A.F.K.); (M.L.C.)
| | - Lucas Blanes
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba 81350-010, Brazil;
| |
Collapse
|
49
|
Flores-Medina S, Figueroa Damian R, Arreola-Ramírez G, Plazola-Camacho N, Villeda-Gabriel G, Ochoa SA, Cruz-Córdova A, Xicohtencatl-Cortes J, Arellano-Galindo J. Severe Cytomegalovirus Congenital Infection With Neurological Compromise a Case Series Study in Mexico: Severe CMV and Neurological Compromise. Case Rep Infect Dis 2024; 2024:7510447. [PMID: 39449816 PMCID: PMC11502122 DOI: 10.1155/2024/7510447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 07/02/2024] [Accepted: 08/22/2024] [Indexed: 10/26/2024] Open
Abstract
Four cases of serious congenital cytomegalovirus (CMV) infections are described in this report. All cases were diagnosed postnatally using cerebrospinal fluid (3/4) or blood PCR (1/4) and histochemical study of the placenta (4/4). All infants were born prematurely. Maternal factors identified as significant were younger age at pregnancy and those from low-income social strata. The major clinical findings among patients with congenital CMV infection were hydrocephalus and persistent thrombocytopenia. The children's clinical condition did not improve over the course of the disease, leading to complications associated with extreme prematurity. Two of the children died, one of whom had severe brain malformations and showed neurological compromise at follow-up, seizures, motor impairment, and severe cognitive delay. It is essential to perform antenatal screening for possible CMV infection among pregnant women, even in countries with high population seropositivity, such as Mexico, to establish prenatal interventions to reduce the risk of fetal damage.
Collapse
Affiliation(s)
- Saúl Flores-Medina
- Departamento de Infectología e Inmunología, Instituto Nacional de Perinatología, Ciudad de México, Mexico
- CECyT 15 “DAE”, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Ricardo Figueroa Damian
- Departamento de Infectología e Inmunología, Instituto Nacional de Perinatología, Ciudad de México, Mexico
| | - Gabriela Arreola-Ramírez
- Departamento de Seguimiento Pediátrico, Instituto Nacional de Perinatología, Ciudad de México, Mexico
| | - Noemi Plazola-Camacho
- Departamento de Infectología e Inmunología, Instituto Nacional de Perinatología, Ciudad de México, Mexico
| | - Graciela Villeda-Gabriel
- Departamento de Infectología e Inmunología, Instituto Nacional de Perinatología, Ciudad de México, Mexico
| | - Sara A. Ochoa
- Laboratorio de Investigación en Bacteriología Intestinal, Hospital Infantil de Mexico Federico Gómez, Ciudad de México, Mexico
| | - Ariadnna Cruz-Córdova
- Laboratorio de Inmunoquimica, Hospital Infantil de Mexico Federico Gómez, Ciudad de México, Mexico
| | - Juan Xicohtencatl-Cortes
- Laboratorio de Investigación en Bacteriología Intestinal, Hospital Infantil de Mexico Federico Gómez, Ciudad de México, Mexico
| | - José Arellano-Galindo
- Unidad de Investigación en Enfermedades Infecciosas, Hospital Infantil de México Federico Gómez, Ciudad de México, Mexico
- Departamento de Medicina, Centro Interdisciplinario de Ciencias de la Salud Unidad Milpa Alta, Instituto Politécnico Nacional, Ciudad de México, Mexico
| |
Collapse
|
50
|
Balegamire SJ, Mâsse B, Audibert F, Lamarre V, Giguere Y, Forest JC, Boucoiran I. Association Between Maternal Cytomegalovirus Seropositivity, Preterm Birth, and Preeclampsia in Two Cohorts From Quebec, Canada: A Mediation Analysis. Am J Reprod Immunol 2024; 92:e13941. [PMID: 39436114 DOI: 10.1111/aji.13941] [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: 06/21/2024] [Revised: 08/14/2024] [Accepted: 09/23/2024] [Indexed: 10/23/2024] Open
Abstract
PROBLEM Preterm birth and preeclampsia significantly contribute to infant morbidity and mortality, posing critical public health concerns. Viral infections, particularly Cytomegalovirus (CMV), associated with chronic inflammation, may play a role in these adverse pregnancy outcomes. The contribution of CMV to preterm birth and preeclampsia requires further investigation. METHOD OF STUDY Data from 6048 pregnant women from two prospective Quebec cohorts, recruited between May 2005 and August 2012, were analyzed. First-trimester CMV serology was the exposure variable. Associations were assessed using multivariable logistic regression adjusted by inverse probability treatment weighting (IPTW) of propensity scores. Mediation analyses estimated the direct effect of maternal CMV serostatus on preterm birth, excluding mediation by preeclampsia. RESULTS Preterm birth and preeclampsia proportions were 5.1% (95% CI: 4.6-5.7) and 1.9% (95% CI: 1.6-2.3), respectively. Multivariable logistic regression adjusted by IPTW showed associations between CMV seropositivity and preterm birth (OR 1.20, 95% CI: 1.02-1.41) and CMV seropositivity and preeclampsia (OR 1.41, 95% CI: 1.08-1.84). Mediation analysis indicated that 97% of the total effect of CMV seropositivity on preterm birth is direct, with the remaining 3% mediated by preeclampsia. CONCLUSIONS CMV seropositivity appears to be a risk factor for both preterm birth and preeclampsia. The effect of maternal CMV seropositivity on preterm birth is primarily direct, not mediated by preeclampsia. Future studies should explore the impact of preventive measures against CMV infection on the incidence of preterm delivery and preeclampsia.
Collapse
Affiliation(s)
- Safari Joseph Balegamire
- Department of Social and Preventive Medicine, École de Santé Publique de Université de Montréal, Montreal, Quebec, Canada
- Women and Children's Infectious Diseases Center, CHU Sainte-Justine Research Center, Montreal, Canada
| | - Benoît Mâsse
- Department of Social and Preventive Medicine, École de Santé Publique de Université de Montréal, Montreal, Quebec, Canada
- Applied Clinical Research Unit, CHU Sainte Justine Research Center, Montreal, Canada
| | - François Audibert
- Department of Obstetrics and Gynecology, Division of Maternofetal Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Valerie Lamarre
- Department of Obstetrics and Gynecology, Division of Maternofetal Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Yves Giguere
- CHU de Québec-Université Laval Research Center, Quebec City, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Jean-Claude Forest
- CHU de Québec-Université Laval Research Center, Quebec City, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Isabelle Boucoiran
- Department of Social and Preventive Medicine, École de Santé Publique de Université de Montréal, Montreal, Quebec, Canada
- Women and Children's Infectious Diseases Center, CHU Sainte-Justine Research Center, Montreal, Canada
- Department of Obstetrics and Gynecology, Division of Maternofetal Medicine, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|