1
|
Jeong NY, Cho H, Kim HJ, Choi NK. A broad assessment of rotavirus vaccine safety in infants in Korea: Insights from a data-driven signal detection approach. Hum Vaccin Immunother 2025; 21:2465161. [PMID: 39936376 PMCID: PMC11834447 DOI: 10.1080/21645515.2025.2465161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/15/2025] [Accepted: 02/07/2025] [Indexed: 02/13/2025] Open
Abstract
In light of the widespread use of rotavirus vaccines, there is a pressing need to perform thorough, large-scale surveillance to actively monitor safety. This study aimed to identify potential adverse events following rotavirus vaccination in infants. Using a nationwide linked database of the national immunization registry and health insurance claims data, we identified infants vaccinated with the first dose of rotavirus vaccine between January 2016 and October 2022. The self-controlled tree-temporal scan statistics method analyzed all incident diagnoses recorded within 56 days post-vaccination and evaluated all temporal risk windows. Among 1,720,778 rotavirus vaccine recipients 64,752 infants contributed to the analysis, yielding 72,970 incident diagnoses. Of these, 28 clusters were categorized as known adverse drug reactions (ADRs), including infection following immunization (Days 1-2, p<.001), viral infection (Days 1-5, p<.001), urticaria and erythema (Days 3-9, p<.001), acute upper respiratory infections (Days 28-42, p<.001), and pneumonia (Days 9-19 or 28-42, p<.001). Seventeen clusters were classified as ADR-related events, such as the ones clinically related to ADR or lower-level diagnostic codes of ADR. The remaining 26 clusters were classified as signals, including sepsis (Days 1-20, p<.001), meningitis (Days 1-23, p<.001), liver disease (Days 4-11, p<.001), and tubulo-interstitial nephritis (Days 11-38, p<.001). A cluster of intussusceptions was only found in monovalent vaccine-stratified analysis (Days 5-8, p = 0.005). This study confirmed known ADRs following rotavirus vaccination, while identifying potential safety signals requiring further investigation. These findings emphasize the importance of active vaccine surveillance and underscore the need for epidemiological studies with validated outcome definitions to confirm causal relationships between rotavirus vaccination and the detected outcomes.
Collapse
Affiliation(s)
- Na-Young Jeong
- Health Science Convergence Research Institute, Ewha Womans University, Seoul, Republic of Korea
| | - Haerin Cho
- Department of Health Convergence, College of Science and Industry Convergence, Ewha Womans University, Seoul, Republic of Korea
| | - Hee-Jin Kim
- Department of Health Convergence, College of Science and Industry Convergence, Ewha Womans University, Seoul, Republic of Korea
| | - Nam-Kyong Choi
- Department of Health Convergence, College of Science and Industry Convergence, Ewha Womans University, Seoul, Republic of Korea
- Graduate School of Industrial Pharmaceutical Science, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
2
|
Rotundo S, Tassone MT, Lionello R, Fusco P, Serapide F, Russo A. Emerging Prognostic and Predictive Biomarkers for Human Cytomegalovirus Infection During Pregnancy: Unmet Needs and Future Perspectives. Viruses 2025; 17:705. [PMID: 40431716 PMCID: PMC12115395 DOI: 10.3390/v17050705] [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: 04/01/2025] [Revised: 05/11/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Human cytomegalovirus (HCMV) infection during pregnancy is a leading cause of congenital infections worldwide, posing significant risks to fetal health. Despite advances in prenatal care, managing HCMV infection remains challenging. Early detection, accurate risk assessment, and timely intervention are critical to mitigating the adverse outcomes associated with congenital HCMV (cHCMV), such as neurodevelopmental delays and hearing loss. However, the current landscape of biomarkers for HCMV infection in pregnancy is marked by several unmet needs. These gaps in biomarker development and application limit our ability to predict fetal transmission, assess the risk of fetal damage, and prognosticate long-term outcomes. Addressing these challenges through the identification and validation of novel biomarkers could revolutionize the management of HCMV in pregnancy, leading to improved outcomes for both mothers and their children. This review examines the critical unmet needs regarding HCMV biomarkers during pregnancy, emphasizing the priority areas for further research and innovation.
Collapse
Affiliation(s)
- Salvatore Rotundo
- Infectious Disease Unit, “San Giovanni di Dio” Hospital, 88900 Crotone, Italy
- Department of Medical and Surgical Sciences, “Magna Graecia” University, 88100 Catanzaro, Italy; (M.T.T.); (F.S.); (A.R.)
| | - Maria Teresa Tassone
- Department of Medical and Surgical Sciences, “Magna Graecia” University, 88100 Catanzaro, Italy; (M.T.T.); (F.S.); (A.R.)
| | - Rosaria Lionello
- Infectious and Tropical Disease Unit, “Renato Dulbecco” Teaching Hospital, 88100 Catanzaro, Italy; (R.L.); (P.F.)
| | - Paolo Fusco
- Infectious and Tropical Disease Unit, “Renato Dulbecco” Teaching Hospital, 88100 Catanzaro, Italy; (R.L.); (P.F.)
| | - Francesca Serapide
- Department of Medical and Surgical Sciences, “Magna Graecia” University, 88100 Catanzaro, Italy; (M.T.T.); (F.S.); (A.R.)
- Infectious and Tropical Disease Unit, “Renato Dulbecco” Teaching Hospital, 88100 Catanzaro, Italy; (R.L.); (P.F.)
| | - Alessandro Russo
- Department of Medical and Surgical Sciences, “Magna Graecia” University, 88100 Catanzaro, Italy; (M.T.T.); (F.S.); (A.R.)
- Infectious and Tropical Disease Unit, “Renato Dulbecco” Teaching Hospital, 88100 Catanzaro, Italy; (R.L.); (P.F.)
| |
Collapse
|
3
|
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
|
4
|
El Hachem G, Poder TG, Carey CM, Gantt S, Kakkar F, Sab M, Renaud C, Boucoiran I. Prenatal Screening for CMV Primary Infection: A Cost-Utility Model. BJOG 2025; 132:805-815. [PMID: 39895506 PMCID: PMC11969920 DOI: 10.1111/1471-0528.18080] [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: 04/16/2024] [Revised: 11/12/2024] [Accepted: 01/10/2025] [Indexed: 02/04/2025]
Abstract
OBJECTIVE Congenital cytomegalovirus (CMV) infection is a major cause of deafness and neurodevelopmental disability in children. Our objective was to assess the cost utility of first-trimester serological CMV screening, compared to screening of high-risk pregnancies and no serological screening. DESIGN A decision-analytic model was created to compare the cost utility of three strategies from a healthcare sector perspective: universal first-trimester serological screening, screening only of high-risk pregnant women (both including antiviral prophylaxis in cases of primary infection) and serological testing triggered by foetal morphological ultrasound (no CMV serological screening). SETTING Canada. POPULATION Hypothetical population of 80 000 pregnant women. METHODS Probability, expected values and cost estimates were derived from published literature and local hospital and national insurance data. MAIN OUTCOME MEASURE Cost per maternal and infant quality-adjusted life year (QALY) lost. RESULTS Universal serological screening was superior to both screening of high-risk women and no screening (utility of -0.42, -0.63 and - 0.87 QALY lost, respectively). Sensitivity analysis demonstrated that universal screening was the most cost-effective strategy regardless of the incidence of primary infection, the acceptability of amniocentesis and the efficacy of antiviral prophylaxis. In the Monte Carlo analyses, universal serological screening was the most cost-effective option in 96.36% of simulations. Universal serological screening would allow detection of 152 cases of primary maternal CMV infection and would prevent 29 cases of congenital CMV infection annually. CONCLUSION Our findings support the adoption of a population-based prenatal screening programme for the prevention of congenital CMV infection.
Collapse
Affiliation(s)
- Gebrael El Hachem
- School of Public HealthUniversité de MontréalMontrealQuebecCanada
- Women and Children's Infectious Diseases Center, CHU Sainte‐Justine Research CenterMontrealCanada
| | - Thomas G. Poder
- School of Public HealthUniversité de MontréalMontrealQuebecCanada
- Research Center of the IUSMMCIUSSS de l'Est de l'Île de MontréalMontrealCanada
| | - Catherine Mc Carey
- Women and Children's Infectious Diseases Center, CHU Sainte‐Justine Research CenterMontrealCanada
- Department of Obstetrics and GynecologyUniversité de MontréalMontrealCanada
| | - Soren Gantt
- Women and Children's Infectious Diseases Center, CHU Sainte‐Justine Research CenterMontrealCanada
- Department of PediatricsUniversité de MontréalMontrealCanada
| | - Fatima Kakkar
- Women and Children's Infectious Diseases Center, CHU Sainte‐Justine Research CenterMontrealCanada
- Department of PediatricsUniversité de MontréalMontrealCanada
| | - Marc Sab
- Faculty of MedicineUniversité Catholique de LouvainBrusselsBelgium
| | - Christian Renaud
- Women and Children's Infectious Diseases Center, CHU Sainte‐Justine Research CenterMontrealCanada
- Department of PediatricsUniversité de MontréalMontrealCanada
| | - Isabelle Boucoiran
- School of Public HealthUniversité de MontréalMontrealQuebecCanada
- Women and Children's Infectious Diseases Center, CHU Sainte‐Justine Research CenterMontrealCanada
| |
Collapse
|
5
|
Wiemers P, Graf I, Addo MM, Arck PC, Diemert A. Mothers and mosquitoes: climate change contributes to the spread of vector-borne pathogens posing a substantial threat to pregnant women. Semin Immunopathol 2025; 47:25. [PMID: 40272573 PMCID: PMC12021716 DOI: 10.1007/s00281-025-01050-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 03/23/2025] [Indexed: 04/25/2025]
Abstract
Infectious diseases have threatened individuals and societies since the dawn of humanity. Certain population groups, including pregnant women, young children and the elderly, are particularly vulnerable to severe infections. Over the past few centuries, advances in medical standards and the availability of vaccines have reduced infection-related mortality and morbidity rates in industrialized countries. However, the global rise in temperatures and increased precipitation present a new challenge, facilitating the broader distribution of disease vectors, such as mosquitoes, bugs and ticks, to higher altitudes and latitudes. Consequently, epidemic and pandemic outbreaks associated with these vectors, such as Zika, West Nile, dengue, yellow fever, chikungunya and malaria, are increasingly impacting diverse populations. This review comprehensively examines how infections associated with climate change disproportionately affect the health and well-being of pregnant women and their unborn children. There has been a noticeable emergence of vector-borne diseases in Europe. Consequently, we stress the importance of implementing measures that effectively protect pregnant women from these increasing infections globally and regionally. We advocate for initiatives to safeguard pregnant women from these emerging threats, beginning with enhanced education to raise awareness about the evolving risks this particularly vulnerable population faces.
Collapse
Affiliation(s)
- Pauline Wiemers
- Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Isabel Graf
- Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marylyn M Addo
- Institute for Infection Research and Vaccine Development, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Petra C Arck
- Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- German Center for Child and Adolescent Health, Partner Site Hamburg, Hamburg, Germany.
| | - Anke Diemert
- Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- German Center for Child and Adolescent Health, Partner Site Hamburg, Hamburg, Germany.
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20251, Hamburg, Germany.
| |
Collapse
|
6
|
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
|
7
|
Mundy MA, Demers D, Brossay L. Lung NK cells are sufficient to control viral dissemination during respiratory MCMV infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf039. [PMID: 40174911 DOI: 10.1093/jimmun/vkaf039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 01/29/2025] [Indexed: 04/04/2025]
Abstract
Murine cytomegalovirus (MCMV) respiratory dissemination schemes, which mimic natural infection routes, have only recently become an area of investigation. Using an intratracheal (i.t.) infection method, we discovered that the respiratory infection route yields differential infection kinetics compared to the widely used intraperitoneal (i.p.) infection method. Remarkably, we find that respiratory infection results in limited dissemination, with the virus being mostly contained in the pulmonary tissue. Importantly, using Rag1, Ly49H, and natural killer (NK) cell-deficient animals, we find that lung conventional NK (cNK) cells play a critical role in preventing MCMV-induced morbidity. Mechanistically, we show that indirect activation of lung NK cells via interleukin (IL)-12 and type 1 interferon (IFN) inflammatory cytokines is dispensable, while direct activation via Ly49H is essential in preventing morbidity from i.t. infection. Additionally, we did not find a significant role for ILC2 or tissue-resident NK (trNK) cells in the prevention of viral dissemination, and we did not observe an increase in the abundance of these cells. These findings uncover an unanticipated role for pulmonary cNK cells in preventing viral dissemination from infected lungs.
Collapse
Affiliation(s)
- Miles A Mundy
- Division of Biology and Medicine, Department of Molecular Microbiology & Immunology, Brown University, Providence, RI, United States
| | - Delia Demers
- Division of Biology and Medicine, Department of Molecular Microbiology & Immunology, Brown University, Providence, RI, United States
| | - Laurent Brossay
- Division of Biology and Medicine, Department of Molecular Microbiology & Immunology, Brown University, Providence, RI, United States
| |
Collapse
|
8
|
Dzubay SK, Gagliuso AH, Arora M, Doshi U, Caughey AB. Universal screening and valacyclovir for first trimester primary cytomegalovirus: a cost-effectiveness analysis. Am J Obstet Gynecol 2025:S0002-9378(25)00089-4. [PMID: 39947573 DOI: 10.1016/j.ajog.2025.02.009] [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: 10/04/2024] [Revised: 02/04/2025] [Accepted: 02/06/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND Universal prenatal screening for cytomegalovirus is not currently recommended in the United States, as no effective interventions have previously been available. With growing evidence that treating maternal first trimester primary cytomegalovirus infections with valacyclovir reduces vertical transmission, universal screening may become an important tool in congenital cytomegalovirus prevention. OBJECTIVE This study examined the cost-effectiveness of a universal screening approach for maternal cytomegalovirus in the first trimester of pregnancy followed by valacyclovir treatment in positive cases for prevention of the sequelae of congenital cytomegalovirus. STUDY DESIGN A decision-analytic model was constructed to compare outcomes of universal screening and subsequent valacyclovir treatment in a theoretical cohort of 2,869,141 individuals, the estimated number of pregnant people in the United States who receive prenatal care by the first trimester. Individuals found to be immunoglobulin G positive, immunoglobulin M positive, and to have low immunoglobulin G avidity were considered to have primary cytomegalovirus infection and received valacyclovir. Outcomes included cases of vertical cytomegalovirus transmission, abortions, stillbirths, neonatal deaths, cases of hearing loss, cases of neurodevelopmental disabilities, costs, and quality-adjusted life years. Model inputs were derived from literature. Sensitivity analyses were performed via tornado analysis, univariable sensitivity analysis, and multivariable sensitivity analysis with Monte Carlo simulation. RESULTS In our study, universal screening in the first trimester for primary cytomegalovirus and subsequent treatment with valacyclovir in positive cases decreased adverse outcomes by preventing 2898 vertical transmissions, 94 abortions, 19 stillbirths, 11 neonatal deaths, 460 cases of hearing loss, and 263 cases of neurodevelopmental disability. Universal screening and subsequent treatment was the dominant strategy as it saved 242.2 million dollars and led to 3437 additional quality-adjusted life years. Tornado analysis demonstrated that there were no threshold values which would change the model results, when all variables were varied across a predetermined range. Univariable sensitivity analyses demonstrated that even with decreasing specificity of screening tests, decreasing maternal utility of neonatal hearing loss, and increasing the price of screening up to 17 times the current cost, universal screening remained the cost-effective strategy. Monte Carlo analysis demonstrated that the screening strategy remained cost-saving in 100% of trials. CONCLUSION Our results demonstrate that screening for first trimester primary cytomegalovirus may be a cost-saving intervention, as identification of cases allows for the provision of treatment, which in turn reduces vertical cytomegalovirus transmission and costly sequelae.
Collapse
Affiliation(s)
- Sarah K Dzubay
- Department of Obstetrics & Gynecology, Oregon Health & Science University, Portland, OR
| | - Amelia H Gagliuso
- Department of Obstetrics & Gynecology, Oregon Health & Science University, Portland, OR
| | - Megha Arora
- Department of Obstetrics & Gynecology, Oregon Health & Science University, Portland, OR
| | - Uma Doshi
- Department of Obstetrics & Gynecology, University of California Los Angeles, Los Angeles, CA
| | - Aaron B Caughey
- Department of Obstetrics & Gynecology, Oregon Health & Science University, Portland, OR.
| |
Collapse
|
9
|
Li Y, Yang HS, Klasse PJ, Zhao Z. The significance of antigen-antibody-binding avidity in clinical diagnosis. Crit Rev Clin Lab Sci 2025; 62:9-23. [PMID: 39041650 DOI: 10.1080/10408363.2024.2379286] [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: 03/12/2024] [Revised: 06/07/2024] [Accepted: 07/09/2024] [Indexed: 07/24/2024]
Abstract
Immunoglobulin G (IgG) and immunoglobulin M (IgM) testing are commonly used to determine infection status. Typically, the detection of IgM indicates an acute or recent infection, while the presence of IgG alone suggests a chronic or past infection. However, relying solely on IgG and IgM antibody positivity may not be sufficient to differentiate acute from chronic infections. This limitation arises from several factors. The prolonged presence of IgM can complicate diagnostic interpretations, and false positive IgM results often arise from antibody cross-reactivity with various antigens. Additionally, IgM may remain undetectable in prematurely collected samples or in individuals who are immunocompromised, further complicating accurate diagnosis. As a result, additional diagnostic tools are required to confirm infection status. Avidity is a measure of the strength of the binding between an antigen and antibody. Avidity-based assays have been developed for various infectious agents, including toxoplasma, cytomegalovirus (CMV), SARS-CoV-2, and avian influenza, and are promising tools in clinical diagnostics. By measuring the strength of antibody binding, they offer critical insights into the maturity of the immune response. These assays are instrumental in distinguishing between acute and chronic or past infections, monitoring disease progression, and guiding treatment decisions. The development of automated platforms has optimized the testing process by enhancing efficiency and minimizing the risk of manual errors. Additionally, the recent advent of real-time biosensor immunoassays, including the label-free immunoassays (LFIA), has further amplified the capabilities of these assays. These advances have expanded the clinical applications of avidity-based assays, making them useful tools for the diagnosis and management of various infectious diseases. This review is structured around several key aspects of IgG avidity in clinical diagnosis, including: (i) a detailed exposition of the IgG affinity maturation process; (ii) a thorough discussion of the IgG avidity assays, including the recently emerged biosensor-based approaches; and (iii) an examination of the applications of IgG avidity in clinical diagnosis. This review is intended to contribute toward the development of enhanced diagnostic tools through critical assessment of the present landscape of avidity-based testing, which allows us to identify the existing knowledge gaps and highlight areas for future investigation.
Collapse
Affiliation(s)
- Yaxin Li
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - He S Yang
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - P J Klasse
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA
| | - Zhen Zhao
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
10
|
Rasmussen SA, Perrotta K, Conover E, Curran CP, Običan SG. Updated Joint Position Statement on Vaccines From the Society for Birth Defects Research and Prevention and the Organization of Teratology Information Specialists. Birth Defects Res 2025; 117:e2433. [PMID: 39823158 DOI: 10.1002/bdr2.2433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 12/31/2024] [Accepted: 01/02/2025] [Indexed: 01/19/2025]
Affiliation(s)
- Sonja A Rasmussen
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kirstie Perrotta
- MotherToBaby California, San Diego, California, USA
- Department of Pediatrics, University of California, San Diego, California, USA
| | - Elizabeth Conover
- Department of Genetic Medicine, Munroe Meyer Institute, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Christine Perdan Curran
- Department of Biological Sciences, Northern Kentucky University, Highland Heights, Kentucky, USA
| | - Sarah G Običan
- Department of Obstetrics and Gynecology, University of South Florida Morsani College of Medicine, Tampa, Florida, USA
| |
Collapse
|
11
|
Palav HC, Padwal VS, Velhal SM, Yadav S, Bhonde GS, Kalsurkar V, Agrawal S, Set R, Shastri J, Shah F, Shah I, Satoskar P, Patel V, Bhor VM. Recent HCMV infection in early pregnancy associates with congenital transmission & adverse pregnancy outcome: A prospective cohort study. Indian J Med Res 2024; 160:614-624. [PMID: 39913512 PMCID: PMC11801775 DOI: 10.25259/ijmr_89_2024] [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: 03/14/2024] [Accepted: 09/02/2024] [Indexed: 02/11/2025] Open
Abstract
Background & objectives Human Cytomegalovirus (HCMV) infection, leading to >90 per cent seropositivity in women of reproductive age from India, is the largest cause of congenital infections worldwide. HCMV infection status was prospectively monitored together with congenital transmission (cCMV) and adverse pregnancy outcomes (APO) in a public health setting where maternal or neonatal screening was not in practice. Methods Eighty three pregnant women, with (n=45) and without (n=38) bad obstetric history (BOH), were monitored for HCMV infection by ELISA-(IgM, IgG, IgG avidity) for all TORCH (Toxoplasma, Rubella, HCMV, HSV 1 & 2) pathogens along with HCMV-specific chemiluminescent microparticle immunoassay (CMIA) and nested polymerase chain reaction (PCR). Descriptive statistics were applied on data sets to determine associations between maternal infection status, pregnancy outcome and cCMV in 52 mother-neonate dyads. Results Combined avidity, PCR-based and HCMV IgM screening, compared to the latter alone, was successful in identifying incident infection during early pregnancy. Pregnancy loss was associated strongly with BOH and concurrent HCMV infection. Features associated with APO and cCMV, were high PCR positivity (first trimester) and high rates of HCMV-specific IgM and intermediate IgG avidity (P=0.0211, 0.0455). Also, recent HCMV infection (intermediate IgG avidity), observed mainly in the BOH group, but not recurrent infection (IgM positivity), in first and second trimesters, was associated with neonatal saliva positivity and adverse outcomes, including neonatal death (P=0.0762). Exposure to other TORCH pathogens, while detected, did not include IgM positivity or low/intermediate IgG. Conclusion This study highlights the significance of conducting early, multi-pronged screening for maternal HCMV infection during pregnancy, especially in public health settings with high HCMV seroprevalence.
Collapse
Affiliation(s)
- Harsha Chandrashekhar Palav
- Department of Viral Immunopathogenesis Lab, ICMR – National Institute for Research in Reproductive and Child Health, Mumbai, Maharashtra, India
| | - Varsha Sakharam Padwal
- Department of Viral Immunopathogenesis Lab, ICMR – National Institute for Research in Reproductive and Child Health, Mumbai, Maharashtra, India
| | - Shilpa Milind Velhal
- Department of Viral Immunopathogenesis Lab, ICMR – National Institute for Research in Reproductive and Child Health, Mumbai, Maharashtra, India
| | - Sapna Yadav
- Department of Viral Immunopathogenesis Lab, ICMR – National Institute for Research in Reproductive and Child Health, Mumbai, Maharashtra, India
| | - Gauri Sanjay Bhonde
- Department of Molecular Immunology and Microbiology, ICMR – National Institute for Research in Reproductive and Child Health, Mumbai, Maharashtra, India
| | - Varsha Kalsurkar
- Department of Viral Immunopathogenesis Lab, ICMR – National Institute for Research in Reproductive and Child Health, Mumbai, Maharashtra, India
| | - Sachee Agrawal
- Department of Microbiology, T.N. Medical College & B.Y.L. Nair Hospital, Mumbai, Maharashtra, India
| | - Reena Set
- Department of Microbiology, T.N. Medical College & B.Y.L. Nair Hospital, Mumbai, Maharashtra, India
| | - Jayanthi Shastri
- Department of Microbiology, T.N. Medical College & B.Y.L. Nair Hospital, Mumbai, Maharashtra, India
| | - Forum Shah
- Nowrosjee Wadia Maternity Hospital, Mumbai, Maharashtra, India
| | - Ira Shah
- Bai Jerbai Wadia Hospital for Children, Mumbai, Maharashtra, India
| | | | - Vainav Patel
- Department of Viral Immunopathogenesis Lab, ICMR – National Institute for Research in Reproductive and Child Health, Mumbai, Maharashtra, India
| | - Vikrant Madhukar Bhor
- Department of Molecular Immunology and Microbiology, ICMR – National Institute for Research in Reproductive and Child Health, Mumbai, Maharashtra, India
| |
Collapse
|
12
|
Vande Walle C, Maris F, Schiettecatte E, Herregods N. The value of magnetic resonance imaging in congenital cytomegalovirus infection: a systematic review. Pediatr Radiol 2024; 54:2157-2174. [PMID: 39419855 DOI: 10.1007/s00247-024-06051-y] [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: 08/06/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 10/19/2024]
Abstract
Congenital cytomegalovirus (cCMV) infection can lead to severe neurodevelopmental and hearing impairments. Imaging techniques can be used both pre- and postnatally to assess early signs of infection. The objective was to provide a systematic review of current literature regarding magnetic resonance imaging (MRI) and its value to predict clinical outcome in children with cCMV. PubMed, Embase, and Web of Science were searched for studies investigating MRI in cCMV between 2016-2024. Risk of bias was assessed using Newcastle-Ottawa quality assessment scales. Descriptive synthesis was performed. Twenty studies were included. MRI detected brain abnormalities in 5.0-53.0% of infected patients prenatally and 26.9-69.0% postnatally. The three most frequently detected abnormalities included white matter lesions, subependymal cysts, and ventricular dilatation. Symptoms at birth, first trimester seroconversion, and high viral load were associated with abnormal MRI; however, brain abnormalities were still found in 33-37% of clinically asymptomatic patients. Prenatal MRI had a negative predictive value of 94-100% and a positive predictive value of 12-60% for predicting adverse clinical outcome. Five in six studies found an association between MRI abnormalities and neurodevelopmental impairments, five in eight with (congenital) hearing loss. MRI detected additional abnormalities in 5.6-19.4% of children with normal ultrasound. In conclusion, MRI can detect a wide range of brain abnormalities, both pre- and postnatally, in symptomatic and asymptomatic patients. MRI can be a helpful tool in the prediction of clinical impairments and seems complementary to ultrasound. Therefore, both fetal and neonatal MRI should be considered in the standard work-up of all cCMV-infected children.
Collapse
Affiliation(s)
| | | | - Eva Schiettecatte
- Ghent University Hospital, Corneel Heymanslaan 10, Ghent, 9000, Belgium
| | - Nele Herregods
- Ghent University Hospital, Corneel Heymanslaan 10, Ghent, 9000, Belgium
| |
Collapse
|
13
|
Gyawali J, Pudasainee-Kapri S, Agrawal S, Khatri DB, Adhikari S, Dhunagana PP. Cytomegalovirus-associated pancytopenia in a four-month-old infant: a case report. BMC Infect Dis 2024; 24:1277. [PMID: 39528986 PMCID: PMC11555845 DOI: 10.1186/s12879-024-10191-9] [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: 09/02/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024] Open
Abstract
Cytomegalovirus (CMV) is a beta-herpes virus causing common infections, often asymptomatic in healthy individuals. However, it poses increased risks to immunocompromised individuals and can cause congenital infections, leading to severe disabilities. CMV infection can cause significant hematological abnormality. A four-month-old female infant was admitted for decreased feeding for two days. She was severely pale, without hepatosplenomegaly. In initial laboratory investigations hemoglobin, platelet count, and white blood cells were decreased. The patient was transfused with whole blood and referred to a tertiary care center. Further workup, including bone marrow biopsy, showed hypocellular marrow. The Urine CMV Polymerase Chain Reaction (PCR) test returned highly positive with a viral load of 1,700,000 copies/mL. This patient was diagnosed with CMV-associated bone marrow suppression, and she was prescribed valganciclovir at a dosage of 16 mg/kg/dose every 12 h for 6 months. She had shown significant hematologic parameter improvement during subsequent follow-up. Pancytopenia in infancy should include a differential diagnosis for CMV infection. The early recognition and correct infection management, including antiviral therapy and symptomatic treatment, yield a better prognosis.
Collapse
Affiliation(s)
- Jeevan Gyawali
- Internal Medicine Department, Chirayu National Hospital and Medical Institute (CNHMI), Maharajgunj, Kathmandu, 44600, Nepal.
| | | | - Sumit Agrawal
- Pediatric Department, Kanti Children's Hospital, Maharajgunj, Kathmandu, 44600, Nepal
| | - Dhan Bahadur Khatri
- Internal Medicine Department, Chirayu National Hospital and Medical Institute (CNHMI), Maharajgunj, Kathmandu, 44600, Nepal
- Pediatric Department, Chirayu National Hospital and Medical Institute (CNHMI), Maharajgunj, Kathmandu, 44600, Nepal
| | - Sugat Adhikari
- Primary Care Physician, Shreegaun Primary Healthcare Center, Maharajgunj, Dang, 22400, Nepal
| | - Prem Prasad Dhunagana
- Pediatric Department, Patan Academy of Health Sciences - School of Medicine, Lagankhel, Lalitpur, 44700, Nepal
| |
Collapse
|
14
|
Skarzynski PH, Obrycka A, Kolodziejak A, Lorens A, Gos E, Zdanowicz R, Skarzynski H. Auditory Development of Young Children with Profound Hearing Loss, Cochlear Implants, and Congenital CMV Infection. J Clin Med 2024; 13:6734. [PMID: 39597878 PMCID: PMC11595024 DOI: 10.3390/jcm13226734] [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: 10/08/2024] [Revised: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: The aim of this study was to assess auditory development in young children with profound hearing loss, cochlear implants (CIs), and congenital cytomegalovirus (cCMV) infection and to determine the effect of comorbidities on their development. Methods: The study group (cCMV group) consisted of 47 CI children-18 girls and 29 boys-who had been diagnosed as having prelingual hearing loss due to cCMV infection (with or without comorbidities); the mean age at CI activation was 15.2 months (range: 9.7-23.8; SD = 3.5). The reference group (no cCMV) consisted of 117 similar children (57 girls and 60 boys) who had profound sensorineural hearing loss not caused by cCMV infection; they had no comorbidities. The mean age at CI activation in the second group was 14.3 months (range: 7.9-23.5; SD = 4.0). Auditory development in all children was assessed with the LittlEARS Auditory Questionnaire (LEAQ) at CI activation and at about 1, 5, 9, 14, and 24 months of CI use. Results: The mean LEAQ total score increased over a similar time frame from 9.8 pts to 28.9 pts in the cCMV group without comorbidities, from 4.5 pts to 18.5 pts in the cCMV group with comorbidities, and from 9.2 to 31.6 pts in the reference group with no cCMV infection. Conclusions: Early cochlear implantation in children with sensorineural hearing loss due to congenital CMV infection and no comorbidities promotes their early auditory development in a similar way to children without cCMV infection.
Collapse
Affiliation(s)
- Piotr H. Skarzynski
- Teleaudiology and Screening Department, World Hearing Center, Institute of Physiology and Pathology of Hearing, 02-042 Warsaw, Poland; (E.G.); (R.Z.)
- Institute of Sensory Organs, 05-830 Kajetany, Poland
| | - Anita Obrycka
- Implant and Auditory Perception Department, World Hearing Center, Institute of Physiology and Pathology of Hearing, 02-042 Warsaw, Poland; (A.O.); (A.L.)
| | - Aleksandra Kolodziejak
- Teleaudiology and Screening Department, World Hearing Center, Institute of Physiology and Pathology of Hearing, 02-042 Warsaw, Poland; (E.G.); (R.Z.)
| | - Artur Lorens
- Implant and Auditory Perception Department, World Hearing Center, Institute of Physiology and Pathology of Hearing, 02-042 Warsaw, Poland; (A.O.); (A.L.)
| | - Elzbieta Gos
- Teleaudiology and Screening Department, World Hearing Center, Institute of Physiology and Pathology of Hearing, 02-042 Warsaw, Poland; (E.G.); (R.Z.)
| | - Rita Zdanowicz
- Teleaudiology and Screening Department, World Hearing Center, Institute of Physiology and Pathology of Hearing, 02-042 Warsaw, Poland; (E.G.); (R.Z.)
| | - Henryk Skarzynski
- Otorhinolaryngosurgery Clinic, World Hearing Center, Institute of Physiology and Pathology of Hearing, 02-042 Warsaw, Poland;
| |
Collapse
|
15
|
Chan ES, Suchet I, Somerset D, de Koning L, Chadha R, Soliman N, Kuret V, Yu W, Lauzon J, Thomas MA, Poon E, Zhou HY. Maternal Cytomegalovirus (CMV) Serology: The Diagnostic Limitations of CMV IgM and IgG Avidity in Detecting Congenital CMV Infection. Pediatr Dev Pathol 2024; 27:530-544. [PMID: 39270128 PMCID: PMC11568646 DOI: 10.1177/10935266241253477] [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] [Indexed: 09/15/2024]
Abstract
INTRODUCTION Congenital cytomegalovirus (cCMV) is a common congenital viral infection. Testing for cCMV usually begins with assessing maternal CMV serology, specifically IgM and IgG antibodies. A negative maternal CMV IgM suggests a low risk of recent maternal CMV infection, thereby suggesting a low risk of cCMV in the fetus. Consequently, cCMV is often ruled out when maternal CMV IgM is negative. METHODS In our perinatal autopsy and placental pathology database, we identified 5 cases of cCMV despite negative maternal CMV IgM results in the second trimester. RESULTS In all 5 cases, fetal abnormalities were first detected by ultrasound in the second trimester, prompting maternal CMV testing. Since second trimester maternal CMV IgM was negative in all cases, cCMV was considered unlikely, thus precluding further prenatal CMV testing in 4 of these cases. The diagnosis of cCMV was subsequently made through placental and/or autopsy examinations. Following this diagnosis, retrospective CMV serology and IgG avidity testing was performed on stored frozen first-trimester maternal blood samples in 3 cases. Among these, the first-trimester samples in 2 cases were IgG+, IgM+, and exhibited low IgG avidity, suggesting a primary maternal CMV infection around the time of conception. In the third case, both first and second-trimester maternal blood samples were IgG+, IgM-, and showed high IgG avidity, suggesting a non-primary maternal CMV infection (i.e., reactivation or reinfection of CMV). CONCLUSION A negative maternal CMV IgM in the second trimester cannot exclude cCMV infection. While CMV IgG avidity testing and analysis of stored frozen first-trimester maternal blood samples provide valuable insights, they have limitations. CMV PCR performed on amniotic fluid is a useful prenatal diagnostic tool. For cases of unexplained fetal abnormalities or death, autopsy and placental examination are recommended.
Collapse
Affiliation(s)
- Elaine S. Chan
- Department of Pathology and Laboratory Medicine, University of Calgary Cumming School of Medicine, Calgary, AB, Canada
- Alberta Children’s Hospital, Calgary, AB, Canada
- Alberta Precision Laboratories, Calgary, AB, Canada
| | | | - David Somerset
- EFW Radiology, Calgary, AB, Canada
- Department of Obstetrics and Gynaecology, University of Calgary Cumming School of Medicine, Calgary, AB, Canada
| | - Lawrence de Koning
- Department of Pathology and Laboratory Medicine, University of Calgary Cumming School of Medicine, Calgary, AB, Canada
- Alberta Children’s Hospital, Calgary, AB, Canada
- Alberta Precision Laboratories, Calgary, AB, Canada
| | - Rati Chadha
- EFW Radiology, Calgary, AB, Canada
- Department of Obstetrics and Gynaecology, University of Calgary Cumming School of Medicine, Calgary, AB, Canada
| | - Nancy Soliman
- EFW Radiology, Calgary, AB, Canada
- Department of Obstetrics and Gynaecology, University of Calgary Cumming School of Medicine, Calgary, AB, Canada
| | - Verena Kuret
- EFW Radiology, Calgary, AB, Canada
- Department of Obstetrics and Gynaecology, University of Calgary Cumming School of Medicine, Calgary, AB, Canada
| | - Weiming Yu
- Department of Pathology and Laboratory Medicine, University of Calgary Cumming School of Medicine, Calgary, AB, Canada
- Alberta Children’s Hospital, Calgary, AB, Canada
- Alberta Precision Laboratories, Calgary, AB, Canada
| | - Julie Lauzon
- Department of Medical Genetics, University of Calgary Cumming School of Medicine, Calgary, AB, Canada
| | - Mary Ann Thomas
- Department of Medical Genetics, University of Calgary Cumming School of Medicine, Calgary, AB, Canada
| | - Elaine Poon
- Department of Pathology and Laboratory Medicine, University of Calgary Cumming School of Medicine, Calgary, AB, Canada
- Alberta Precision Laboratories, Calgary, AB, Canada
| | - Hong Yuan Zhou
- Department of Pathology and Laboratory Medicine, University of Calgary Cumming School of Medicine, Calgary, AB, Canada
- Alberta Precision Laboratories, Calgary, AB, Canada
| |
Collapse
|
16
|
Schleiss MR, Crooks CM, Karthigeyan KP, Kruc RM, Otero CE, Wang HY(S, Permar SR, Plotkin SA, Gautam R. Proceedings of the Conference "CMV Vaccine Development-How Close Are We?" (27-28 September 2023). Vaccines (Basel) 2024; 12:1231. [PMID: 39591134 PMCID: PMC11598149 DOI: 10.3390/vaccines12111231] [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: 07/23/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 11/28/2024] Open
Abstract
Congenital cytomegalovirus (cCMV) is the most common infectious cause of disability in children, including sensorineural hearing loss. There is interest in developing a pre-conception vaccine that could confer protective immunity on a woman of child-bearing age, hence resulting in a reduced cCMV disease burden. Other populations, including solid organ transplant (SOT) and hematopoietic stem cell transplant (HSCT) patients, could also benefit from CMV vaccination. To review and discuss vaccines that are in clinical development, a workshop, sponsored by the National Institutes of Health (NIH) and the National Institute of Allergy and Infectious Diseases (NIAID), was empaneled. At this workshop, correlates of protective immunity against CMV, epidemiologic features of CMV transmission, and vaccine platforms in development were reviewed. Representatives from academia, pharma, and the NIH engaged in discussion on the current state-of-the-art in CMV vaccinology. A summary of the presentations from this is provided in this report.
Collapse
Affiliation(s)
- Mark R. Schleiss
- Division of Infectious Diseases, Department of Pediatrics, University of Minnesota Medical School, 2001 6th Street SE, Minneapolis, MN 55455, USA
| | - Chelsea M. Crooks
- BB-869-H, Belfer Research Building, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA; (C.M.C.); (K.P.K.); (C.E.O.)
| | - Krithika P. Karthigeyan
- BB-869-H, Belfer Research Building, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA; (C.M.C.); (K.P.K.); (C.E.O.)
| | - Rebecca M. Kruc
- Department of Pediatrics, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA;
| | - Claire E. Otero
- BB-869-H, Belfer Research Building, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA; (C.M.C.); (K.P.K.); (C.E.O.)
| | - Hsuan-Yuan (Sherry) Wang
- BB-869-H, Belfer Research Building, Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA; (C.M.C.); (K.P.K.); (C.E.O.)
| | - Sallie R. Permar
- Department of Pediatrics, Weill Cornell Medicine, 1300 York Ave Box 65, New York, NY 10065, USA;
| | - Stanley A. Plotkin
- Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Boulevard, Building 421, Philadelphia, PA 19104, USA
| | - Rajeev Gautam
- Program Officer at Virology Branch, Division of Microbiology and Infectious Diseases, NIAID, NIH, 5601 Fisher’s Lane, Rockville, MD 20892, USA;
| |
Collapse
|
17
|
Farrokhi A, Atre T, Salitra S, Aletaha M, Márquez AC, Gynn M, Fidanza M, Jo S, Rolf N, Simmons K, Duque-Afonso J, Cleary ML, Seif AE, Kollmann T, Gantt S, Reid GSD. Early-life infection depletes preleukemic cells in a mouse model of hyperdiploid B-cell acute lymphoblastic leukemia. Blood 2024; 144:809-821. [PMID: 38875504 PMCID: PMC11375503 DOI: 10.1182/blood.2024025038] [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: 04/19/2024] [Revised: 05/29/2024] [Accepted: 05/29/2024] [Indexed: 06/16/2024] Open
Abstract
ABSTRACT Epidemiological studies report opposing influences of infection on childhood B-cell acute lymphoblastic leukemia (B-ALL). Although infections in the first year of life appear to exert the largest impact on leukemia risk, the effect of early pathogen exposure on the fetal preleukemia cells (PLC) that lead to B-ALL has yet to be reported. Using cytomegalovirus (CMV) infection as a model early-life infection, we show that virus exposure within 1 week of birth induces profound depletion of transplanted E2A-PBX1 and hyperdiploid B-ALL cells in wild-type recipients and in situ-generated PLC in Eμ-ret mice. The age-dependent depletion of PLC results from an elevated STAT4-mediated cytokine response in neonates, with high levels of interleukin (IL)-12p40-driven interferon (IFN)-γ production inducing PLC death. Similar PLC depletion can be achieved in adult mice by impairing viral clearance. These findings provide mechanistic support for potential inhibitory effects of early-life infection on B-ALL progression and could inform novel therapeutic or preventive strategies.
Collapse
Affiliation(s)
- Ali Farrokhi
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Tanmaya Atre
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Samuel Salitra
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Maryam Aletaha
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Ana Citlali Márquez
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Matthew Gynn
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Mario Fidanza
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Sumin Jo
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Nina Rolf
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Karen Simmons
- Division of Infectious Diseases, Department of Pediatrics, The University of British Columbia, Vancouver, BC, Canada
| | - Jesus Duque-Afonso
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA
| | - Michael L. Cleary
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA
| | - Alix E. Seif
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA
| | - Tobias Kollmann
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Soren Gantt
- Department of Microbiology, Infection, and Immunology, Université de Montreal, Montreal, QC, Canada
| | - Gregor S. D. Reid
- Michael Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Division of Oncology, Hematology and Bone Marrow Transplant, Department of Pediatrics, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
18
|
Liberati C, Sturniolo G, Brigadoi G, Cavinato S, Visentin S, Cosmi E, Donà D, Rampon O. Burden of Congenital CMV Infection: A Narrative Review and Implications for Public Health Interventions. Viruses 2024; 16:1311. [PMID: 39205285 PMCID: PMC11360585 DOI: 10.3390/v16081311] [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: 07/19/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024] Open
Abstract
Cytomegalovirus causes the most common congenital infection worldwide. With most infants asymptomatic at birth, the few affected may present with variable clinical scenarios, from isolated hearing loss to severe neurologic impairment. Public health interventions include all actions at the health system, community, and individual levels that aim at reducing the burden of congenital Cytomegalovirus. This review examines the literature on maternal and neonatal screening programs in light of current evidence for treatment and the development of vaccines against Cytomegalovirus. Potential biases and benefits of these interventions are outlined, with the objective of increasing awareness about the problem and providing readers with data and critical tools to participate in this ongoing debate.
Collapse
Affiliation(s)
- Cecilia Liberati
- Department of Women’s and Children’s Health, Pediatric Infectious Disease, Padua University Hospital, 35126 Padua, Italy; (C.L.); (G.S.); (D.D.); (O.R.)
| | - Giulia Sturniolo
- Department of Women’s and Children’s Health, Pediatric Infectious Disease, Padua University Hospital, 35126 Padua, Italy; (C.L.); (G.S.); (D.D.); (O.R.)
| | - Giulia Brigadoi
- Department of Women’s and Children’s Health, Pediatric Infectious Disease, Padua University Hospital, 35126 Padua, Italy; (C.L.); (G.S.); (D.D.); (O.R.)
| | - Silvia Cavinato
- Infectious and Tropical Diseases Unit, Padua University Hospital, 35126 Padua, Italy;
| | - Silvia Visentin
- Department of Women’s and Children’s Health, Gynecological and Obstetric Clinic, Padua University Hospital, 35126 Padua, Italy; (S.V.); (E.C.)
| | - Erich Cosmi
- Department of Women’s and Children’s Health, Gynecological and Obstetric Clinic, Padua University Hospital, 35126 Padua, Italy; (S.V.); (E.C.)
| | - Daniele Donà
- Department of Women’s and Children’s Health, Pediatric Infectious Disease, Padua University Hospital, 35126 Padua, Italy; (C.L.); (G.S.); (D.D.); (O.R.)
| | - Osvalda Rampon
- Department of Women’s and Children’s Health, Pediatric Infectious Disease, Padua University Hospital, 35126 Padua, Italy; (C.L.); (G.S.); (D.D.); (O.R.)
| |
Collapse
|
19
|
Greig J, Bamford A, Chadwick D, Darley A, Gamoudi D, Palit J. British HIV Association guidelines on the management of opportunistic infection in people living with HIV: Considerations in pregnancy 2024. HIV Med 2024; 25 Suppl 3:3-19. [PMID: 38811357 DOI: 10.1111/hiv.13645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2024] [Indexed: 05/31/2024]
Affiliation(s)
- J Greig
- Sheffield Teaching Hospitals NHS Foundation Trust, UK
| | - A Bamford
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - D Chadwick
- Centre for Clinical Infection, James Cook University Hospital, Middlesbrough, UK
| | - A Darley
- Nottingham University Hospitals NHS Trust, UK
| | | | - J Palit
- Sheffield Teaching Hospitals NHS Foundation Trust, UK
| |
Collapse
|
20
|
Ye X, Shih DJH, Ku Z, Hong J, Barrett DF, Rupp RE, Zhang N, Fu TM, Zheng WJ, An Z. Transcriptional signature of durable effector T cells elicited by a replication defective HCMV vaccine. NPJ Vaccines 2024; 9:70. [PMID: 38561339 PMCID: PMC10984989 DOI: 10.1038/s41541-024-00860-w] [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: 01/13/2023] [Accepted: 02/26/2024] [Indexed: 04/04/2024] Open
Abstract
Human cytomegalovirus (HCMV) is a leading infectious cause of birth defects and the most common opportunistic infection that causes life-threatening diseases post-transplantation; however, an effective vaccine remains elusive. V160 is a live-attenuated replication defective HCMV vaccine that showed a 42.4% efficacy against primary HCMV infection among seronegative women in a phase 2b clinical trial. Here, we integrated the multicolor flow cytometry, longitudinal T cell receptor (TCR) sequencing, and single-cell RNA/TCR sequencing approaches to characterize the magnitude, phenotype, and functional quality of human T cell responses to V160. We demonstrated that V160 de novo induces IE-1 and pp65 specific durable polyfunctional effector CD8 T cells that are comparable to those induced by natural HCMV infection. We identified a variety of V160-responsive T cell clones which exhibit distinctive "transient" and "durable" expansion kinetics, and revealed a transcriptional signature that marks durable CD8 T cells post-vaccination. Our study enhances the understanding of human T-cell immune responses to V160 vaccination.
Collapse
Affiliation(s)
- Xiaohua Ye
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
- Center for Infectious Disease Research, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - David J H Shih
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, USA
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR
| | - Zhiqiang Ku
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Junping Hong
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Diane F Barrett
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Richard E Rupp
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Tong-Ming Fu
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - W Jim Zheng
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
21
|
Mohammed K. Prevalence and Risk Factors of Rubella and Cytomegalovirus Infections Among Pregnant Women in Makkah: Implications for Screening and Vaccination Programs. Cureus 2024; 16:e57269. [PMID: 38686268 PMCID: PMC11057681 DOI: 10.7759/cureus.57269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2024] [Indexed: 05/02/2024] Open
Abstract
Introduction Contracting rubella virus or cytomegalovirus (CMV) while pregnant can lead to severe health issues for both the mother and the unborn child. This study aims to determine the prevalence of these infections in pregnant women and identify associated risk factors. Methods A total of 146 pregnant women consented to participate in this research. Data were collected through a detailed questionnaire and blood samples were obtained from each participant. Blood was drawn into vacutainer tubes, and plasma was separated and stored at -20°C for analysis. We utilized specific enzyme-linked immunosorbent assays (ELISA) for immunoglobulin G (IgG) and Immunoglobulin M (IgM) to detect antibodies against rubella and CMV in the plasma samples. Results The study revealed that the prevalence rates of IgG antibodies for rubella and CMV were 68.5% and 97.3%, respectively. No samples tested positive for IgM antibodies for either disease. A significant relationship was found between chronic rubella infection and women aged 26 to 35 years (p < 0.05). A significant association was also observed between chronic CMV infection and women with lower income (p < 0.05). Conclusion This study confirms the widespread presence of chronic rubella and CMV infections among pregnant women in Makkah. The findings highlight the impact of socioeconomic factors on infection rates and underscore the importance of implementing vaccination programs to mitigate the severity of these infections in pregnant women and protect fetal health.
Collapse
Affiliation(s)
- Khalil Mohammed
- Epidemiology and Medical Statistics, Faculty of Public Health and Health Informatics, Umm Al-Qura University, Makkah, SAU
| |
Collapse
|
22
|
Sartori P, Baud D, de Tejada BM, Farin A, Rossier MC, Rieder W, Rouiller S, Robyr R, Grant G, Eggel B, Bruno A, Pretalli M, Gainon B, Capoccia-Brugger R, Ammon-Chansel L, De Courten C, Beurret-Lepori N, Baert J, Vial Y, Pomar L. Cytomegalovirus infection during pregnancy: cross-sectional survey of knowledge and prevention practices of healthcare professionals in French-speaking Switzerland. Virol J 2024; 21:45. [PMID: 38383491 PMCID: PMC10882847 DOI: 10.1186/s12985-024-02318-w] [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: 11/06/2023] [Accepted: 02/14/2024] [Indexed: 02/23/2024] Open
Abstract
BACKGROUND Lack of Cytomegalovirus (CMV) knowledge among healthcare professionals has been proven to be the main threat to pregnant women's awareness, preventing them from reducing the risk of infection. The aims of this study were to assess the knowledge and practices of French-speaking Swiss perinatal professionals in terms of CMV prevention, as well as the sociodemographic-professional factors that influence them. METHODS This observational study used a cross-sectional design to collect data-via an anonymous electronic questionnaire in French distributed to gynecologists-obstetricians, general practitioners and midwives via various channels: e-mails and social networks of partner centers, professional associations, and conferences. The 41-item questionnaire collected data on sociodemographic and professional characteristics, general CMV knowledge, national recommendation knowledge and prevention practices. Univariable and multivariable analyses were performed. RESULTS A total of 110 gynecologist-obstetricians, 5 general practitioners and 226 midwives participated in the study. While more than 80% of practitioners were familiar with protective hygiene measures, significant gaps were highlighted concerning the transmission routes, as well as the signs of short- and long-term congenital CMV infection. Regarding practice, 63.3% of participants provided information on CMV to their patients, mainly during the first antenatal visit. Among those who did not, lack of knowledge and forgetfulness were the two main reasons cited. Concerning systematic screening, 45.7% of participants offered it to their patients, and 37.3% only offered it to "at risk" groups. The existence of national guidelines on CMV was known by 62.0% of participants. Multivariable analysis revealed that working as a gynecologist-obstetrician was independently associated with higher score of preventive practices, while performing ultrasound or preconception consultations was independently associated with a higher score of general CMV knowledge, and working in a university hospital was independently associated with a higher score of Swiss recommendation knowledge. A level of training higher than the basic medical or midwifery diploma and participation in fetal medicine symposia both promote a higher score of CMV knowledge and prevention practices in line with current recommendations. CONCLUSION This study confirms the significant gaps in CMV knowledge among French-speaking Swiss caregivers along with the heterogeneity of their prevention practices. To raise awareness among pregnant women and reduce the burden of congenital CMV infections, improving professional knowledge through access to specific training and standardizing practices should be a national priority.
Collapse
Affiliation(s)
- Pauline Sartori
- School of Health Sciences (HESAV), HES-SO University of Applied Sciences and Arts Western Switzerland, Avenue de Beaumont 21, 1011, Lausanne, Switzerland
- Department Woman-Mother-Child, Lausanne University Hospital and University of Lausanne, 1011, Lausanne, Switzerland
| | - David Baud
- Department Woman-Mother-Child, Lausanne University Hospital and University of Lausanne, 1011, Lausanne, Switzerland
| | - Begoña Martinez de Tejada
- Obstetrics Division, Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University Hospitals of Geneva, 1205, Geneva, Switzerland
| | - Alexandre Farin
- Obstetrics Unit, Hospital Riviera-Chablais, 1847, Rennaz, Switzerland
| | | | - Wawrzyniec Rieder
- Department Woman-Mother-Child, Lausanne University Hospital and University of Lausanne, 1011, Lausanne, Switzerland
- Dianecho, 1205, Geneva, Switzerland
| | - Sylvie Rouiller
- Service of Gynecology and Obstetrics, Ensemble Hospitalier de La Côte, 1110, Morges, Switzerland
| | | | - Gaston Grant
- Department of Gynecology and Obstetrics, Cantonal Hospital, 1752, Villars-Sur-Glâne, Switzerland
- Maternal-Fetal Medicine, Point-f Women's Medical Centre, 1700, Fribourg, Switzerland
| | - Béatrice Eggel
- Department Woman-Mother-Child, Lausanne University Hospital and University of Lausanne, 1011, Lausanne, Switzerland
| | - Adrien Bruno
- School of Health Sciences (HESAV), HES-SO University of Applied Sciences and Arts Western Switzerland, Avenue de Beaumont 21, 1011, Lausanne, Switzerland
| | - Maeva Pretalli
- Maternity Ward, Hôpital du Jura, 2800, Delémont, Switzerland
| | - Bertrand Gainon
- Maternity Ward, Hôpital du Jura, 2800, Delémont, Switzerland
| | - Romina Capoccia-Brugger
- Department of Obstetrics and Gynecology, Réseau Hospitalier Neuchâtelois, 2000, Neuchâtel, Switzerland
| | | | - Clarisse De Courten
- Department of Gynaecology and Obstetrics, Regional Hospital EHNV, 1400, Yverdon, Switzerland
| | | | - Jonathan Baert
- Department Woman-Mother-Child, Lausanne University Hospital and University of Lausanne, 1011, Lausanne, Switzerland
| | - Yvan Vial
- Department Woman-Mother-Child, Lausanne University Hospital and University of Lausanne, 1011, Lausanne, Switzerland
| | - Léo Pomar
- School of Health Sciences (HESAV), HES-SO University of Applied Sciences and Arts Western Switzerland, Avenue de Beaumont 21, 1011, Lausanne, Switzerland.
- Department Woman-Mother-Child, Lausanne University Hospital and University of Lausanne, 1011, Lausanne, Switzerland.
| |
Collapse
|
23
|
Radoi CL, Zlatian O, Balasoiu M, Dragomir TL, Sorop MI, Bagiu IC, Boeriu E, Susan M, Sorop B, Oprisoni LA, Iliescu DG. Seroprevalence of Anti-Cytomegalovirus Antibodies in Pregnant Women from South-West Romania. Microorganisms 2024; 12:268. [PMID: 38399672 PMCID: PMC10893531 DOI: 10.3390/microorganisms12020268] [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: 12/25/2023] [Revised: 01/17/2024] [Accepted: 01/22/2024] [Indexed: 02/25/2024] Open
Abstract
Cytomegalovirus (CMV), in addition to other agents, is part of the TORCH complex (Toxoplasma gondii, Rubella virus, Cytomegalovirus, Herpes simplex viruses, and other agents). CMV infection is the most frequent cause of congenital malformations. This study aimed to establish the variation of prevalence of anti-CMV antibodies in pregnant women from the South-West region of Romania, according to demographic factors, such as age and area of residence, in two separate time periods (2013-2016 and 2019-2022). We collected from the hospital records the age, place of residence, and anti-CMV antibody test results using immune electrochemiluminescence and chemiluminescence. This study found that the seroprevalence of anti-CMV IgM antibodies increased slightly from 2013-2016 to 2019-2022, from 1.92% to 2.26%, and for IgG antibodies from 93.68% to 94.96%. In both groups was observed a descending trend of anti-CMV IgM seroprevalence with an increase in age, showing a decrease in seroprevalence from 3.57% to 1.09% in pregnant women from rural areas in the 31-35 years age group, while in urban areas, we observed a decrease in seroprevalence from 11.11% to 3.06% in the <20 years age group. The IgG seroprevalence showed an increase both in rural areas (from 93.97% to 95.52%) and urban areas (from 93.52% to 94.27%). In both groups, seroprevalence was higher in rural areas compared to urban regions. These results show a high rate of immunization against CMV in pregnant women in South-West Romania, which led to a low risk of acquiring the primary infection during pregnancy. However, the increase in the rate of primary CMV infections in pregnancy suggests the need for prioritizing screening programs and improving the existing protocols to enhance maternal and child healthcare.
Collapse
Affiliation(s)
- Cristiana Luiza Radoi
- Doctoral School, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Ovidiu Zlatian
- Medical Laboratory, County Clinical Emergency Hospital of Craiova, 200349 Craiova, Romania; (O.Z.); (M.B.)
- Microbiology Department, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Maria Balasoiu
- Medical Laboratory, County Clinical Emergency Hospital of Craiova, 200349 Craiova, Romania; (O.Z.); (M.B.)
- Microbiology Department, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Tiberiu-Liviu Dragomir
- Department of Internal Medicine I, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania;
| | - Madalina Ioana Sorop
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Iulia Cristina Bagiu
- Department of Microbiology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania;
- Multidisciplinary Research Center on Antimicrobial Resistance (MULTI-REZ), Microbiology Department, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Estera Boeriu
- Department of Pediatrics, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| | - Monica Susan
- Department of Internal Medicine I, Centre for Preventive Medicine,”Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania;
| | - Bogdan Sorop
- Department of Obstetrics and Gynecology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania;
| | - Licinia Andrada Oprisoni
- Department of Pediatrics, Discipline of Pediatric Oncology and Hematology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania;
| | - Dominic Gabriel Iliescu
- Obstetrics and Gynecology Department, County Clinical Emergency Hospital of Craiova, 200349 Craiova, Romania;
- Obstetrics and Gynecology Department, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| |
Collapse
|
24
|
Mirsalehi N, Yavarian J, Ghavami N, Naseri M, Khodakhah F, Shatizadeh Malekshahi S, Zadheidar S, Mokhtari-Azad T, Shafiei-Jandaghi NZ. Congenital cytomegalovirus infection in newborns suspected of congenital rubella syndrome in Iran: a cross-sectional study. BMC Pediatr 2024; 24:31. [PMID: 38200453 PMCID: PMC10777647 DOI: 10.1186/s12887-023-04502-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Following rubella virus control, the most important cause of congenital infections is human cytomegalovirus (HCMV). Congenital CMV (cCMV) may happen both in primary and non-primary maternal infections. The present study aimed to screen cCMV in symptomatic newborns suspected of congenital rubella syndrome (CRS) in Iran. METHODS Out of 1629 collected infants' serum samples suspected of CRS but negative for rubella IgM, 524 samples were selected regarding cCMV complications. These samples were divided into two age groups: 1- one month and younger, 2- older than 1 month up to one year. Anti-HCMV IgM detection was performed on these serums. Then HCMV IgG avidity assay and HCMV DNA detection were carried out on all samples with positive and borderline results in IgM detection. RESULTS Herein, 3.67% of symptomatic infants aged one month and younger had positive and borderline HCMV IgM, 12.5% of which had a low avidity index (AI). HCMV IgM detection rate among symptomatic infants older than one month to one year was 14.5%. Identified genotypes in this study were gB-1(63.63%), gB2 (18.18%), and gB3 (18.18%), respectively. CONCLUSIONS This comprehensive study was performed on serum samples of symptomatic infants clinically suspected of cCMV from all over Iran. There was a good correlation between serology findings and PCR.
Collapse
Affiliation(s)
- Negar Mirsalehi
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Jila Yavarian
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Nastaran Ghavami
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Naseri
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Farshad Khodakhah
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Sevrin Zadheidar
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Talat Mokhtari-Azad
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Nazanin-Zahra Shafiei-Jandaghi
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
25
|
Calado AM, Seixas F, Pires MDA. Updating an Overview of Teratology. Methods Mol Biol 2024; 2753:1-38. [PMID: 38285332 DOI: 10.1007/978-1-0716-3625-1_1] [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] [Indexed: 01/30/2024]
Abstract
In this chapter, the authors aim to update an overview of the principles of teratology, beginning with the definition of teratology, the critical point at which this process occurs, and some of the most common etiological agents that improve our understanding of teratology.Modern teratology has greatly improved in recent years with advances in new methods in molecular biology, toxicology, animal laboratory science, and genetics, increasing our knowledge of ambient influences. Nevertheless, there is a lot to do to reduce the influence of hazardous intervening agents, whether they target our genetics or not, that can negatively affect pregnancy and induce congenital development disorders, including morphological, biochemical, or behavioral defects.Certain agents might indeed be related to certain defects, but we have not been able to identify the cause of most congenital defects, which highlights the importance of finding and testing out new genetics techniques and conducting laboratory animal science to unravel the etiology and pathogenicity of each congenital defect.
Collapse
Affiliation(s)
- Ana Margarida Calado
- Animal and Veterinary Research Centre (CECAV), UTAD, and Associate Laboratory for Animal and Veterinary Science (AL4Animals), Department of Veterinary Sciences, School of Agrarian and Veterinary Sciences (ECAV), University of Trás-os-Montes e Alto Douro (UTAD), Vila Real, Portugal
| | - Fernanda Seixas
- Animal and Veterinary Research Centre (CECAV), UTAD, and Associate Laboratory for Animal and Veterinary Science (AL4Animals), Department of Veterinary Sciences, School of Agrarian and Veterinary Sciences (ECAV), University of Trás-os-Montes e Alto Douro (UTAD), Vila Real, Portugal
| | - Maria Dos Anjos Pires
- Animal and Veterinary Research Centre (CECAV), UTAD, and Associate Laboratory for Animal and Veterinary Science (AL4Animals), Department of Veterinary Sciences, School of Agrarian and Veterinary Sciences (ECAV), University of Trás-os-Montes e Alto Douro (UTAD), Vila Real, Portugal.
| |
Collapse
|
26
|
STOKES CALEB, J. MELVIN ANN. Viral Infections of the Fetus and Newborn. AVERY'S DISEASES OF THE NEWBORN 2024:450-486.e24. [DOI: 10.1016/b978-0-323-82823-9.00034-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
27
|
Chen YN, Hsu KH, Huang CG, Chiang MC, Chu SM, Chen CL, Hsu JF, Chueh HY. Clinical Characteristics of Infants with Symptomatic Congenital and Postnatal Cytomegalovirus Infection-An 11-Year Multicenter Cohort Study in Taiwan. CHILDREN (BASEL, SWITZERLAND) 2023; 11:17. [PMID: 38255331 PMCID: PMC10813870 DOI: 10.3390/children11010017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/12/2023] [Accepted: 12/21/2023] [Indexed: 01/24/2024]
Abstract
(1) Background: Cytomegalovirus (CMV) infection is a prevalent viral disease among infants. The prevalence typically ranges from 0.2% to 2.4% among all newborns. There are limited data regarding the demographic characteristics of infants with symptomatic CMV infections. (2) Methods: In this retrospective cohort study using the Chang Gung Memorial Hospital multicenter database, infants with CMV infection determined by a positive urine culture, positive blood polymerase chain reaction assay or positive immunoglobulin M result for CMV from 2011 through 2021 were included. Clinical characteristics at initial diagnosis, management and outcomes were investigated. Congenital CMV (cCMV) infection is diagnosed within three weeks after birth; postnatal CMV (pCMV) is diagnosed when CMV is detected after the first 3 weeks of life. (3) Results: Among the 505 CMV-infected infants identified, 272 were included in the analysis. According to the age at initial presentation, 21 infants had cCMV infection and 251 had pCMV infection. Higher incidences of prematurity and being small for gestational age and a lower Z score for weight at diagnosis were observed in the cCMV group. While thrombocytopenia (61.9%) was the leading presentation in the cCMV group, hepatitis (59.8%) and prolonged jaundice (21.9%) were more common in the pCMV group. (4) Conclusions: Utilizing an 11-year multicenter database, we demonstrated the characteristics of infants with CMV infection in Taiwan and highlighted the demographic disparities and differing symptoms between the cCMV and pCMV groups. These findings emphasize the necessity for future research to refine screening policies, explore treatment options, and establish follow-up protocols for affected infants.
Collapse
Affiliation(s)
- Yu-Ning Chen
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan 33382, Taiwan; (Y.-N.C.); (K.-H.H.); (M.-C.C.); (S.-M.C.); (J.-F.H.)
| | - Kai-Hsiang Hsu
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan 33382, Taiwan; (Y.-N.C.); (K.-H.H.); (M.-C.C.); (S.-M.C.); (J.-F.H.)
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Graduate Institute of Clinical Medical Science, Chang Gung University, Taoyuan 33302, Taiwan
| | - Chung-Guei Huang
- Department of Laboratory Medicine, Chang Gung Memorial Hospital, Taoyuan 33382, Taiwan;
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Ming-Chou Chiang
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan 33382, Taiwan; (Y.-N.C.); (K.-H.H.); (M.-C.C.); (S.-M.C.); (J.-F.H.)
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Shih-Ming Chu
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan 33382, Taiwan; (Y.-N.C.); (K.-H.H.); (M.-C.C.); (S.-M.C.); (J.-F.H.)
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Chyi-Liang Chen
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan 33382, Taiwan;
| | - Jen-Fu Hsu
- Division of Neonatology, Department of Pediatrics, Chang Gung Memorial Hospital, Taoyuan 33382, Taiwan; (Y.-N.C.); (K.-H.H.); (M.-C.C.); (S.-M.C.); (J.-F.H.)
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Ho-Yen Chueh
- Department of Obstetrics and Gynaecology, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan 33302, Taiwan
| |
Collapse
|
28
|
Gupta A, Lawrence SM, Fraley SI. A broad-based probe-free qPCR assay for detection and discrimination of three human herpes viruses. J Virol Methods 2023; 322:114824. [PMID: 37778538 PMCID: PMC11175599 DOI: 10.1016/j.jviromet.2023.114824] [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: 05/22/2023] [Revised: 09/04/2023] [Accepted: 09/28/2023] [Indexed: 10/03/2023]
Abstract
Primary infection or reactivation of latent human cytomegalovirus (HCMV) or herpes simplex viruses (HSV) 1 or 2 during pregnancy can transmit the virus in utero or during natural childbirth to the fetus. The majority of these infections are asymptomatic at birth but may present later with potentially lethal disseminated infection or meningitis (HSV), or long-term neurodevelopmental sequelae including sensorineural hearing loss or neurodevelopmental impairments (HCMV). Unfortunately, early signs and symptoms of disseminated viral infections may be misdiagnosed as bacterial sepsis. Therefore, immediate testing for viral etiologies may not be ordered or even considered by skilled clinicians. In asymptomatic HCMV infections, early detection is necessary to monitor for and treat future neurologic sequelae. In acutely ill-appearing infants, specific detection of viruses against other disease-causing agents is vital to inform correct patient management, including early administration of the correct antimicrobial(s). An ideal test should be rapid, inexpensive, require low sample volumes, and demonstrate efficacy in multiple tissue matrices to aid in timely clinical decision-making for neonatal infections. This work discusses the development of a rapid probe-free qPCR assay for HSV and HCMV that enables early and specific detection of these viruses in neonates. The assay's probe free chemistry would allow easier extension to a broad-based multiplexed pathogenic panel as compared to assays utilizing sequence-specific probes or nested PCR.
Collapse
Affiliation(s)
- Anshu Gupta
- Jacobs School of Engineering, Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Shelley M Lawrence
- Department of Pediatrics, Division of Neonatology, University of Utah, Salt Lake City, UT, USA.
| | - Stephanie I Fraley
- Jacobs School of Engineering, Department of Bioengineering, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
29
|
Gao F, Mora MC, Constantinides M, Coenon L, Multrier C, Vaillant L, Zhang T, Villalba M. g-NK cells from umbilical cord blood are phenotypically and functionally different than g-NK cells from peripheral blood. Oncoimmunology 2023; 12:2283353. [PMID: 38126036 PMCID: PMC10732642 DOI: 10.1080/2162402x.2023.2283353] [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: 09/13/2023] [Accepted: 11/10/2023] [Indexed: 12/23/2023] Open
Abstract
FcRγ-deficient natural killer (NK) cells, designated as g-NK cells, exhibit enhanced antibody-dependent cellular cytotoxicity (ADCC) capacity and increased IFN-γ and TNF-α production, rendering them promising for antiviral and antitumor responses. g-NK cells from peripheral blood (PB) are often associated with prior human cytomegalovirus (HCMV) infection. However, the prevalence, phenotype, and function of g-NK cells in umbilical cord blood (UCB-g-NK) remain unclear. Here, we demonstrate significant phenotypical differences between UCB-g-NK and PB-g-NK cells. Unlike PB-g-NK cells, UCB-g-NK cells did not show heightened cytokine production upon CD16 engagement, in contrast to the conventional NK (c-NK) cell counterparts. Interestingly, following in vitro activation, UCB-g-NK cells also exhibited elevated levels of IFN-γ production, particularly when co-cultured with HCMV and plasma from g-NK+ adults. Furthermore, g-NK+ plasma from PB even facilitated the in vitro expansion of UCB-g-NK cells. These findings underscore the phenotypic and functional heterogeneity of g-NK cells based on their origin and demonstrate that components within g-NK+ plasma may directly contribute to the acquisition of an adult phenotype by the "immature" UCB-g-NK cells.
Collapse
Affiliation(s)
- Fei Gao
- Department of Pathology, School of Basic Medicine, Central South University, Changsha, China
- IRMB, INSERM, CHRU de Montpellier, University Montpellier, Montpellier, France
| | | | | | - Loïs Coenon
- IRMB, INSERM, CHRU de Montpellier, University Montpellier, Montpellier, France
| | - Caroline Multrier
- IRMB, INSERM, CHRU de Montpellier, University Montpellier, Montpellier, France
| | - Loïc Vaillant
- IRMB, INSERM, CHRU de Montpellier, University Montpellier, Montpellier, France
| | - Tianxiang Zhang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Martin Villalba
- IRMB, INSERM, CHRU de Montpellier, University Montpellier, Montpellier, France
- Institut du Cancer Avignon-Provence Sainte Catherine, Avignon, France
- IRMB, INSERM, CHRU de Montpellier, CNRS, Univ Montpellier, Montpellier, France
| |
Collapse
|
30
|
Chu C, Yu S, Min F, Sun L, Liu M, Meng Q. Establishment and application of a point-of-care testing and diagnosis method for early immediate expression gene IE1 of cytomegalovirus in maternal urine based on isothermal amplification. Virus Res 2023; 337:199229. [PMID: 37769815 PMCID: PMC10579523 DOI: 10.1016/j.virusres.2023.199229] [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: 08/28/2023] [Revised: 09/17/2023] [Accepted: 09/25/2023] [Indexed: 10/03/2023]
Abstract
BACKGROUND Human Cytomegalovirus virus (HCMV) is a worldwide virus that causes no serious symptoms in most adults. However, HCMV infection during pregnancy, it may lead to a series of serious complications, such as hearing loss, mental retardation, visual impairment, microcephaly and developmental retardation. AIM The aim of this study was to develop a simple, low dependence on equipment and accurate method for HCMV detection based on the recombinase polymerase amplification (RPA) and lateral flow chromatography strip (LFS) reading. METHODS In order to meet the feasibility of HCMV early screening, three pairs of RPA primers were designed based on the UL123 gene encoding IE1, which was expressed immediately in the early stage of HCMV. In order to improve the specificity of the reaction and satisfy the visual detection, a specific probe was designed to insert THF site between upstream and downstream primers, fluorescein isothiocyanate (FITC) and C3spacer were used to modify the 5' end and the 3' end respectively, and Biotin was used to modify the 5' end of the reverse primer. HCMV standard strain AD169 was enriched by ARPE-19 cells culture, and its genome was extracted. The primers and probes were screened by RPA-LFS test, and the optimal reaction temperature and time were determined The specificity was verified in different viruses, bacteria and parasites. The standard curve was drawn based on the constructed recombinant plasmid of pMD18T-HCMV-UL123 and used for HCMV genomic DNA quantification and determination of the detection sensitivity. Urine samples from artificial HCMV contamination or clinical collection were prepared to evaluate the consistency with the results of real-time quantitative PCR. RESULTS The results showed that the primers and probes for HCMV RPA-LFS detection based on UL123 gene were successfully screened, the amplification of HCMV genomic DNA with as low as 30 copies could be completed at 37 °C within 15 min, it did not react with Human herpesvirus 1, Streptococcus pyogenes, Candida albicans, Listeria monocytogenes, Y. enterocolitica, Klebsiella Pneumoniae, Enterobacter cloacae, Citrobacter freundii, Vibrio alginnolyfificus, Vibrio parahaemolyticus, S. typhimurium, Staphylococcus aureus, Pseudomonas aeruginosa and Trichomonas vaginalis. The positive rate of PCR was 96.67 % in 30 simulated urine samples and 100 % in 127 clinical urine samples with the same UL123 gene detection. CONCLUSIONS To sum up, we developed a diagnostic method for HCMV based on UL123 gene combined with RPA and LFS, which is low dependent on equipment, fast, sensitive and specific, provide reference for point-of-care testing HCMV in grass-roots laboratories and remote areas.
Collapse
Affiliation(s)
- Chu Chu
- Obstetrical Department, Lianyungang Maternal and Child Health Care Hospital, Lianyungang, Jiangsu 222006, China
| | - Shijiao Yu
- Obstetrical Department, Lianyungang Maternal and Child Health Care Hospital, Lianyungang, Jiangsu 222006, China
| | - Fanli Min
- Obstetrical Department, Lianyungang Maternal and Child Health Care Hospital, Lianyungang, Jiangsu 222006, China
| | - Lizhou Sun
- Obstetrical Department, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 222006, China
| | - Meilin Liu
- Obstetrical Department, Lianyungang Maternal and Child Health Care Hospital, Lianyungang, Jiangsu 222006, China.
| | - Qian Meng
- Obstetrical Department, Lianyungang Maternal and Child Health Care Hospital, Lianyungang, Jiangsu 222006, China.
| |
Collapse
|
31
|
Suominen H, Suominen N, Syrjänen K, Waterboer T, Grénman S, Syrjänen S, Louvanto K. Effect of a Second Pregnancy on the HPV Serology in Mothers Followed Up in the Finnish Family HPV Study. Viruses 2023; 15:2109. [PMID: 37896886 PMCID: PMC10612095 DOI: 10.3390/v15102109] [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: 09/10/2023] [Revised: 10/10/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
The impact of pregnancy on human papillomavirus (HPV) natural antibody levels is not fully understood. We tested the seroprevalence and levels of HPV 6, 11, 16, 18 and 45 antibodies at different time points among 89 women with a second pregnancy and 238 nonpregnant women during their 36-month followup. All participants were unvaccinated for HPV and pregnant at the enrollment of the study. Serum samples were collected from the mothers at baseline and at the 12-month, 24-month, and 36-month followup visits. No statistically significant differences in mean antibody levels were observed in women who developed a second pregnancy compared to their nonpregnant counterparts. Between these two groups, statistically significant differences in serostatus were observed, particularly if the second pregnancy was ongoing at the 24-month timepoint. Accordingly, women with a second pregnancy were more likely to be seronegative for HPV 6, 11, 18, and 45 as compared to the nonpregnant women, the reverse being true for HPV16. In contrast, the women with an ongoing second pregnancy showed a higher prevalence of HPV16 seropositivity at the 36-month followup. These data suggest that a second pregnancy does not seem to have a major impact on the levels of HPV antibodies, but it might influence the serological outcomes.
Collapse
Affiliation(s)
- Helmi Suominen
- Department of Obstetrics and Gynecology, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland;
| | - Nelli Suominen
- Department of Obstetrics and Gynecology, Turku University Hospital and University of Turku, 20014 Turku, Finland; (N.S.); (S.G.)
- Department of Obstetrics and Gynecology, Vaasa Central Hospital, 65130 Vaasa, Finland
| | | | - Tim Waterboer
- Division of Infections and Cancer Epidemiology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
| | - Seija Grénman
- Department of Obstetrics and Gynecology, Turku University Hospital and University of Turku, 20014 Turku, Finland; (N.S.); (S.G.)
| | - Stina Syrjänen
- Department of Pathology, Turku University Hospital, 20014 Turku, Finland;
- Department of Oral Pathology and Oral Radiology, Institute of Dentistry, Faculty of Medicine, University of Turku, 20014 Turku, Finland
| | - Karolina Louvanto
- Department of Obstetrics and Gynecology, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland;
- Department of Oral Pathology and Oral Radiology, Institute of Dentistry, Faculty of Medicine, University of Turku, 20014 Turku, Finland
- Department of Obstetrics and Gynecology, Tampere University Hospital, 33100 Tampere, Finland
| |
Collapse
|
32
|
Creisher PS, Perry JL, Zhong W, Lei J, Mulka KR, Ryan WH, Zhou R, Akin EH, Liu A, Mitzner W, Burd I, Pekosz A, Klein SL. Adverse outcomes in SARS-CoV-2-infected pregnant mice are gestational age-dependent and resolve with antiviral treatment. J Clin Invest 2023; 133:e170687. [PMID: 37581940 PMCID: PMC10575736 DOI: 10.1172/jci170687] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 08/10/2023] [Indexed: 08/17/2023] Open
Abstract
SARS-CoV-2 infection during pregnancy is associated with severe COVID-19 and adverse fetal outcomes, but the underlying mechanisms remain poorly understood. Moreover, clinical studies assessing therapeutics against SARS-CoV-2 in pregnancy are limited. To address these gaps, we developed a mouse model of SARS-CoV-2 infection during pregnancy. Outbred CD1 mice were infected at E6, E10, or E16 with a mouse-adapted SARS-CoV-2 (maSCV2) virus. Outcomes were gestational age-dependent, with greater morbidity, reduced antiviral immunity, greater viral titers, and impaired fetal growth and neurodevelopment occurring with infection at E16 (third trimester equivalent) than with infection at either E6 (first trimester equivalent) or E10 (second trimester equivalent). To assess the efficacy of ritonavir-boosted nirmatrelvir, which is recommended for individuals who are pregnant with COVID-19, we treated E16-infected dams with mouse-equivalent doses of nirmatrelvir and ritonavir. Treatment reduced pulmonary viral titers, decreased maternal morbidity, and prevented offspring growth restriction and neurodevelopmental impairments. Our results highlight that severe COVID-19 during pregnancy and fetal growth restriction is associated with heightened virus replication in maternal lungs. Ritonavir-boosted nirmatrelvir mitigated maternal morbidity along with fetal growth and neurodevelopment restriction after SARS-CoV-2 infection. These findings prompt the need for further consideration of pregnancy in preclinical and clinical studies of therapeutics against viral infections.
Collapse
Affiliation(s)
- Patrick S. Creisher
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Jamie L. Perry
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Weizhi Zhong
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Jun Lei
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Kathleen R. Mulka
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - W. Hurley Ryan
- Department of Environmental Health and Engineering, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Ruifeng Zhou
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Elgin H. Akin
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Anguo Liu
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Wayne Mitzner
- Department of Environmental Health and Engineering, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Irina Burd
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Environmental Health and Engineering, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Sabra L. Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
33
|
Guo L, Liu X, Su X. The role of TEMRA cell-mediated immune senescence in the development and treatment of HIV disease. Front Immunol 2023; 14:1284293. [PMID: 37901239 PMCID: PMC10602809 DOI: 10.3389/fimmu.2023.1284293] [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: 08/28/2023] [Accepted: 09/29/2023] [Indexed: 10/31/2023] Open
Abstract
Human Immunodeficiency Virus (HIV) has plagued human society for a long time since its discovery, causing a large number of patients to suffer and costing hundreds of millions of medical services every year. Scientists have found that HIV and antiretroviral therapy accelerate immune aging by inducing mitochondrial dysfunction, and that terminal effector memory T cells (TEMRA cells) are crucial in immune aging. This specific subset of effector memory T cells has terminally differentiated properties and exhibits high cytotoxicity and proinflammatory capacity. We therefore explored and described the interplay between exhaustion features, essential markers, functions, and signaling pathways from previous studies on HIV, antiretroviral therapy, immune senescence, and TEMRA cells. Their remarkable antiviral capacity is then highlighted by elucidating phenotypic changes in TEMRA cells during HIV infection, describing changes in TEMRA cells before, during, and after antiretroviral therapy and other drug treatments. Their critical role in complications and cytomegalovirus (CMV)-HIV superinfection is highlighted. These studies demonstrate that TEMRA cells play a key role in the antiviral response and immune senescence during HIV infection. Finally, we review current therapeutic strategies targeting TEMRA cells that may be clinically beneficial, highlight their potential role in HIV-1 vaccine development, and provide perspectives and predictions for related future applications.
Collapse
Affiliation(s)
- Lihui Guo
- Department of Burns and Plastic Surgery, Yanbian University Hospital, Yanji, China
- Department of Rheumatology and Immunology, The First Hospital of China Medical University, China Medical University, Shen Yang, China
| | - Xudong Liu
- Department of Rheumatology and Immunology, The First Hospital of China Medical University, China Medical University, Shen Yang, China
| | - Xin Su
- Department of Burns and Plastic Surgery, Yanbian University Hospital, Yanji, China
| |
Collapse
|
34
|
Singh S, Maheshwari A, Boppana S. CMV-induced Hearing Loss. NEWBORN (CLARKSVILLE, MD.) 2023; 2:249-262. [PMID: 38348106 PMCID: PMC10860330 DOI: 10.5005/jp-journals-11002-0081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/15/2024]
Abstract
Congenital cytomegalovirus (cCMV) infection is the most common fetal viral infection and contributes to about 25% of childhood hearing loss by the age of 4 years. It is the leading nongenetic cause of sensorineural hearing loss (SNHL). Infants born to seroimmune mothers are not completely protected from SNHL, although the severity of their hearing loss may be milder than that seen in those whose mothers had a primary infection. Both direct cytopathic effects and localized inflammatory responses contribute to the pathogenesis of cytomegalovirus (CMV)-induced hearing loss. Hearing loss may be delayed onset, progressive or fluctuating in nature, and therefore, a significant proportion will be missed by universal newborn hearing screening (NHS) and warrants close monitoring of hearing function at least until 5-6 years of age. A multidisciplinary approach is required for the management of hearing loss. These children may need assistive hearing devices or cochlear implantation depending on the severity of their hearing loss. In addition, early intervention services such as speech or occupational therapy could help better communication, language, and social skill outcomes. Preventive measures to decrease intrauterine CMV transmission that have been evaluated include personal protective measures, passive immunoprophylaxis and valacyclovir treatment during pregnancy in mothers with primary CMV infection. Several vaccine candidates are currently in testing and one candidate vaccine in phase 3 trials. Until a CMV vaccine becomes available, behavioral and educational interventions may be the most effective strategy to prevent maternal CMV infection.
Collapse
Affiliation(s)
- Srijan Singh
- Department of Neonatology, Kailash Hospital, Noida, Uttar Pradesh, India
- Global Newborn Society (https://www.globalnewbornsociety.org/), Clarksville, Maryland, United States of America
| | - Akhil Maheshwari
- Global Newborn Society (https://www.globalnewbornsociety.org/), Clarksville, Maryland, United States of America
- Department of Pediatrics, Louisiana State University, Shreveport, Louisiana, United States of America
| | - Suresh Boppana
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| |
Collapse
|
35
|
Moglad EH, Hassan AO, Atta Elmanan MS, Saeed SM, Abdalla WM, Mohammedsalih KA, Ali HT, Abd Elaziz MS, Ahmed HH. Seroepidemiological Survey of Cytomegalovirus Infection among Pregnant Women in Sudan. Pol J Microbiol 2023; 72:269-275. [PMID: 37668434 PMCID: PMC10508969 DOI: 10.33073/pjm-2023-026] [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: 03/19/2023] [Accepted: 06/09/2023] [Indexed: 09/06/2023] Open
Abstract
Human Cytomegalovirus (HCMV) is a leading healthcare problem associated with stillbirth and congenital abnormalities. Determining the seroprevalence and the possible risk factors related to HCMV infections may be a cornerstone in preventing its complications. This cross-sectional study was conducted in Kassala and River Nile States to determine the seroprevalence and risk factors associated with HCMV infection in pregnant women. One hundred eighty-four (n = 184) blood specimens were collected from pregnant women from February 2018 to January 2020. Enzyme-linked immunosorbent assay (ELISA) was used to detect HCMV-specific IgG and IgM antibodies. Socio-demographical characteristics of the women were collected using structured questionnaires. The results showed that HCMV IgG was detected in 170 (92.4%) of the blood specimens, and IgM was detected in 29/93 (31.2%). There was a significant relationship between the history of miscarriage and the presence of IgG and IgM with a p-value = 0.001 and between HCMV IgM and gestational stage (p-value = 0.028). The study found a strikingly high seroprevalence of HCMV infections among pregnant women in the investigated States. This high percentage of illiterate housewives living in rural areas makes it possible to reduce the incidence of HCMV infection in pregnant women by improving their knowledge, attitude, and practice regarding the route of viral transmission, which may reflect in lowering the rate of congenital diseases in their infants.
Collapse
Affiliation(s)
- Ehssan H. Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam bin Abdulaziz University, Alkharj, Kingdom of Saudi Arabia
- Department of Microbiology and Parasitology, Institute of Medicinal and Aromatic Plants Research, The National Center for Research, Khartoum, Sudan
| | - Ahmed O. Hassan
- Department of Microbiology, College of Medical Laboratory Science, Sudan University of Science and Technology, Khartoum, Sudan
| | - Mawada S. Atta Elmanan
- Department of Microbiology, College of Medical Laboratory Science, Sudan University of Science and Technology, Khartoum, Sudan
| | - Samar M. Saeed
- Department of Microbiology, College of Medical Laboratory Science, Sudan University of Science and Technology, Khartoum, Sudan
| | - Wafaa Mohammed Abdalla
- Department of Microbiology, College of Medical Laboratory Science, Sudan University of Science and Technology, Khartoum, Sudan
| | | | - Hatim T. Ali
- Department of Obstetrics and Gynecology, Faculty of Medicine, King Khalid University, Abha, Kingdom of Saudi Arabia
| | - Mohammed Siddig Abd Elaziz
- Department of Histology, Pathology and Cytology, College of Medical Laboratory Science, Sudan University of Science and Technology, Khartoum, Sudan
- Scientific Affair, University of Science and Technology, Khartoum, Sudan
| | - Hind Haidar Ahmed
- Department of Microbiology, College of Medical Laboratory Science, Sudan University of Science and Technology, Khartoum, Sudan
| |
Collapse
|
36
|
Aldè M, Binda S, Primache V, Pellegrinelli L, Pariani E, Pregliasco F, Di Berardino F, Cantarella G, Ambrosetti U. Congenital Cytomegalovirus and Hearing Loss: The State of the Art. J Clin Med 2023; 12:4465. [PMID: 37445500 DOI: 10.3390/jcm12134465] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 06/29/2023] [Accepted: 07/02/2023] [Indexed: 07/15/2023] Open
Abstract
In developed countries, congenital cytomegalovirus (cCMV) infection is the most common congenital viral infection, representing the leading non-genetic cause of sensorineural hearing loss (HL). Diagnosis of cCMV infection can be performed by detection of CMV DNA in urine or saliva within 2-3 weeks after birth, or later in dried blood samples on the Guthrie card. Currently, there are many controversies regarding the preventive, diagnostic, and therapeutic approaches to cCMV infection. HL secondary to cCMV is highly variable in onset, side, degree, audiometric configuration, and threshold changes over time. Therefore, it is of paramount importance to perform a long and thorough audiological follow-up in children with cCMV infection to ensure early identification and prompt treatment of progressive and/or late-onset HL. Early cochlear implantation appears to be a valid solution not only for children with bilateral profound HL, but also for those with single-sided deafness, improving localization ability and understanding speech in noisy environments. Moreover, the decision to apply a unilateral cochlear implant in children with cCMV is strengthened by the non-negligible possibility of hearing deterioration of the contralateral ear over time.
Collapse
Affiliation(s)
- Mirko Aldè
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
- Audiology Unit, Department of Specialist Surgical Sciences, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Sandro Binda
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| | - Valeria Primache
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| | - Laura Pellegrinelli
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| | - Elena Pariani
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| | - Fabrizio Pregliasco
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy
| | - Federica Di Berardino
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
- Audiology Unit, Department of Specialist Surgical Sciences, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Giovanna Cantarella
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
- Otolaryngology Unit, Department of Specialist Surgical Sciences, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Umberto Ambrosetti
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| |
Collapse
|
37
|
Lalagkas PN, Iliou J, Rigo R, Miarons M, Fernández-Alarcon B, Bestard O, Cruzado JM, Melilli E, Torras J, Grinyó JM, Lloberas N, Colom H. Comparison of Three Renal Function Formulas for Ganciclovir/Valganciclovir Dose Individualization in CMV-Infected Solid Organ Transplantation Patients Using a Population Approach. Clin Pharmacokinet 2023; 62:861-880. [PMID: 37140726 PMCID: PMC10256675 DOI: 10.1007/s40262-023-01237-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2023] [Indexed: 05/05/2023]
Abstract
BACKGROUND AND OBJECTIVE The gold standard treatment of established cytomegalovirus infection or prevention in solid organ transplantation is the intravenous administration of ganciclovir (GCV) or oral administration of valganciclovir (VGCV), both adjusted to the renal function. In both instances, there is a high interindividual pharmacokinetic variability, mainly owing to the wide range of variation of both the renal function and body weight. Therefore, accurate estimation of the renal function is crucial for GCV/VGCV dose optimization. This study aimed to compare three different formulas for estimating the renal function in solid organ transplantation patients with cytomegalovirus infection, for individualizing antiviral therapy with GCV/VGCV, using a population approach. METHODS A population pharmacokinetic analysis was performed using NONMEM 7.4. A total of 650 plasma concentrations obtained after intravenous GCV and oral VGCV administrations were analyzed, from intensive and sparse sampling designs. Three different population pharmacokinetic models were built with the renal function given by Cockcroft-Gault, Modification of Diet in Renal Disease, or Chronic Kidney Disease EPIdemiology Collaboration (CKD-EPI) formulas. Pharmacokinetic parameters were allometrically scaled to body weight. RESULTS The CKD-EPI formula was identified as the best predictor of between-patient variability in GCV clearance. Internal and external validation techniques showed that the CKD-EPI model had better stability and performed better compared with the others. CONCLUSIONS The model based on the more accurate estimation of the renal function with the CKD-EPI formula and body weight as a size metric most used in the clinical practice can refine initial dose recommendations and contribute to GCV and VGCV dose individualization when required in the prevention or treatment of cytomegalovirus infection in solid organ transplantation patients.
Collapse
Affiliation(s)
- Panagiotis Nikolaos Lalagkas
- Biopharmaceutics and Pharmacokinetics Unit, Department of Pharmacy and Pharmaceutical Technology and Physical-Chemistry, School of Pharmacy and Food Sciences, University of Barcelona, Avda. Joan XXIII, s/n, 08028, Barcelona, Spain
| | - Jorge Iliou
- Biopharmaceutics and Pharmacokinetics Unit, Department of Pharmacy and Pharmaceutical Technology and Physical-Chemistry, School of Pharmacy and Food Sciences, University of Barcelona, Avda. Joan XXIII, s/n, 08028, Barcelona, Spain
| | - Raul Rigo
- Biochemistry Department, IDIBELL, Hospital Universitari de Bellvitge, Barcelona, Spain
| | - Marta Miarons
- Pharmacy Service, Hospital de la Vall d'Hebron, Barcelona, Spain
| | - Beatriz Fernández-Alarcon
- Biopharmaceutics and Pharmacokinetics Unit, Department of Pharmacy and Pharmaceutical Technology and Physical-Chemistry, School of Pharmacy and Food Sciences, University of Barcelona, Avda. Joan XXIII, s/n, 08028, Barcelona, Spain
| | - Oriol Bestard
- Nephrology Service, Hospital Universitari de Bellvitge, IDIBELL, Lab Exp Nephrology 4122, Pav. Govern, 4a planta, UB, Feixa Llarga s/n, L'Hospitalet de Llobregat, Llobregat, 08907, Barcelona, Spain
| | - Josep M Cruzado
- Nephrology Service, Hospital Universitari de Bellvitge, IDIBELL, Lab Exp Nephrology 4122, Pav. Govern, 4a planta, UB, Feixa Llarga s/n, L'Hospitalet de Llobregat, Llobregat, 08907, Barcelona, Spain
| | - Edoardo Melilli
- Nephrology Service, Hospital Universitari de Bellvitge, IDIBELL, Lab Exp Nephrology 4122, Pav. Govern, 4a planta, UB, Feixa Llarga s/n, L'Hospitalet de Llobregat, Llobregat, 08907, Barcelona, Spain
| | - Joan Torras
- Nephrology Service, Hospital Universitari de Bellvitge, IDIBELL, Lab Exp Nephrology 4122, Pav. Govern, 4a planta, UB, Feixa Llarga s/n, L'Hospitalet de Llobregat, Llobregat, 08907, Barcelona, Spain
| | - Josep M Grinyó
- Department of Clinical Sciences, University of Barcelona, Barcelona, Spain
| | - Nuria Lloberas
- Nephrology Service, Hospital Universitari de Bellvitge, IDIBELL, Lab Exp Nephrology 4122, Pav. Govern, 4a planta, UB, Feixa Llarga s/n, L'Hospitalet de Llobregat, Llobregat, 08907, Barcelona, Spain.
| | - Helena Colom
- Biopharmaceutics and Pharmacokinetics Unit, Department of Pharmacy and Pharmaceutical Technology and Physical-Chemistry, School of Pharmacy and Food Sciences, University of Barcelona, Avda. Joan XXIII, s/n, 08028, Barcelona, Spain.
| |
Collapse
|
38
|
Canfield D, Gabby L, Vaziri Fard E, Gyamfi-Bannerman C. Cytomegalovirus in Pregnancy. Obstet Gynecol Clin North Am 2023; 50:263-277. [PMID: 37149309 DOI: 10.1016/j.ogc.2023.02.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Cytomegalovirus is a pervasive DNA herpesvirus that, while clinically insignificant to an immunocompetent adult host, can cause significant morbidity to a congenitally infected fetus. Although detection is often possible with several common ultrasonographic markers and good diagnostic accuracy using polymerase chain reaction testing of amniotic fluid, there are no proven prenatal prevention or antenatal treatment options. Therefore, universal screening is not currently recommended in pregnancy. Strategies that have been studied in the past include immunoglobulins, antivirals, and the development of a vaccine. In this review, we will further discuss the themes above, along with future direction for prevention and treatment.
Collapse
Affiliation(s)
- Dana Canfield
- Division of Maternal Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Diego, CA, USA
| | - Lauryn Gabby
- Division of Maternal Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Diego, CA, USA
| | - Elmira Vaziri Fard
- Department of Pathology and Lab Medicine, University of California, San Diego, CA, USA
| | - Cynthia Gyamfi-Bannerman
- Division of Maternal Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Diego, CA, USA.
| |
Collapse
|
39
|
Trinh QD, Pham NTK, Takada K, Ushijima H, Komine-Aizawa S, Hayakawa S. Roles of TGF-β1 in Viral Infection during Pregnancy: Research Update and Perspectives. Int J Mol Sci 2023; 24:ijms24076489. [PMID: 37047462 PMCID: PMC10095195 DOI: 10.3390/ijms24076489] [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: 03/04/2023] [Revised: 03/25/2023] [Accepted: 03/28/2023] [Indexed: 04/14/2023] Open
Abstract
Transforming growth factor-beta 1 (TGF-β1) is a pleiotropic growth factor playing various roles in the human body including cell growth and development. More functions of TGF-β1 have been discovered, especially its roles in viral infection. TGF-β1 is abundant at the maternal-fetal interface during pregnancy and plays an important function in immune tolerance, an essential key factor for pregnancy success. It plays some critical roles in viral infection in pregnancy, such as its effects on the infection and replication of human cytomegalovirus in syncytiotrophoblasts. Interestingly, its role in the enhancement of Zika virus (ZIKV) infection and replication in first-trimester trophoblasts has recently been reported. The above up-to-date findings have opened one of the promising approaches to studying the mechanisms of viral infection during pregnancy with links to corresponding congenital syndromes. In this article, we review our current and recent advances in understanding the roles of TGF-β1 in viral infection. Our discussion focuses on viral infection during pregnancy, especially in the first trimester. We highlight the mutual roles of viral infection and TGF-β1 in specific contexts and possible functions of the Smad pathway in viral infection, with a special note on ZIKV infection. In addition, we discuss promising approaches to performing further studies on this topic.
Collapse
Affiliation(s)
- Quang Duy Trinh
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Ngan Thi Kim Pham
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Kazuhide Takada
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Hiroshi Ushijima
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Shihoko Komine-Aizawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Satoshi Hayakawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| |
Collapse
|
40
|
Sartori P, Egloff C, Hcini N, Vauloup Fellous C, Périllaud-Dubois C, Picone O, Pomar L. Primary, Secondary, and Tertiary Prevention of Congenital Cytomegalovirus Infection. Viruses 2023; 15:v15040819. [PMID: 37112800 PMCID: PMC10146889 DOI: 10.3390/v15040819] [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: 01/09/2023] [Revised: 02/11/2023] [Accepted: 03/20/2023] [Indexed: 04/29/2023] Open
Abstract
Cytomegalovirus infection is the most common congenital infection, affecting about 1% of births worldwide. Several primary, secondary, and tertiary prevention strategies are already available during the prenatal period to help mitigate the immediate and long-term consequences of this infection. In this review, we aim to present and assess the efficacy of these strategies, including educating pregnant women and women of childbearing age on their knowledge of hygiene measures, development of vaccines, screening for cytomegalovirus infection during pregnancy (systematic versus targeted), prenatal diagnosis and prognostic assessments, and preventive and curative treatments in utero.
Collapse
Affiliation(s)
- Pauline Sartori
- School of Health Sciences (HESAV), University of Applied Sciences and Arts Western Switzerland, 1011 Lausanne, Switzerland
- Department Woman-Mother-Child, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
| | - Charles Egloff
- Assistance Publique-Hôpitaux de Paris APHP, Nord, Service de Gynécologie Obstétrique, Hôpital Louis Mourier, 92700 Colombes, France
- Université de Paris, 75006 Paris, France
- INSERM, IAME, B.P. 416, 75870 Paris, France
| | - Najeh Hcini
- Department of Obstetrics and Gynaecology, West French Guiana Hospital Center, French 97320, Guyana
- CIC Inserm 1424 et DFR Santé Université Guyane, 97320 ST Laurent du Maroni, France
| | - Christelle Vauloup Fellous
- Université Paris-Saclay, INSERM U1193, 94804 Villejuif, France
- Laboratoire de Virologie, AP-HP, Hôpital Paul-Brousse, 94804 Villejuif, France
- Groupe de Recherche sur les Infections Pendant la Grossesse (GRIG), 75000 Paris, France
| | - Claire Périllaud-Dubois
- Université de Paris, 75006 Paris, France
- INSERM, IAME, B.P. 416, 75870 Paris, France
- Virology Laboratory, AP-HP, Sorbonne Université, Hôpital Saint-Antoine, F-75012 Paris, France
| | - Olivier Picone
- Assistance Publique-Hôpitaux de Paris APHP, Nord, Service de Gynécologie Obstétrique, Hôpital Louis Mourier, 92700 Colombes, France
- Université de Paris, 75006 Paris, France
- INSERM, IAME, B.P. 416, 75870 Paris, France
- Groupe de Recherche sur les Infections Pendant la Grossesse (GRIG), 75000 Paris, France
| | - Léo Pomar
- School of Health Sciences (HESAV), University of Applied Sciences and Arts Western Switzerland, 1011 Lausanne, Switzerland
- Department Woman-Mother-Child, Lausanne University Hospital and University of Lausanne, 1011 Lausanne, Switzerland
| |
Collapse
|
41
|
Diena D, Allesina A, Fop F, Mella A, Cavallo R, Costa C, Dolla C, Gallo E, De Rosa FG, Lavacca A, Giraudi R, Mariano F, Biancone L. Relationship between Cytomegalovirus Viremia and Long-Term Outcomes in Kidney Transplant Recipients with Different Donor Ages. Microorganisms 2023; 11:microorganisms11020458. [PMID: 36838423 PMCID: PMC9961719 DOI: 10.3390/microorganisms11020458] [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: 11/01/2022] [Revised: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
OBJECTIVES To explore the Cytomegalovirus (CMV) burden on the long-term post-transplant course in different donor ages, we evaluated the incidence and risk factors for CMV in our kidney-transplanted patients (KTs) with extensive adoption of expanded-criteria donors (ECDs). METHODS Retrospective evaluation of 929 consecutive first KTs (49.5% receiving an organ from a donor ≥ 60 years) performed between 01-2003 and 12-2013. Overall survival was estimated using Kaplan-Meier curves; cumulative incidence function was additionally analyzed to consider the potential role of death with a functioning graft as a competitive event with graft dysfunction and to avoid overestimation. Apart from regular DNAemia monitoring in all patients, prophylaxis was adopted in high-risk groups (D+/R- or recipients of anti-thymocyte globulin induction), with pre-emptive therapy in the remaining groups. RESULTS CMV incidence was 19.5% (4-34.9% according to serostatus combination: D-/R-, D-/R+, D+/R+, D+/R-). Donor and recipient age, recipient pre-transplant hypertension, DR antigen compatibility, cold ischemia time, and post-transplant early complications, including rejection, urologic and renal artery stenosis, and lower renal function and proteinuria ≥ 0.5 g/day at one year after KT were associated with CMV. CMV determined lower death-censored graft survival (DCGS) (p < 0.01), with a prominent effect in R+ (p < 0.01) and without impact in R- (p = 0.32 in D-/R- and p = 0.006 in D+/R-). Interestingly, CMV occurrence influenced DCGS only in KTs who received grafts from donors < 50 or 50-69 years old (p < 0.01), while it was not significant with older donors (p = 0.07). The analysis of the cumulative incidence of graft loss accounting for death as a competing risk confirmed all these findings. In multivariate analysis, CMV replication/disease in the first year was an independent predictor for DCGS (HR 1.73 [1.3-2.3]). CONCLUSIONS In a large population with extensive ECD adoption, CMV viremia in the first year demonstrates its harmful effect with an independent role for graft loss and significant impact among R+ recipients and KTs with donors < 70 years.
Collapse
Affiliation(s)
- Davide Diena
- Renal Transplant Center “A. Vercellone”, Nephrology, Dialysis, and Renal Transplant Division, “Città Della Salute e Della Scienza” Hospital, Department of Medical Sciences, University of Turin, 10126 Turin, Italy
- Renal Unit, Santa Croce e Carle Hospital, 12100 Cuneo, Italy
| | - Anna Allesina
- Renal Transplant Center “A. Vercellone”, Nephrology, Dialysis, and Renal Transplant Division, “Città Della Salute e Della Scienza” Hospital, Department of Medical Sciences, University of Turin, 10126 Turin, Italy
| | - Fabrizio Fop
- Renal Transplant Center “A. Vercellone”, Nephrology, Dialysis, and Renal Transplant Division, “Città Della Salute e Della Scienza” Hospital, Department of Medical Sciences, University of Turin, 10126 Turin, Italy
| | - Alberto Mella
- Renal Transplant Center “A. Vercellone”, Nephrology, Dialysis, and Renal Transplant Division, “Città Della Salute e Della Scienza” Hospital, Department of Medical Sciences, University of Turin, 10126 Turin, Italy
| | - Rossana Cavallo
- Microbiology and Virology Unit, University of Turin, 10126 Turin, Italy
| | - Cristina Costa
- Microbiology and Virology Unit, University of Turin, 10126 Turin, Italy
| | - Caterina Dolla
- Renal Transplant Center “A. Vercellone”, Nephrology, Dialysis, and Renal Transplant Division, “Città Della Salute e Della Scienza” Hospital, Department of Medical Sciences, University of Turin, 10126 Turin, Italy
| | - Ester Gallo
- Renal Transplant Center “A. Vercellone”, Nephrology, Dialysis, and Renal Transplant Division, “Città Della Salute e Della Scienza” Hospital, Department of Medical Sciences, University of Turin, 10126 Turin, Italy
| | - Francesco Giuseppe De Rosa
- Department of Medical Sciences, Infectious Diseases, University of Turin, A.O.U. Città Della Salute e Della Scienza di Torino, 10126 Turin, Italy
| | - Antonio Lavacca
- Renal Transplant Center “A. Vercellone”, Nephrology, Dialysis, and Renal Transplant Division, “Città Della Salute e Della Scienza” Hospital, Department of Medical Sciences, University of Turin, 10126 Turin, Italy
| | - Roberta Giraudi
- Renal Transplant Center “A. Vercellone”, Nephrology, Dialysis, and Renal Transplant Division, “Città Della Salute e Della Scienza” Hospital, Department of Medical Sciences, University of Turin, 10126 Turin, Italy
| | - Filippo Mariano
- Renal Transplant Center “A. Vercellone”, Nephrology, Dialysis, and Renal Transplant Division, “Città Della Salute e Della Scienza” Hospital, Department of Medical Sciences, University of Turin, 10126 Turin, Italy
| | - Luigi Biancone
- Renal Transplant Center “A. Vercellone”, Nephrology, Dialysis, and Renal Transplant Division, “Città Della Salute e Della Scienza” Hospital, Department of Medical Sciences, University of Turin, 10126 Turin, Italy
- Correspondence: ; Tel.: +39-011-6336797
| |
Collapse
|
42
|
Liao EN, Stephans J, Taketa E, Mohamad NI, Kaur Khalsa I, Naugle K, Chan DK. Factors associated with congenital cytomegalovirus infection detected by dried blood spot testing in children with hearing loss. Int J Pediatr Otorhinolaryngol 2023; 165:111446. [PMID: 36657329 DOI: 10.1016/j.ijporl.2023.111446] [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: 06/13/2022] [Revised: 12/11/2022] [Accepted: 01/06/2023] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Congenital cytomegalovirus (cCMV) infection in infants leads to an increased risk of developing sensorineural hearing loss (SNHL), even if they are asymptomatic at birth. There are currently no national guidelines for universal screening for CMV, placing children with cCMV at a high risk for unidentified and untreated HL, which in turn places them at greater risk for lasting impacts on quality of life and cognitive and behavioral abilities. We sought to describe the sociodemographic and hearing loss characteristics of children with HL due to cCMV. DESIGN We performed a retrospective cohort study of patients 0-18 years of age who completed CMV dried blood spot (DBS) testing in our HL clinic before April 1, 2022. Home ZIP codes were entered into the Healthy Places Index (HPI) database to quantify the health of the community in which the patient lived. Severity of HL was determined by pure tone averages (PTA) of hearing thresholds for frequencies of 500Hz, 1000Hz, 2000Hz, and 4000Hz. Progression was defined as those who referred on newborn hearing screen and then had a >15 dB increase in PTA, and those who passed newborn hearing screen and were found to have HL later in life. Logistic regression was used to compare variables. RESULTS Of 365 children who received a CMV DBS test, 15 (4%) had a positive test, indicating the presence of cCMV infection, and 350 (96%) had a negative test. 192 (53%) were male, 212 (58%) were URM, 202 (55%) had public insurance, the median number of ICD-10 codes was 2 (range 0-53), and the median HPI percentile score was 71.2 (range: 3.4-99.9). Although CMV DBS testing was ordered for those with suspicion of SNHL, ultimately diagnostic testing found 333 (91%) with SNHL, 4 (1%) with CHL, 13 (4%) with mixed HL, 9 (3%) with auditory neuropathy spectrum disorder, and 5 (4%) with unspecified HL, and 11 (3%) without HL. Of the 353 patients with HL, 126 (36%) had unilateral, 156 (44%) had symmetric bilateral, and 71 (20%) had asymmetric bilateral HL; 183 (52%) had progressive and 138 (39%) had stable HL. In children with SNHL (n = 333), we tested the association of socio-demographic and audiologic factors with cCMV. Those with asymmetric bilateral SNHL (OR 5.19, 95% CI 1.81-14.90) or profound SNHL (>90 dB) in either ear (OR 13.91, 95% CI 3.82-50.67) had higher odds of having cCMV. Those with symmetric bilateral SNHL had lower odds of a positive CMV DBS test result (OR 0.17, 95% CI 0.02-0.76). All sociodemographic variables, medical comorbidities, and other audiologic variables were not associated with CMV DBS test results. CONCLUSION Congenital CMV infection is associated with asymmetric bilateral and profound SNHL. More research is warranted to determine best practices for universal screening for cCMV to identify these children.
Collapse
Affiliation(s)
- Elizabeth N Liao
- Department of Otolaryngology-Head & Neck Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Jihyun Stephans
- Department of Otolaryngology-Head & Neck Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Emily Taketa
- Department of Otolaryngology-Head & Neck Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Noura I Mohamad
- Department of Otolaryngology-Head & Neck Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Inderpreet Kaur Khalsa
- Department of Otolaryngology-Head & Neck Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Kendyl Naugle
- Department of Otolaryngology-Head & Neck Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Dylan K Chan
- Department of Otolaryngology-Head & Neck Surgery, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
43
|
Mappa I, De Vito M, Flacco ME, di Mascio D, D'Antonio F, Rizzo G. Prenatal predictors of adverse perinatal outcome in congenital cytomegalovirus infection: a retrospective multicenter study. J Perinat Med 2023; 51:102-110. [PMID: 35993868 DOI: 10.1515/jpm-2022-0286] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 07/07/2022] [Indexed: 01/17/2023]
Abstract
OBJECTIVES To identify predictors of adverse perinatal outcome in congenital cytomegalovirus (CMV) infection. METHODS In a multicenter study fetuses with congenital CMV infection diagnosed by PCR on amniotic fluid and normal prenatal imaging at the time of diagnosis were included. Primary outcome was the occurrence of structural anomalies at follow-up ultrasound or prenatal magnetic resonance imaging (MRI). Secondary outcomes were the occurrence of anomalies detected exclusively postnatally and the rate of symptomatic infection. RESULTS One hundred and four fetuses with congenital CMV were included in the study. Anomalies were detected at follow-up ultrasound or MRI in 18.3% (19/104) cases. Additional anomalies were found after birth in 11.9% (10/84) of cases and 15.5% (13/85) of newborns showed clinical symptoms related to CMV infection. There was no difference in either maternal age (p=0.3), trimester (p=0.4) of infection and prenatal therapy (p=0.4) between fetuses with or whiteout anomalies at follow-up. Conversely, median viral load in the amniotic fluid was higher in fetuses with additional anomalies at follow-up (p=0.02) compared to those without. At multivariate logistic regression analysis, high viral load in the amniotic fluid, defined as ≥100,000 copies/mL was the only independent predictor for the occurrence of anomalies detected exclusively at follow-up ultrasound assessment or MRI, with an OR of 3.12. CONCLUSIONS Viral load in the amniotic fluid is a strong predictor of adverse perinatal outcome in congenital CMV infection. The results of this study emphasize the importance of adequate follow up even in case of negative neurosonography to better predict postnatal adverse outcomes of infected newborns, especially in amniotic fluid high viral load.
Collapse
Affiliation(s)
- Ilenia Mappa
- Department of Obstetrics and Gynecology, Fondazione Policlinico Tor Vergata, University of Rome Tor Vergata, Rome, Italy
| | - Marika De Vito
- Department of Obstetrics and Gynecology, Fondazione Policlinico Tor Vergata, University of Rome Tor Vergata, Rome, Italy
| | | | - Daniele di Mascio
- Department of Maternal and Child Health and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Francesco D'Antonio
- Centre for Fetal Care and High-risk Pregnancy, Department of Obstetrics and Gynecology, University of Chieti, Chiety, Italy
| | - Giuseppe Rizzo
- Department of Obstetrics and Gynecology, Fondazione Policlinico Tor Vergata, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
44
|
Kirschen GW, Burd I. Modeling of vertical transmission and pathogenesis of cytomegalovirus in pregnancy: Opportunities and challenges. FRONTIERS IN VIROLOGY 2023; 3:1106634. [PMID: 36908829 PMCID: PMC9997718 DOI: 10.3389/fviro.2023.1106634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In addition to facilitating nutrient, oxygen, and waste transfer between developing fetus and mother, the placenta provides important immune barrier function against infection. Elucidation of the complexity of placental barrier function at the maternal-fetal interface has been greatly aided through experimental model organism systems. In this review, we focus on models of vertical transmission of cytomegalovirus (CMV), a ubiquitous double-stranded DNA viruses whose vertical transmission during pregnancy can lead to devastating neurological and obstetric sequelae. We review the current evidence related to guinea pig and murine models of congenital CMV infection, discuss the possible translatability of a non-human primate model, and conclude with recently developed technology using human placental organoids.
Collapse
Affiliation(s)
- Gregory W Kirschen
- Department of Gynecology and Obstetrics, The Johns Hopkins Hospital, Baltimore, MD, United States
| | - Irina Burd
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland, Baltimore, MD, United States
| |
Collapse
|
45
|
Challenges, Recent Advances and Perspectives in the Treatment of Human Cytomegalovirus Infections. Trop Med Infect Dis 2022; 7:tropicalmed7120439. [PMID: 36548694 PMCID: PMC9784992 DOI: 10.3390/tropicalmed7120439] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 11/30/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Human cytomegalovirus (HCMV) is ubiquitous worldwide and elicits global health problems. The diseases associated with HCMV are a serious threat to humans, especially for the sick, infant, elderly and immunocompromised/immunodeficient individuals. Although traditional antiviral drugs (e.g., ganciclovir, valganciclovir, cidofovir, foscarnet) can be used to treat or prevent acute HCMV infections, their efficacy is limited because of toxicity, resistance issues, side effects and other problems. Fortunately, novel drugs (e.g., letermovir and maribavir) with less toxicity and drug/cross-resistance have been approved and put on the market in recent years. The nucleic acid-based gene-targeting approaches including the external guide sequences (EGSs)-RNase, the clustered regularly interspaced short palindromic repeats (CRISPRs)/CRISPRs-associated protein 9 (Cas9) system and transcription activator-like effector nucleases (TALENs) have been investigated to remove both lytic and latent CMV in vitro and/or in vivo. Cell therapy including the adoptive T cell therapy (ACT) and immunotherapy have been tried against drug-resistant and recurrent HCMV in patients receiving hematopoietic stem cell transplantation (HSCT) or solid organ transplant (SOT), and they have also been used to treat glioblastoma (GBM) associated with HCMV infections. These newly developed antiviral strategies are expected to yield fruitful results and make a significant contribution to the treatment of HCMV infections. Despite this progress, the nucleic acid-based gene-targeting approaches are still under study for basic research, and cell therapy is adopted in a small study population size or only successful in case reports. Additionally, no current drugs have been approved to be indicated for latent infections. Therefore, the next strategy is to develop antiviral strategies to elevate efficacy against acute and/or latent infections and overcome challenges such as toxicity, resistance issues, and side effects. In this review, we would explore the challenges, recent advances and perspectives in the treatment of HCMV infections. Furthermore, the suitable therapeutic strategies as well as the possibility for compassionate use would be evaluated.
Collapse
|
46
|
Human Maternal-Fetal Interface Cellular Models to Assess Antiviral Drug Toxicity during Pregnancy. REPRODUCTIVE MEDICINE 2022. [DOI: 10.3390/reprodmed3040024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Pregnancy is a period of elevated risk for viral disease severity, resulting in serious health consequences for both the mother and the fetus; yet antiviral drugs lack comprehensive safety and efficacy data for use among pregnant women. In fact, pregnant women are systematically excluded from therapeutic clinical trials to prevent potential fetal harm. Current FDA-recommended reproductive toxicity assessments are studied using small animals which often do not accurately predict the human toxicological profiles of drug candidates. Here, we review the potential of human maternal-fetal interface cellular models in reproductive toxicity assessment of antiviral drugs. We specifically focus on the 2- and 3-dimensional maternal placental models of different gestational stages and those of fetal embryogenesis and organ development. Screening of drug candidates in physiologically relevant human maternal-fetal cellular models will be beneficial to prioritize selection of safe antiviral therapeutics for clinical trials in pregnant women.
Collapse
|
47
|
Walsh CJ, Rosenberg SL, Hale EW. Obstetric complications in mothers with ADHD. FRONTIERS IN REPRODUCTIVE HEALTH 2022; 4:1040824. [PMID: 36419963 PMCID: PMC9678343 DOI: 10.3389/frph.2022.1040824] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/18/2022] [Indexed: 10/22/2023] Open
Abstract
BACKGROUND There is currently limited research on the intersection of pregnancy and ADHD and the unique pregnancy risk factors for mothers with an ADHD diagnosis. With an increased population of patients with ADHD in the recent decades and an increase in ADHD medication use during pregnancy it is important to consider what unique risks mothers with ADHD face during the perinatal period. OBJECTIVE Investigate a variety of outcomes in maternal ADHD. METHODS We identified female patients with a diagnosis of pregnancy and ADHD diagnosis. We also further separated the ADHD cohort for separate sub-analyses based on medication type. Odds ratios and relative risk were calculated from outcome incidence within each cohort. Cohorts were balanced on age, sex, and race. RESULTS We identified 45,737 pregnant females with ADHD. We matched these patients to pregnant females without ADHD, for a total of 42,916 pairs. Compared to the group without ADHD, mothers with ADHD had higher rates of every outcome except for HPV infection, which was statistically insignificant (P = 0.768). The odds ratios ranged from 1.08 for anemia complicating pregnancy to 2.63 for depressive episodes. Most outcomes were between 1.2 and 1.8 times more likely to occur in the cohort with ADHD. CONCLUSION This study presents substantial advancements in our knowledge of pregnancy-related ADHD care. Armed with an increased awareness of these potential complications and their relationship with ADHD, obstetricians, psychiatrists, and providers of all specialties may be able to reduce the rate of complications within this specific patient population.
Collapse
Affiliation(s)
| | | | - Elijah W. Hale
- School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
48
|
Plotogea M, Isam AJ, Frincu F, Zgura A, Bacinschi X, Sandru F, Duta S, Petca RC, Edu A. An Overview of Cytomegalovirus Infection in Pregnancy. Diagnostics (Basel) 2022; 12:diagnostics12102429. [PMID: 36292118 PMCID: PMC9600407 DOI: 10.3390/diagnostics12102429] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/22/2022] [Accepted: 10/03/2022] [Indexed: 11/16/2022] Open
Abstract
The objective of this review was to bring to attention cytomegalovirus (CMV) infection during pregnancy, taking into consideration all relevant aspects, such as maternal diagnosis, fetal infection and prevention, prenatal diagnosis, and postnatal prognosis. A literature review was performed regarding adult and congenital infection. General information regarding this viral infection and potential related medical conditions was provided, considering the issues of maternal infection during pregnancy, transmission to the fetus, and associated congenital infection management. Prenatal diagnosis includes maternal serum testing and the confirmation of the infection in amniotic fluid or fetal blood. Additionally, prenatal diagnosis requires imaging techniques, ultrasound, and complementary magnetic resonance to assess cortical and extracortical anomalies. Imaging findings can predict both fetal involvement and the postnatal prognosis of the newborn, but they are difficult to assess, even for highly trained physicians. In regard to fetal sequelae, the early diagnosis of a potential fetal infection is crucial, and methods to decrease fetal involvement should be considered. Postnatal evaluation is also important, because many newborns may be asymptomatic and clinical anomalies can be diagnosed when sequelae are permanent.
Collapse
Affiliation(s)
- Mihaela Plotogea
- Department of Obstetrics and Gynecology, “Nicolae Malaxa” Clinical Hospital, 022441 Bucharest, Romania
| | - Al Jashi Isam
- Faculty of Medicine, “Titu Maiorescu” University, 031593 Bucharest, Romania
- Correspondence: (A.J.I.); (F.F.)
| | - Francesca Frincu
- Department of Obstetrics and Gynecology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Correspondence: (A.J.I.); (F.F.)
| | - Anca Zgura
- Department of Obstetrics and Gynecology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Xenia Bacinschi
- Department of Obstetrics and Gynecology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Florica Sandru
- Department of Obstetrics and Gynecology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Simona Duta
- Department of Obstetrics and Gynecology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Razvan Cosmin Petca
- Department of Obstetrics and Gynecology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Antoine Edu
- Department of Obstetrics and Gynecology, “Nicolae Malaxa” Clinical Hospital, 022441 Bucharest, Romania
- Department of Obstetrics and Gynecology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
49
|
Jin Y, Liu X, Chen S, Xiang J, Peng Z, Sun Y. Analysis of the Results of Cytomegalovirus Testing Combined with Genetic Testing in Children with Congenital Hearing Loss. J Clin Med 2022; 11:jcm11185335. [PMID: 36142981 PMCID: PMC9504080 DOI: 10.3390/jcm11185335] [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: 07/22/2022] [Revised: 09/04/2022] [Accepted: 09/09/2022] [Indexed: 11/28/2022] Open
Abstract
To improve the etiological diagnosis of congenital hearing loss by combining whole-exome sequencing (WES) with cytomegalovirus (CMV) testing and to explore the potential benefits of adding CMV screening to newborn hearing screening, 80 children under 2 years of age with bilateral sensorineural hearing loss were recruited. Peripheral venous blood was extracted from the children for WES analysis. Saliva after mouthwash and the first urine in the morning were collected and used as samples to quantify CMV DNA copy number in urine and saliva by qPCR; among the 80 children with congenital deafness, 59 (74%) were found to have genetic variants that may cause congenital deafness, including 44 with GJB2 or SLC26A4 gene variant, 1 with STRC gene variant, and 14 with other genetic variants. A total of 12 children carried deafness gene variants associated with a syndrome; CMV test results showed that in two children, the CMV DNA copy number in saliva was >1000/mL, which indicates that they were CMV-positive, and their genetic test results were negative. A neonatal CMV test combined with genetic screening can improve the etiological diagnosis rate of congenital deafness, and the direct evidence of neonatal CMV infection deserves further verification.
Collapse
Affiliation(s)
- Yuan Jin
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaozhou Liu
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Sen Chen
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jiale Xiang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhiyu Peng
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yu Sun
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Correspondence:
| |
Collapse
|
50
|
Evaluation of neurological and cardiac development of newborn infants born to mothers infected with COVID-19. JOURNAL OF SURGERY AND MEDICINE 2022. [DOI: 10.28982/josam.7293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Background/Aim: In the coronavirus disease 2019 (COVID-19) pandemic, which has been affecting the world for the last 2 years, pulmonary, cardiovascular, and neurological adverse effects of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have been reported. These negative influences entail a risk for fetal progress. In this study, by performing a detailed clinical evaluation, postnatal ultrasonography, and echocardiography, we aimed to investigate potential neurological and cardiac complications of newborns born to pregnant women infected with COVID-19.
Methods: This prospective and cross-sectional study was conducted between January and July 2021. Newborn infants (0–28 days postpartum) born to mothers with proven COVID-19 infection by positive RT-PCR test during pregnancy were enrolled. Fetal cardiac development was evaluated by a pediatric cardiologist with an echocardiographic examination. Fetal neurologic evaluation was performed by a pediatric neurologist using both neurologic examination and transfontanelle ultrasonography (TFUS). Infants were reevaluated every 2 months until 6 months of age.
Results: Thirty-three female and 32 male infants born to 64 pregnant women, one being a twin birth, were included in the study. Seven women developed COVID-19 infection in the first trimester, 11 in the second trimester, and 46 in the third trimester. Neurological examination and TFUS were normal in all newborns except one with microcephaly. The etiologic cause could not be detected in this infant, and his neurodevelopment was normal in the follow-up. The cardiac examination did not reveal any significant disorders. Eleven infants failed the standard “Auditory Brainstem Response” (ABR) hearing screening test, so a second test was performed. Only two infants required further investigation after the second test.
Conclusion: We did not observe any neurologic and cardiologic teratogenic effects associated with COVID-19 infection during pregnancy.
Collapse
|