1
|
Barlinn R, Helbig A, Rønnestad A, Hilde K, Klingenberg C, Lang AM. Congenital cytomegalovirus infection can be prevented. TIDSSKRIFT FOR DEN NORSKE LEGEFORENING 2025; 145:25-0092. [PMID: 40272141 DOI: 10.4045/tidsskr.25.0092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025] Open
|
2
|
Kang H, Wang L, Chen Y, Dong T, Pan Y, Liu Q. Screening and prenatal diagnosis of fetal cytomegalovirus infection: experience in a western Chinese city. BMC Infect Dis 2025; 25:542. [PMID: 40240971 PMCID: PMC12004672 DOI: 10.1186/s12879-025-10910-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: 03/03/2025] [Accepted: 04/03/2025] [Indexed: 04/18/2025] Open
Abstract
OBJECTIVE To explore the application value of serology testing and fetal ultrasound examination in screening for fetal cytomegalovirus(CMV) infection. To explore the application value of CMV polymerase chain reaction (PCR) in amnio fluids in prenatal diagnosis of fetal CMV infection. METHODS This is a retrospective study performed at Chengdu Women's and Children's Central Hospital between 2021 and 2024. The baseline of preconception CMV infections and recent infections were investigated via serology testing. Pregnant women with suspicious serology for recent infection and/or ultrasound abnormalities suggestive of fetal infection underwent amniocentesis to obtain amniotic fluid for CMV PCR. The pregnancy outcomes of women with suspicious serology for recent infection and those who underwent amniocentesis were analyzed. RESULTS The seroprevalence was 95.65%(55262/57778) and 0.99%(571/57778) for immunoglobulin G(IgG) and immunoglobulin M (IgM) antibodies respectively. Among the 315 pregnant women with positive IgM results who performed CMV IgG avidity index and/or dynamic IgM/ IgG tests, only 2.22%(7/315) confirmed recent infection and 2.54%(8/315) suspected recent infection: three terminated pregnancies including one with positive CMV PCR results and two with other reasons, and 12 gave birth to asymptomatic babies. 576 pregnant women with suspicious serology for recent infection and/or ultrasound abnormalities suggestive of fetal infection performed CMV PCR in amniotic fluid. Among them, we got 438 pregnancy outcomes, including 25 terminated pregnancies, eight stillbirths, and 405 live births. None of the 405 live births had symptoms related to CMV infection at birth, except two with mild hearing loss without known cause. Four fetal infections were diagnosed, including three who performed amniocentesis and CMV PCR and one accidentally diagnosed by copy number variation sequencing(CNV-seq). Three of them were negative for IgM in first-trimester screening. CONCLUSION Ultrasound screening plays a more important role than serology screening in areas with a high seroprevalence of CMV antibodies. Assays for both chromosome disorders and CMV PCR in amniotic fluids should be suggested to pregnant women with ultrasound abnormalities suggestive of fetal infection.
Collapse
Affiliation(s)
- Han Kang
- Prenatal diagnosis department, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, 1617 RiYue Road, Chengdu, China
| | - Lingxi Wang
- Prenatal diagnosis department, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, 1617 RiYue Road, Chengdu, China
| | - Yifei Chen
- Prenatal diagnosis department, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, 1617 RiYue Road, Chengdu, China
| | - Ting Dong
- Prenatal diagnosis department, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, 1617 RiYue Road, Chengdu, China
| | - Yuchun Pan
- Prenatal diagnosis department, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, 1617 RiYue Road, Chengdu, China
| | - Qingsong Liu
- Prenatal diagnosis department, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, 1617 RiYue Road, Chengdu, China.
| |
Collapse
|
3
|
Coskun E, Kakkar F, Riley LE, Ciaranello AL, Prabhu M. Evaluation and Management of Congenital Cytomegalovirus Infection. Obstet Gynecol 2025; 145:297-306. [PMID: 39847776 DOI: 10.1097/aog.0000000000005840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/12/2024] [Indexed: 01/25/2025]
Abstract
The purpose of this review is to serve as an update on congenital cytomegalovirus (CMV) evaluation and management for obstetrician-gynecologists and to provide a framework for counseling birthing people at risk for or diagnosed with a primary CMV infection or reactivation or reinfection during pregnancy. A DNA virus, CMV is the most common congenital viral infection and the most common cause of nongenetic childhood hearing loss in the United States. The risk of congenital CMV infection from transplacental viral transfer depends on the gestational age at the time of maternal infection and whether the infection is primary or nonprimary. Although the risk of congenital CMV infection is lower with infection at earlier gestational ages, clinical sequelae are more severe with maternal infections earlier in gestation. At present, routine screening for maternal CMV infection is not recommended by U.S. guidelines. When maternal primary infection is confirmed in early pregnancy, emerging data support consideration of maternal antiviral therapy to prevent congenital CMV infection. When congenital CMV infection is confirmed, typically after an abnormal prenatal ultrasound result, there are more limited data on the utility of maternal antiviral therapy. Universal newborn screening for congenital CMV infection is not mandatory in most U.S. states at present. Newborns diagnosed with congenital CMV infection undergo an extensive evaluation to determine whether neurologic symptoms are present, which guides postnatal evaluation and management. In this review, we discuss the diagnosis and management of maternal CMV infection, the risk and diagnosis of congenital CMV infection, prevention and potential treatment of congenital CMV infection in utero, and neonatal congenital CMV infection diagnosis and management.
Collapse
Affiliation(s)
- Elif Coskun
- Medical Practice Evaluation Center, the Division of Infectious Disease, and the Division of Maternal Fetal Medicine, Massachusetts General Hospital, Boston, Massachusetts; the Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine, University of Montreal, Montreal, Quebec, Canada; and the Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, New York
| | | | | | | | | |
Collapse
|
4
|
Govind S, Lang PO, Bürkle A, Moreno-Villanueva M, Franceschi C, Capri M, Bernard J, Weinberger B, Grubeck-Loebenstein B, Fiegl S, Gonos ES, Sikora E, Jansen E, Dollé MET, Grune T, Breusing N, Aspinall R. Detection of HHV-5 HHV-6a HHV-6b and HHV-7 in the urine: potential use as a non-invasive diagnostic tool for immune profiling. Immun Ageing 2024; 21:84. [PMID: 39609853 PMCID: PMC11606101 DOI: 10.1186/s12979-024-00490-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 11/15/2024] [Indexed: 11/30/2024]
Abstract
Decline in immune function with age has been studied extensively, but approaches to immune restoration have been hampered by the lack of simple methods of identifying individuals whose immune system is in decline. Our approach has been to identify individuals whose immune decline has led to a loss of control of common latent viral infections and their consequent reactivation. Viruses excreted in urine were detected and quantified and we believe this approach could provide a 'surrogate marker' for identifying immune compromised individuals. Here we report the detection of human herpes virus (HHV) 5, 6a, 6b and 7 in the urine of healthy individuals over a wide age range and their correlation with T cell receptor excision circle (TREC) data. The results did not show a clear correlation between TREC values and the detection of individual specific viruses or viral load values when measured singly. However, a correlation was found between low TREC values and the detection of several different human herpes viruses in the urine in males. We present evidence suggesting that for males, the detection of three or more different human herpes viruses in the urine could identify individuals with declining immune function as evidenced by their significantly lower TREC levels.
Collapse
Affiliation(s)
- Shelia Govind
- Medicines and Healthcare Products Regulatory Agency (MHRA), South Mimms Laboratories, Potters Bar, UK
| | | | - Alexander Bürkle
- Molecular Toxicology Group, Department of Biology, University of Konstanz, Box 628, Konstanz, 78457, Germany
| | - María Moreno-Villanueva
- Molecular Toxicology Group, Department of Biology, University of Konstanz, Box 628, Konstanz, 78457, Germany
- Human Performance Research Centre, Department of Sport Science, University of Konstanz, Box 30, Konstanz, 78457, Germany
| | - Claudio Franceschi
- Institute of Biogerontology, Lobachevsky State University, Nizhny Novgorod, Russia
| | - Miriam Capri
- Department of Medical and Surgical Sciences, University of Bologna-Alma Mater Studiorum, Bologna, Italy
- Alma Mater Research Institute On Global Challenges and Climate Change (Alma Climate), University of Bologna, Bologna, Italy
| | | | - Birgit Weinberger
- Institute for Biomedical Aging Research, Universität Innsbruck, Innsbruck, Austria
| | | | | | | | - Ewa Sikora
- Polish Academy of Sciences, Warsaw, Poland
| | - Eugène Jansen
- Centre for Health Protection, National Institute for Public Health and the Environment, PO Box 1, 3720 BA Bilthoven, Utrecht, The Netherlands
| | - Martijn E T Dollé
- National Institute for Public Health and the Environment, Utrecht, The Netherlands
| | - Tilman Grune
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, 14558, Germany
| | - Nicolle Breusing
- Department of Applied Nutritional Science/Dietetics, Institute of Nutritional Medicine, University of Hohenheim, 70599, Stuttgart, Germany
| | - Richard Aspinall
- Centre for Intelligent Healthcare, Coventry University, Priory Street, COVENTRY, CV1 5FB, Conventry, UK.
| |
Collapse
|
5
|
Supplisson O, Visseaux B, Haim-Boukobza S, Boutolleau D, Alizon S, Burrel S, Sofonea MT. Seroprevalence of human herpes viruses in France, 2018-2022: a multilevel regression and poststratification approach. Infect Dis (Lond) 2024; 56:931-945. [PMID: 38946531 DOI: 10.1080/23744235.2024.2365906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 07/02/2024] Open
Abstract
BACKGROUND Information related to herpes simplex virus 1 and 2 (HSV-1 and 2), varicella-zoster virus (VZV), Epstein-Barr virus (EBV), and cytomegalovirus (CMV) seroprevalence in France is either lacking, incomplete, or outdated, despite their public health burden. METHOD We used routinely collected serological data between 2018 and 2022 to estimate HSV-1, HSV-2, VZV, EBV, and CMV seroprevalence in France. To account for demographic differences between our analytic samples and the French population and get estimates for sparsely sampled districts and age classes, we used a multilevel regression and poststratification approach combined with Bayesian model averaging via stacking weights. RESULTS The observed seroprevalence (number of positive tests/number of tests) were 64.6% (93,294/144,424), 16.9% (24,316/144,159), 93.0% (141,419/152,084), 83.4% (63,199/75, 781), and 49.0% (23,276/47,525), respectively, for HSV-1, HSV-2, VZV, EBV, and CMV. Between 2018 and 2022, France had a model-based average (equal-tailed interval at 95%) expected seroprevalence equal to 61.1% (60.7,61.5), 14.5% (14.2,14.81), 89.5% (89.3,89.8), 85.6% (85.2,86.0), and 50.5% (49.3,51.7), respectively, for HSV-1, HSV-2, VZV, EBV, and CMV infections. We found an almost certain lower expected seroprevalence in Metropolitan France than in overseas territories for all viruses but VZV, for which it was almost certainly greater. The expected seroprevalences were likely greater among females for all viruses. LIMITATIONS Our results relied on the assumption that individuals were sampled at random conditionally to variables used to build the poststratification table. IMPLICATIONS The analysis highlights spatial and demographic patterns in seroprevalence that should be considered for designing tailored public health policies.
Collapse
Affiliation(s)
- Olivier Supplisson
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, Paris, France
- Sorbonne Université, Paris, France
| | | | | | - David Boutolleau
- AP-HP, Sorbonne Université, Centre National de Référence Herpèsvirus (laboratoire Associé), Service de Virologie, Hôpital Pitié-Salpêtrière, Paris, France
- Sorbonne Université, INSERM UMR-S 1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique (iPLESP), Paris, France
| | - Samuel Alizon
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS, INSERM, Université PSL, Paris, France
| | - Sonia Burrel
- CHU de Bordeaux, Service de virologie, Bordeaux, France
- CNRS UMR 5234, Fundamental Microbiology and Pathogenicity, Université de Bordeaux, Bordeaux, France
| | - Mircea T Sofonea
- Pathogenesis and Control of Chronic and Emerging Infections (PCCEI), Université de Montpellier, Inserm, EFS, Montpellier, France and CHU de Nîmes, Nîmes, France
| |
Collapse
|
6
|
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
|
7
|
Vicente M, Carocha AI, Rijo C, Cohen Á, Martins ML. Cytomegalovirus congenital infection: long-term outcomes in a valaciclovir treated population. J Perinat Med 2024; 52:778-782. [PMID: 38943320 DOI: 10.1515/jpm-2023-0535] [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: 12/15/2023] [Accepted: 05/18/2024] [Indexed: 07/01/2024]
Abstract
OBJECTIVES This case series aims to evaluate the long-term outcomes of congenital cytomegalovirus (CMV) infection in a population treated with valaciclovir during pregnancy. The study focuses on assessing the prevalence of long-term sequelae in infants with confirmed CMV fetal infection. METHODS A retrospective analysis was conducted on 33 pregnancies corresponding to 34 fetus with confirmed CMV congenital infection. They were followed from November 2004 to December 2020. Valaciclovir treatment was initiated after confirmation of fetal infection, and fetal outcomes were monitored through serial ultrasounds, neurosonography, and fetal magnetic resonance imaging (MRI). Postnatal assessments included: PCR confirmation, symptoms evaluation at birth, and long-term follow-up protocols for visual, auditory, and neurodevelopmental assessment. RESULTS Therapy was started at a median gestational age of 24 weeks. Of the 34 newborns 79.4 % were asymptomatic at birth. Median follow-up time was 6 years and 32.35 % developed long-term sequelae. Neurosensorial hearing loss (SNHL) was the predominant sequelae. In the cases which developed sequelae 54.5 % had imaging findings, and all with major findings developed long-term sequelae. CONCLUSIONS In our treated population we had a higher asymptomatic rate at birth comparing with a non-treated population, similar to those found in previous studies. We had a long-term sequelae rate of 32.35 %, similar to recent studies on non-treated population, although we registered a slightly lower rate of SNHL. A larger multicenter studies with a longer follow-up time, where treatment is started in the first trimester, is of the utmost importance, so we can truly understand the correlation between these imaging findings, therapy and long-term sequelae.
Collapse
Affiliation(s)
- Maria Vicente
- Fetal Medicine and Surgery Department, Maternidade Dr. Alfredo da Costa - Unidade Local de Saúde de São José , Lisboa, Portugal
| | - Ana I Carocha
- Fetal Medicine and Surgery Department, Maternidade Dr. Alfredo da Costa - Unidade Local de Saúde de São José , Lisboa, Portugal
| | - Claúdia Rijo
- Fetal Medicine and Surgery Department, Maternidade Dr. Alfredo da Costa - Unidade Local de Saúde de São José , Lisboa, Portugal
| | - Álvaro Cohen
- Fetal Medicine and Surgery Department, Maternidade Dr. Alfredo da Costa - Unidade Local de Saúde de São José , Lisboa, Portugal
| | - Maria L Martins
- Fetal Medicine and Surgery Department, Maternidade Dr. Alfredo da Costa - Unidade Local de Saúde de São José , Lisboa, Portugal
| |
Collapse
|
8
|
Ma J, Bruce K, Stevenson PG, Farrell HE. Mouse cytomegalovirus lacking sgg1 shows reduced import into the salivary glands. J Gen Virol 2024; 105:002013. [PMID: 39093048 PMCID: PMC11296724 DOI: 10.1099/jgv.0.002013] [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: 05/16/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024] Open
Abstract
Cytomegaloviruses (CMVs) transmit via chronic shedding from the salivary glands. How this relates to the broad cell tropism they exhibit in vitro is unclear. Human CMV (HCMV) infection presents only after salivary gland infection is established. Murine CMV (MCMV) is therefore useful to analyse early infection events. It reaches the salivary glands via infected myeloid cells. Three adjacent spliced genes designated as m131/129 (MCK-2), sgg1 and sgg1.1, positional homologues of the HCMV UL128/130/131 tropism determinants, are implicated. We show that a sgg1 null mutant is defective in infected myeloid cell entry into the salivary glands, a phenotype distinct from MCMV lacking MCK-2. These data point to a complex, multi-step process of salivary gland colonization.
Collapse
Affiliation(s)
- Jiawei Ma
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Australia
| | - Kimberley Bruce
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Australia
| | - Philip G. Stevenson
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Australia
| | - Helen E. Farrell
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Australia
| |
Collapse
|
9
|
Wang Y, Yan M, Zhang P, Wu X, Huang S, Chen S, Rong Y, Sheng Y, Wang Y, Mao G, Chen L, Wang S, Yang B. Structure elucidation and antiviral activity of a cold water-extracted mannogalactofucan Ts1-1A from Trametes sanguinea against human cytomegalovirus in vitro. Carbohydr Polym 2024; 335:122101. [PMID: 38616079 DOI: 10.1016/j.carbpol.2024.122101] [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: 01/17/2024] [Revised: 03/09/2024] [Accepted: 03/26/2024] [Indexed: 04/16/2024]
Abstract
In this study, we purified a partially acetylated heteropolysaccharide (Ts1-1A) from the fruit bodies of Trametes sanguinea Lloyd through cold water extraction and serial chromatographic separation. The purified polysaccharide Ts1-1A (12.8 kDa) was characterized as a branched mannogalactofucan with a backbone of alternately connected 1,3-linked α-Fucp and 1,6-linked α-Galp, which was partially substituted by non-reducing end units of β-Manp at O-2 and O-3 positions of 1,6-linked α-Galp. Ts1-1A showed pronounced anti-human cytomegalovirus activity at the concentration of 200 and 500 μg/mL in systematical assessments including morphological changes, western blotting, qPCR, indirect immunofluorescence and tissue culture infective dose assays. Moreover, Ts1-1A exerted its antiviral activity at two distinct stages of viral proliferation manifesting as significantly inhibiting viral protein (IE1/2 and p52) expression and reducing viral gene (UL123, UL44 and UL32) replication in the HCMV-infected WI-38 cells. At viral attachment stage, Ts1-1A interacted with HCMV and prevented HCMV from attaching to its host cells. While at early phase of viral replication stage, Ts1-1A suppressed HCMV replication by downregulating NQO1 and HO-1 proteins related to oxidative stress as an antioxidant. To sum up, Ts1-1A is a promising anti-HCMV agent which could be developed for HCMV infection prevention and therapy.
Collapse
Affiliation(s)
- Yiran Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, No. 260 Baichuan Street, Hangzhou 311402, PR China
| | - Mengxia Yan
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, No. 260 Baichuan Street, Hangzhou 311402, PR China
| | - Panpan Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, No. 260 Baichuan Street, Hangzhou 311402, PR China
| | - Xinna Wu
- Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou 310030, PR China
| | - Siyang Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, No. 260 Baichuan Street, Hangzhou 311402, PR China
| | - Siru Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, No. 260 Baichuan Street, Hangzhou 311402, PR China
| | - Yizhou Rong
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, No. 260 Baichuan Street, Hangzhou 311402, PR China
| | - Yunjie Sheng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, No. 260 Baichuan Street, Hangzhou 311402, PR China
| | - Yangyang Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, No. 260 Baichuan Street, Hangzhou 311402, PR China
| | - Genxiang Mao
- Zhejiang Provincial Key Lab of Geriatrics & Geriatrics Institute of Zhejiang Province, Department of Geriatrics, Zhejiang Hospital, Hangzhou 310030, PR China.
| | - Libing Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, No. 260 Baichuan Street, Hangzhou 311402, PR China.
| | - Sanying Wang
- Zhejiang Provincial Key Lab of Geriatrics & Geriatrics Institute of Zhejiang Province, Department of Geriatrics, Zhejiang Hospital, Hangzhou 310030, PR China.
| | - Bo Yang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, No. 260 Baichuan Street, Hangzhou 311402, PR China.
| |
Collapse
|
10
|
Coste-Mazeau P, Hamon M, Ribot É, Hantz S, Alain S. [Implementation of screening for cytomegalovirus congenital infection in a French type 3 maternity]. GYNECOLOGIE, OBSTETRIQUE, FERTILITE & SENOLOGIE 2024; 52:403-409. [PMID: 38272182 DOI: 10.1016/j.gofs.2024.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 01/27/2024]
Abstract
OBJECTIVES Congenital cytomegalovirus (CMV) infection is the most common congenital infection and the leading cause of infectious neurosensorial disability in newborns. We wanted to organize the management of women from the beginning of pregnancy allowing access to antenatal treatment with valaciclovir, recognized since 2020 as limiting materno-fetal transmission. To this end, we set up and evaluated the interest of systematic screening for CMV infection in our maternity. We wanted to organize care for women from the very start of pregnancy. METHODS Retrospective and comparative descriptive study carried out at the CHRU de Limoges from July 2017 to December 2019 (targeted screening), then from January 2020 to June 2022, during which period we implemented systematized screening by iterative serologies at the 3rd, 6th, 8th months and before delivery. Our main evaluation criteria were the seroprevalence of CMV infection and the rate of congenital infection. We then described our cases of infection (primary or secondary) during pregnancy. RESULTS CMV seroprevalence in our pregnant women increased significantly from 52.7% (779/1478 women screened) to 58.4% (3852/6599 women screened) between the 2 study periods (P=0.04). We diagnosed 11 infections during the first part of the study vs. 27 during the second, with a significant increase in primary infections from 0.14% (9/6524 births) to 0.37% (24/6426 births) (P=0.008). Only 3 secondary infections were diagnosed during the second study period. The rate of congenital infections remained stable between the 2 study periods (6 children/6524=0.09% vs. 8 children/6426=0.12%; P=0.57). CONCLUSION Our results confirmed the interest of screening for CMV infection, while modifying the screening strategy we had initiated.
Collapse
Affiliation(s)
- Perrine Coste-Mazeau
- Service de gynécologie obstétrique, hôpital Mère-Enfant, CHRU de Limoges, 8, avenue Dominique-Larrey, 87000 Limoges, France; Inserm, RESINFIT, U 1092, université de Limoges, CHU de Limoges, 1, rue du Pr Descottes, 87000 Limoges, France; Service de virologie, Centre national de référence des Herpès virus, CHU de Limoges, 2, avenue Martin-Luther-King, 87000 Limoges, France.
| | - Magali Hamon
- Service de gynécologie obstétrique, hôpital Mère-Enfant, CHRU de Limoges, 8, avenue Dominique-Larrey, 87000 Limoges, France
| | - Élodie Ribot
- Service de virologie, Centre national de référence des Herpès virus, CHU de Limoges, 2, avenue Martin-Luther-King, 87000 Limoges, France
| | - Sébastien Hantz
- Inserm, RESINFIT, U 1092, université de Limoges, CHU de Limoges, 1, rue du Pr Descottes, 87000 Limoges, France; Service de virologie, Centre national de référence des Herpès virus, CHU de Limoges, 2, avenue Martin-Luther-King, 87000 Limoges, France
| | - Sophie Alain
- Inserm, RESINFIT, U 1092, université de Limoges, CHU de Limoges, 1, rue du Pr Descottes, 87000 Limoges, France; Service de virologie, Centre national de référence des Herpès virus, CHU de Limoges, 2, avenue Martin-Luther-King, 87000 Limoges, France.
| |
Collapse
|
11
|
Bottino P, Pastrone L, Zanotto E, Sidoti F, Cavallo R, Costa C. Molecular diagnosis of Cytomegalovirus infection: clinical performance of the Aptima transcription-mediated amplification assay toward conventional qPCR chemistry on whole blood samples. J Clin Microbiol 2024; 62:e0090623. [PMID: 38349144 PMCID: PMC10935658 DOI: 10.1128/jcm.00906-23] [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: 07/17/2023] [Accepted: 01/16/2024] [Indexed: 03/14/2024] Open
Abstract
Human Cytomegalovirus (HCMV) infection is life-threatening for immunocompromised patients. Quantitative molecular assays on whole blood or plasma are the gold standard for the diagnosis of invasive HCMV infection and for monitoring antiviral treatment in individuals at risk of HCMV disease. For these reasons, an accurate standardization toward the WHO 1st International Standard among different centers and diagnostic kits represents an effort for better clinical management of HCMV-positive patients. Herein, we evaluate, for the first time, the performance of a new transcription-mediated amplification (TMA) assay versus quantitative polymerase chain reaction (qPCR) chemistry, used as a routine method, on whole blood samples. A total of 755 clinical whole blood specimens were collected and tested simultaneously with TMA and qPCR assays. The data showed a qualitative agreement of 99.27% for positive quantified samples and 89.39% for those undetected between the two tested methods. Evaluation of viremia in positive samples highlighted a good correlation between TMA and qPCR chemistries in terms of International Units (ΔLog10 IU/mL: -0.29 ± 0.40). The TMA assay showed a significant correlation with qPCR in patients monitored for up to 3 months, thus allowing an accurate assessment of viremia in transplant patients. Therefore, TMA chemistry showed good agreement with qPCR testing, used as a current diagnostic routine. It also offers important advantages, such as FDA approval on plasma and In Vitro Diagnostic (IVD) on both plasma and whole blood, automated workflow with minimal hands-on time, and random access loading, thus enabling a rapid and reliable diagnostic in HCMV-infected patients. IMPORTANCE In this paper, we describe the clinical performance of a novel transcription-mediated amplification (TMA) assay for the detection and quantification of human Cytomegalovirus (HCMV) DNA from whole blood samples. This is a pivotal analysis in immunocompromised patients [transplanted, HIV-positive, and Hematopoietic Stem Cell (HSC) recipients], and molecular tests with high sensitivity and specificity are necessary to evaluate the HCMV viral load in these patients. To our knowledge, this is the first in-depth evaluation of TMA chemistry for HCMV diagnosis on whole blood samples. Moreover, also technical aspects of this assay make it suitable for clinical diagnostics.
Collapse
Affiliation(s)
- Paolo Bottino
- S.C. Microbiology and Virology U, A.O.U. “Città della Salute e della Scienza di Torino”, Turin, Italy
| | - Lisa Pastrone
- S.C. Microbiology and Virology U, A.O.U. “Città della Salute e della Scienza di Torino”, Turin, Italy
| | - Elisa Zanotto
- S.C. Microbiology and Virology U, A.O.U. “Città della Salute e della Scienza di Torino”, Turin, Italy
| | - Francesca Sidoti
- S.C. Microbiology and Virology U, A.O.U. “Città della Salute e della Scienza di Torino”, Turin, Italy
| | - Rossana Cavallo
- S.C. Microbiology and Virology U, A.O.U. “Città della Salute e della Scienza di Torino”, Turin, Italy
| | - Cristina Costa
- S.C. Microbiology and Virology U, A.O.U. “Città della Salute e della Scienza di Torino”, Turin, Italy
| |
Collapse
|
12
|
Su R, Zhang T, Wang H, Yan G, Wu R, Zhang X, Gao C, Li X, Wang C. New sights of low dose IL-2: Restoration of immune homeostasis for viral infection. Immunology 2024; 171:324-338. [PMID: 37985960 DOI: 10.1111/imm.13719] [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: 08/16/2023] [Accepted: 11/07/2023] [Indexed: 11/22/2023] Open
Abstract
Viral infection poses a significant threat to human health. In addition to the damage caused by viral replication, the immune response it triggers often leads to more serious adverse consequences. After the occurrence of viral infection, in addition to the adverse consequences of infection, chronic infections can also lead to virus-related autoimmune diseases and tumours. At the same time, the immune response triggered by viral infection is complex, and dysregulated immune response may lead to the occurrence of immune pathology and macrophage activation syndrome. In addition, it may cause secondary immune suppression, especially in patients with compromised immune system, which could lead to the occurrence of secondary infections by other pathogens. This can often result in more severe clinical outcomes. Therefore, regarding the treatment of viral infections, restoring the balance of the immune system is crucial in addition to specific antiviral medications. In recent years, scientists have made an interesting finding that low dose IL-2 (ld-IL-2) could potentially have a crucial function in regulating the immune system and reducing the chances of infection, especially viral infection. Ld-IL-2 exerts immune regulatory effects in different types of viral infections by modulating CD4+ T subsets, CD8+ T cells, natural killer cells, and so on. Our review summarised the role of IL-2 or IL-2 complexes in viral infections. Ld-IL-2 may be an effective strategy for enhancing host antiviral immunity and preventing infection from becoming chronic; additionally, the appropriate use of it can help prevent excessive inflammatory response after infection. In the long term, it may reduce the occurrence of infection-related autoimmune diseases and tumours by promoting the restoration of early immune homeostasis. Furthermore, we have also summarised the application of ld-IL-2 in the context of autoimmune diseases combined with viral infections; it may be a safe and effective strategy for restoring immune homeostasis without compromising the antiviral immune response. In conclusion, focusing on the role of ld-IL-2 in viral infections may provide a new perspective for regulating immune responses following viral infections and improving prognosis.
Collapse
Affiliation(s)
- Rui Su
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Tingting Zhang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Hui Wang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Gaofei Yan
- Second department, Hamony Long Stomatological Hospital, Taiyuan, China
| | - Ruihe Wu
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Xin Zhang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Chong Gao
- Department of Pathology, Brigham and Women's Hospital/Children's Hospital Boston, Joint Program in Transfusion Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Xiaofeng Li
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Caihong Wang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| |
Collapse
|
13
|
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]
|
14
|
Paris R, Apter D, Boppana S, D’Aloia M, De Schrevel N, Delroisse JM, Grassano L, Guignard A, Panackal AA, Roman F, Yu J, Yunes EM, Dieussaert I. Incidence of Cytomegalovirus Primary and Secondary Infection in Adolescent Girls: Results From a Prospective Study. J Infect Dis 2023; 228:1491-1495. [PMID: 37340664 PMCID: PMC10681855 DOI: 10.1093/infdis/jiad182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Indexed: 06/22/2023] Open
Abstract
Developing a vaccine to prevent congenital cytomegalovirus (CMV) infection and newborn disability requires an understanding of infection incidence. In a prospective cohort study of 363 adolescent girls (NCT01691820), CMV serostatus, primary infection, and secondary infection were determined in blood and urine samples collected at enrollment and every 4 months for 3 years. Baseline CMV seroprevalence was 58%. Primary infection occurred in 14.8% of seronegative girls. Among seropositive girls, 5.9% had ≥4-fold increase in anti-CMV antibody, and 23.9% shed CMV DNA in urine. Our findings provide insights on infection epidemiology and highlight the need for more standardized markers of secondary infection.
Collapse
Affiliation(s)
| | - Dan Apter
- VL-Medi Clinical Research Center, Helsinki, Finland
| | - Suresh Boppana
- Departments of Pediatrics and Microbiology, University of Alabama at Birmingham, Birmingham, US
| | | | | | | | | | | | | | | | - Jing Yu
- Vaccines, GSK, Rockville, Maryland, US
| | - Elsa M Yunes
- Center for Research on Population Health, National Institute of Public Health, Cuernavaca, Mexico
| | | |
Collapse
|
15
|
Gourin C, Alain S, Hantz S. Anti-CMV therapy, what next? A systematic review. Front Microbiol 2023; 14:1321116. [PMID: 38053548 PMCID: PMC10694278 DOI: 10.3389/fmicb.2023.1321116] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 11/06/2023] [Indexed: 12/07/2023] Open
Abstract
Human cytomegalovirus (HCMV) is one of the main causes of serious complications in immunocompromised patients and after congenital infection. There are currently drugs available to treat HCMV infection, targeting viral polymerase, whose use is complicated by toxicity and the emergence of resistance. Maribavir and letermovir are the latest antivirals to have been developed with other targets. The approval of letermovir represents an important innovation for CMV prevention in hematopoietic stem cell transplant recipients, whereas maribavir allowed improving the management of refractory or resistant infections in transplant recipients. However, in case of multidrug resistance or for the prevention and treatment of congenital CMV infection, finding new antivirals or molecules able to inhibit CMV replication with the lowest toxicity remains a critical need. This review presents a range of molecules known to be effective against HCMV. Molecules with a direct action against HCMV include brincidofovir, cyclopropavir and anti-terminase benzimidazole analogs. Artemisinin derivatives, quercetin and baicalein, and anti-cyclooxygenase-2 are derived from natural molecules and are generally used for different indications. Although they have demonstrated indirect anti-CMV activity, few clinical studies were performed with these compounds. Immunomodulating molecules such as leflunomide and everolimus have also demonstrated indirect antiviral activity against HCMV and could be an interesting complement to antiviral therapy. The efficacy of anti-CMV immunoglobulins are discussed in CMV congenital infection and in association with direct antiviral therapy in heart transplanted patients. All molecules are described, with their mode of action against HCMV, preclinical tests, clinical studies and possible resistance. All these molecules have shown anti-HCMV potential as monotherapy or in combination with others. These new approaches could be interesting to validate in clinical trials.
Collapse
Affiliation(s)
- Claire Gourin
- INSERM, CHU Limoges, University of Limoges, RESINFIT, Limoges, France
| | - Sophie Alain
- INSERM, CHU Limoges, University of Limoges, RESINFIT, Limoges, France
- CHU Limoges, Laboratoire de Bactériologie-Virologie-Hygiène, National Reference Center for Herpesviruses, Limoges, France
| | - Sébastien Hantz
- INSERM, CHU Limoges, University of Limoges, RESINFIT, Limoges, France
- CHU Limoges, Laboratoire de Bactériologie-Virologie-Hygiène, National Reference Center for Herpesviruses, Limoges, France
| |
Collapse
|
16
|
Salomè S, Corrado FR, Mazzarelli LL, Maruotti GM, Capasso L, Blazquez-Gamero D, Raimondi F. Congenital cytomegalovirus infection: the state of the art and future perspectives. Front Pediatr 2023; 11:1276912. [PMID: 38034830 PMCID: PMC10687293 DOI: 10.3389/fped.2023.1276912] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 10/18/2023] [Indexed: 12/02/2023] Open
Abstract
Congenital cytomegalovirus (cCMV) infection is the most common congenital infection, with an estimated incidence of approximately one in 200 infants in high-income settings. Approximately one in four children may experience life-long consequences, including sensorineural hearing loss and neurodisability. Knowledge regarding prevention, diagnosis, and treatment increased in the recent years, but some challenges remain. In this review, we tried to summarize the current knowledge on both the obstetrical and pediatric areas, while also highlighting controversial aspects and future perspectives. There is a need to enhance awareness among the general population and pregnant women through specific information programs. Further research is needed to better define the classification of individuals at birth and to have a deeper understanding of the long-term outcomes for so defined children. Finally, the availability of valaciclovir medication throughout pregnancy, where appropriate, has prompted the assessment of a universal serological antenatal screening. It is recommended to establish a dedicated unit for better evaluation and management of both mothers and children.
Collapse
Affiliation(s)
- S. Salomè
- Division of Neonatology, Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - F. R. Corrado
- Division of Neonatology, Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - L. L. Mazzarelli
- Division of Obstetrician and Gynecologist, Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - G. M. Maruotti
- Division of Obstetrician and Gynecologist, Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - L. Capasso
- Division of Neonatology, Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - D. Blazquez-Gamero
- Pediatric Infectious Diseases Unit, Hospital Universitario 12 de Octubre, Instituto de Investigación Hospital 12 de Octubre (Imas12), Translational Research Network in Pediatric Infectious Diseases (RITIP), Universidad Complutense, Madrid, Spain
| | - F. Raimondi
- Division of Neonatology, Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| |
Collapse
|
17
|
Boppana SB, van Boven M, Britt WJ, Gantt S, Griffiths PD, Grosse SD, Hyde TB, Lanzieri TM, Mussi-Pinhata MM, Pallas SE, Pinninti SG, Rawlinson WD, Ross SA, Vossen ACTM, Fowler KB. Vaccine value profile for cytomegalovirus. Vaccine 2023; 41 Suppl 2:S53-S75. [PMID: 37806805 DOI: 10.1016/j.vaccine.2023.06.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 04/28/2023] [Accepted: 06/02/2023] [Indexed: 10/10/2023]
Abstract
Cytomegalovirus (CMV) is the most common infectious cause of congenital malformation and a leading cause of developmental disabilities such as sensorineural hearing loss (SNHL), motor and cognitive deficits. The significant disease burden from congenital CMV infection (cCMV) led the US National Institute of Medicine to rank CMV vaccine development as the highest priority. An average of 6.7/1000 live births are affected by cCMV, but the prevalence varies across and within countries. In contrast to other congenital infections such as rubella and toxoplasmosis, the prevalence of cCMV increases with CMV seroprevalence rates in the population. The true global burden of cCMV disease is likely underestimated because most infected infants (85-90 %) have asymptomatic infection and are not identified. However, about 7-11 % of those with asymptomatic infection will develop SNHL throughout early childhood. Although no licensed CMV vaccine exists, several candidate vaccines are in development, including one currently in phase 3 trials. Licensure of one or more vaccine candidates is feasible within the next five years. Various models of CMV vaccine strategies employing different target populations have shown to provide substantial benefit in reducing cCMV. Although CMV can cause end-organ disease with significant morbidity and mortality in immunocompromised individuals, the focus of this vaccine value profile (VVP) is on preventing or reducing the cCMV disease burden. This CMV VVP provides a high-level, comprehensive assessment of the currently available data to inform the potential public health, economic, and societal value of CMV vaccines. The CMV VVP was developed by a working group of subject matter experts from academia, public health groups, policy organizations, and non-profit organizations. All contributors have extensive expertise on various elements of the CMV VVP and have described the state of knowledge and identified the current gaps. The VVP was developed using only existing and publicly available information.
Collapse
Affiliation(s)
- Suresh B Boppana
- Departments of Pediatrics and Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Michiel van Boven
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, and Julius Center for Health Sciences and Primary Care, Department of Epidemiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - William J Britt
- Departments of Pediatrics, Microbiology, and Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, USA
| | - Soren Gantt
- Centre de recherche du CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada
| | - Paul D Griffiths
- Emeritus Professor of Virology, University College London, United Kingdom
| | - Scott D Grosse
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, GA 30341, USA
| | - Terri B Hyde
- Global Immunization Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Tatiana M Lanzieri
- Measles, Rubella, and Cytomegalovirus Epidemiology Team, Viral Vaccine Preventable Diseases Branch / Division of Viral Diseases. National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Marisa M Mussi-Pinhata
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Brazil
| | - Sarah E Pallas
- Global Immunization Division, Center for Global Health, U.S. Centers for Disease Control and Prevention, Atlanta, GA 30329-4027, USA
| | - Swetha G Pinninti
- Departments of Pediatrics and Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - William D Rawlinson
- Serology and Virology Division, NSW Health Pathology Randwick, Prince of Wales Hospital, Sydney, Australia; School of Biotechnology and Biomolecular Sciences, and School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Shannon A Ross
- Departments of Pediatrics and Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ann C T M Vossen
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Karen B Fowler
- Departments of Pediatrics and Epidemiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
18
|
Barry PA, Iyer SS, Gibson L. Re-Evaluating Human Cytomegalovirus Vaccine Design: Prediction of T Cell Epitopes. Vaccines (Basel) 2023; 11:1629. [PMID: 38005961 PMCID: PMC10674879 DOI: 10.3390/vaccines11111629] [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: 08/30/2023] [Revised: 10/05/2023] [Accepted: 10/09/2023] [Indexed: 11/26/2023] Open
Abstract
HCMV vaccine development has traditionally focused on viral antigens identified as key targets of neutralizing antibody (NAb) and/or T cell responses in healthy adults with chronic HCMV infection, such as glycoprotein B (gB), the glycoprotein H-anchored pentamer complex (PC), and the unique long 83 (UL83)-encoded phosphoprotein 65 (pp65). However, the protracted absence of a licensed HCMV vaccine that reduces the risk of infection in pregnancy regardless of serostatus warrants a systematic reassessment of assumptions informing vaccine design. To illustrate this imperative, we considered the hypothesis that HCMV proteins infrequently detected as targets of T cell responses may contain important vaccine antigens. Using an extant dataset from a T cell profiling study, we tested whether HCMV proteins recognized by only a small minority of participants encompass any T cell epitopes. Our analyses demonstrate a prominent skewing of T cell responses away from most viral proteins-although they contain robust predicted CD8 T cell epitopes-in favor of a more restricted set of proteins. Our findings raise the possibility that HCMV may benefit from evading the T cell recognition of certain key proteins and that, contrary to current vaccine design approaches, including them as vaccine antigens could effectively take advantage of this vulnerability.
Collapse
Affiliation(s)
- Peter A. Barry
- Department of Pathology and Laboratory Medicine, Center for Immunology and Infectious Diseases, University of California Davis School of Medicine, Sacramento, CA 95817, USA;
- California National Primate Research Center, University of California, Davis, CA 95616, USA
| | - Smita S. Iyer
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15261, USA;
| | - Laura Gibson
- Departments of Medicine and of Pediatrics, Infectious Diseases and Immunology, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| |
Collapse
|
19
|
Moström MJ, Yu S, Tran D, Saccoccio FM, Versoza CJ, Malouli D, Mirza A, Valencia S, Gilbert M, Blair RV, Hansen S, Barry P, Früh K, Jensen JD, Pfeifer SP, Kowalik TF, Permar SR, Kaur A. Protective effect of pre-existing natural immunity in a nonhuman primate reinfection model of congenital cytomegalovirus infection. PLoS Pathog 2023; 19:e1011646. [PMID: 37796819 PMCID: PMC10553354 DOI: 10.1371/journal.ppat.1011646] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 08/29/2023] [Indexed: 10/07/2023] Open
Abstract
Congenital cytomegalovirus (cCMV) is the leading infectious cause of neurologic defects in newborns with particularly severe sequelae in the setting of primary CMV infection in the first trimester of pregnancy. The majority of cCMV cases worldwide occur after non-primary infection in CMV-seropositive women; yet the extent to which pre-existing natural CMV-specific immunity protects against CMV reinfection or reactivation during pregnancy remains ill-defined. We previously reported on a novel nonhuman primate model of cCMV in rhesus macaques where 100% placental transmission and 83% fetal loss were seen in CD4+ T lymphocyte-depleted rhesus CMV (RhCMV)-seronegative dams after primary RhCMV infection. To investigate the protective effect of preconception maternal immunity, we performed reinfection studies in CD4+ T lymphocyte-depleted RhCMV-seropositive dams inoculated in late first / early second trimester gestation with RhCMV strains 180.92 (n = 2), or RhCMV UCD52 and FL-RhCMVΔRh13.1/SIVgag, a wild-type-like RhCMV clone with SIVgag inserted as an immunological marker, administered separately (n = 3). An early transient increase in circulating monocytes followed by boosting of the pre-existing RhCMV-specific CD8+ T lymphocyte and antibody response was observed in the reinfected dams but not in control CD4+ T lymphocyte-depleted dams. Emergence of SIV Gag-specific CD8+ T lymphocyte responses in macaques inoculated with the FL-RhCMVΔRh13.1/SIVgag virus confirmed reinfection. Placental transmission was detected in only one of five reinfected dams and there were no adverse fetal sequelae. Viral whole genome, short-read, deep sequencing analysis confirmed transmission of both reinfection RhCMV strains across the placenta with ~30% corresponding to FL-RhCMVΔRh13.1/SIVgag and ~70% to RhCMV UCD52, consistent with the mixed human CMV infections reported in infants with cCMV. Our data showing reduced placental transmission and absence of fetal loss after non-primary as opposed to primary infection in CD4+ T lymphocyte-depleted dams indicates that preconception maternal CMV-specific CD8+ T lymphocyte and/or humoral immunity can protect against cCMV infection.
Collapse
Affiliation(s)
- Matilda J. Moström
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, United States of America
| | - Shan Yu
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, United States of America
| | - Dollnovan Tran
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, United States of America
| | - Frances M. Saccoccio
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, United States of America
| | - Cyril J. Versoza
- Center for Evolution & Medicine, School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Daniel Malouli
- Oregon Health and Sciences University, Beaverton, Oregon, United States of America
| | - Anne Mirza
- University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Sarah Valencia
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, United States of America
| | - Margaret Gilbert
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, United States of America
| | - Robert V. Blair
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, United States of America
| | - Scott Hansen
- Oregon Health and Sciences University, Beaverton, Oregon, United States of America
| | - Peter Barry
- University of California, Davis, California, United States of America
| | - Klaus Früh
- Oregon Health and Sciences University, Beaverton, Oregon, United States of America
| | - Jeffrey D. Jensen
- Center for Evolution & Medicine, School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Susanne P. Pfeifer
- Center for Evolution & Medicine, School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Timothy F. Kowalik
- University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Sallie R. Permar
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, United States of America
- Weill Cornell Medicine, New York, New York State, United States of America
| | - Amitinder Kaur
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, United States of America
| |
Collapse
|
20
|
Bottino P, Pastrone L, Curtoni A, Bondi A, Sidoti F, Zanotto E, Cavallo R, Solidoro P, Costa C. Antiviral Approach to Cytomegalovirus Infection: An Overview of Conventional and Novel Strategies. Microorganisms 2023; 11:2372. [PMID: 37894030 PMCID: PMC10608897 DOI: 10.3390/microorganisms11102372] [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: 08/28/2023] [Revised: 09/16/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
Human cytomegalovirus (HCMV) is a herpesvirus capable of establishing a lifelong persistence in the host through a chronic state of infection and remains an essential global concern due to its distinct life cycle, mutations, and latency. It represents a life-threatening pathogen for immunocompromised patients, such as solid organ transplanted patients, HIV-positive individuals, and hematopoietic stem cell recipients. Multiple antiviral approaches are currently available and administered in order to prevent or manage viral infections in the early stages. However, limitations due to side effects and the onset of antidrug resistance are a hurdle to their efficacy, especially for long-term therapies. Novel antiviral molecules, together with innovative approaches (e.g., genetic editing and RNA interference) are currently in study, with promising results performed in vitro and in vivo. Since HCMV is a virus able to establish latent infection, with a consequential risk of reactivation, infection management could benefit from preventive treatment for critical patients, such as immunocompromised individuals and seronegative pregnant women. This review will provide an overview of conventional antiviral clinical approaches and their mechanisms of action. Additionally, an overview of proposed and developing new molecules is provided, including nucleic-acid-based therapies and immune-mediated approaches.
Collapse
Affiliation(s)
- Paolo Bottino
- Microbiology and Virology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy; (L.P.); (A.C.); (A.B.); (F.S.); (E.Z.); (R.C.)
| | - Lisa Pastrone
- Microbiology and Virology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy; (L.P.); (A.C.); (A.B.); (F.S.); (E.Z.); (R.C.)
| | - Antonio Curtoni
- Microbiology and Virology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy; (L.P.); (A.C.); (A.B.); (F.S.); (E.Z.); (R.C.)
| | - Alessandro Bondi
- Microbiology and Virology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy; (L.P.); (A.C.); (A.B.); (F.S.); (E.Z.); (R.C.)
| | - Francesca Sidoti
- Microbiology and Virology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy; (L.P.); (A.C.); (A.B.); (F.S.); (E.Z.); (R.C.)
| | - Elisa Zanotto
- Microbiology and Virology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy; (L.P.); (A.C.); (A.B.); (F.S.); (E.Z.); (R.C.)
| | - Rossana Cavallo
- Microbiology and Virology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy; (L.P.); (A.C.); (A.B.); (F.S.); (E.Z.); (R.C.)
| | - Paolo Solidoro
- Pneumology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy;
| | - Cristina Costa
- Microbiology and Virology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy; (L.P.); (A.C.); (A.B.); (F.S.); (E.Z.); (R.C.)
| |
Collapse
|
21
|
Mendilcioglu I, Dogan NU, Ozkan O, Bahceci M, Boynukalin K, Dogan S, Ozkan O. Pregnancy management and outcome after uterus transplantation. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2023; 62:328-335. [PMID: 36468688 DOI: 10.1002/uog.26134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 10/30/2022] [Accepted: 11/18/2022] [Indexed: 06/17/2023]
Abstract
Uterus transplantation is a novel approach in women whose uterus is absent or severely abnormal. However, it is still an experimental procedure that poses risks to both mother and baby. To date, 32 live births after uterus transplantation have been reported in peer-reviewed journals, with several maternal, fetal and neonatal complications. The most common complications were preterm delivery, hypertensive disorders and placenta previa. Four patients experienced episodes of transplant rejection during pregnancy. The appropriate management of complicated and non-complicated pregnancies following uterus transplantation is still unresolved. In this review, obstetric outcomes after uterus transplantation and optimal management during pregnancy are discussed in light of the available data. © 2022 International Society of Ultrasound in Obstetrics and Gynecology.
Collapse
Affiliation(s)
- I Mendilcioglu
- Department of Obstetrics and Gynecology, Akdeniz University, Antalya, Turkey
| | - N U Dogan
- Department of Obstetrics and Gynecology, Akdeniz University, Antalya, Turkey
| | - O Ozkan
- Department of Plastic and Reconstructive Surgery, Akdeniz University, Antalya, Turkey
| | - M Bahceci
- Bahçeci IVF Center, Istanbul, Turkey
| | | | - S Dogan
- Department of Obstetrics and Gynecology, Akdeniz University, Antalya, Turkey
| | - O Ozkan
- Department of Plastic and Reconstructive Surgery, Akdeniz University, Antalya, Turkey
| |
Collapse
|
22
|
Hill M, Mostafa S, Muganda PM, Jeffers-Francis LK, Obeng-Gyasi E. The Association of Cytomegalovirus and Allostatic Load by Country of Birth and Length of Time in the United States. Diseases 2023; 11:101. [PMID: 37606472 PMCID: PMC10443278 DOI: 10.3390/diseases11030101] [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: 07/17/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND Cytomegalovirus (CMV) is a highly prevalent virus with a worldwide distribution. It typically remains dormant in most individuals until reactivation. Immunocompromised states are known to be potential causes for CMV reactivation. Current research has shown a link in the decline of immigrant health among those living in the US for an extended period, though the impact of CMV on this is not clear. METHODS This study investigated the association between country of birth, duration of US residency, allostatic load, and latent cytomegalovirus infection (CMV IgG) in a sample of US adults aged 20-49. The data utilized for our analysis was obtained from the National Health and Nutrition Examination Survey (NHANES) conducted between 2001 and 2004. Allostatic load, an index measuring the cumulative physiological strain on the body as it strives to regain stability in the presence of chronic stress, provided a valuable approach to assess stress within the context of CMV exposure. Logistic regression modeling was employed to estimate odds ratios and confidence intervals for the analysis. The chi-square test of association and Cramer's V statistic were used to assess the correlation among categorical variables, while Pearson's correlation coefficient was applied to evaluate the relationship between continuous variables. The results revealed that individuals born outside the US and those with less than 20 years of residency in the US exhibited significantly higher proportions of positive CMV IgG compared to individuals born in the US. Specifically, individuals born outside the US had more than triple the odds of CMV IgG when adjusting for the AL index (OR = 3.69, p-value = 0.0063). A similar trend was observed when examining AL risk based on the duration of US residency. Furthermore, age and sex were identified as significant predictors (p-value < 0.05) of AL risk, considering the individual's country of birth. In summary, the findings of this study significantly enhance our comprehension of the intricate interplay between cytomegalovirus (CMV) and allostatic load (AL). The investigation sheds light on how CMV and AL interact within specific demographic contexts, providing valuable insights into the underlying risk factors for CMV infection.
Collapse
Affiliation(s)
- Matthew Hill
- Department of Built Environment, North Carolina A&T State University, Greensboro, NC 27411, USA
- Environmental Health and Disease Laboratory, North Carolina A&T State University, Greensboro, NC 27411, USA
| | - Sayed Mostafa
- Department of Mathematics and Statistics, North Carolina A&T State University, Greensboro, NC 27411, USA
| | - Perpetua M. Muganda
- Department of Biology, North Carolina A&T State University, Greensboro, NC 27411, USA
| | | | - Emmanuel Obeng-Gyasi
- Department of Built Environment, North Carolina A&T State University, Greensboro, NC 27411, USA
- Environmental Health and Disease Laboratory, North Carolina A&T State University, Greensboro, NC 27411, USA
| |
Collapse
|
23
|
Farias-Jofre M, Romero R, Xu Y, Levenson D, Tao L, Kanninen T, Galaz J, Arenas-Hernandez M, Liu Z, Miller D, Bhatti G, Seyerle M, Tarca AL, Gomez-Lopez N. Differential immunophenotype of circulating monocytes from pregnant women in response to viral ligands. BMC Pregnancy Childbirth 2023; 23:323. [PMID: 37149573 PMCID: PMC10163583 DOI: 10.1186/s12884-023-05562-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/30/2023] [Indexed: 05/08/2023] Open
Abstract
BACKGROUND Viral infections during pregnancy can have deleterious effects on mothers and their offspring. Monocytes participate in the maternal host defense against invading viruses; however, whether pregnancy alters monocyte responses is still under investigation. Herein, we undertook a comprehensive in vitro study of peripheral monocytes to characterize the differences in phenotype and interferon release driven by viral ligands between pregnant and non-pregnant women. METHODS Peripheral blood was collected from third-trimester pregnant (n = 20) or non-pregnant (n = 20, controls) women. Peripheral blood mononuclear cells were isolated and exposed to R848 (TLR7/TLR8 agonist), Gardiquimod (TLR7 agonist), Poly(I:C) (HMW) VacciGrade™ (TLR3 agonist), Poly(I:C) (HMW) LyoVec™ (RIG-I/MDA-5 agonist), or ODN2216 (TLR9 agonist) for 24 h. Cells and supernatants were collected for monocyte phenotyping and immunoassays to detect specific interferons, respectively. RESULTS The proportions of classical (CD14hiCD16-), intermediate (CD14hiCD16+), non-classical (CD14loCD16+), and CD14loCD16- monocytes were differentially affected between pregnant and non-pregnant women in response to TLR3 stimulation. The proportions of pregnancy-derived monocytes expressing adhesion molecules (Basigin and PSGL-1) or the chemokine receptors CCR5 and CCR2 were diminished in response to TLR7/TLR8 stimulation, while the proportions of CCR5- monocytes were increased. Such differences were found to be primarily driven by TLR8 signaling, rather than TLR7. Moreover, the proportions of monocytes expressing the chemokine receptor CXCR1 were increased during pregnancy in response to poly(I:C) stimulation through TLR3, but not RIG-I/MDA-5. By contrast, pregnancy-specific changes in the monocyte response to TLR9 stimulation were not observed. Notably, the soluble interferon response to viral stimulation by mononuclear cells was not diminished in pregnancy. CONCLUSIONS Our data provide insight into the differential responsiveness of pregnancy-derived monocytes to ssRNA and dsRNA, mainly driven by TLR8 and membrane-bound TLR3, which may help to explain the increased susceptibility of pregnant women to adverse outcomes resulting from viral infection as observed during recent and historic pandemics.
Collapse
Affiliation(s)
- Marcelo Farias-Jofre
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
- Division of Obstetrics and Gynecology, Faculty of Medicine, Pontificia Universidad Católica de Chile, 8330024, Santiago, Chile
| | - Roberto Romero
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, 48201, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, 48824, USA
| | - Yi Xu
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
| | - Dustyn Levenson
- Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Li Tao
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
| | - Tomi Kanninen
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
| | - Jose Galaz
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
- Division of Obstetrics and Gynecology, Faculty of Medicine, Pontificia Universidad Católica de Chile, 8330024, Santiago, Chile
| | - Marcia Arenas-Hernandez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
| | - Zhenjie Liu
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
| | - Derek Miller
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
| | - Gaurav Bhatti
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
| | - Megan Seyerle
- Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Adi L Tarca
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA
- Department of Computer Science, Wayne State University College of Engineering, Detroit, MI, 48202, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, 48201, USA
| | - Nardhy Gomez-Lopez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS), Detroit, MI, 48201, USA.
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, 48201, USA.
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, 48201, USA.
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI, 48201, USA.
| |
Collapse
|
24
|
Greye H, Wex T, Taneva E, Redlich A, Costa SD, Rissmann A. Cytomegalovirus seronegativity rate in pregnant women and primary cytomegalovirus infection during pregnancy in rural Germany. BMC Pregnancy Childbirth 2023; 23:299. [PMID: 37118680 PMCID: PMC10148470 DOI: 10.1186/s12884-023-05612-7] [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: 12/23/2022] [Accepted: 04/14/2023] [Indexed: 04/30/2023] Open
Abstract
BACKGROUND Congenital cytomegalovirus (CMV) infection is the most common congenital infection worldwide and one of the leading causes of congenital hearing loss in newborns. The aim of this study was to determine the seroprevalence rate for cytomegalovirus in pregnant women and the rate of CMV serological testing utilised during pregnancy in a rural region in Germany. METHODS Retrospective data on the prevalence of CMV IgG and IgM antibodies were obtained from 3,800 women, identified in the study group of 19,511 pregnant women from outpatient settings whose samples were collected between 1 and 2014 and 30 April 2018. In addition, the serological CMV status in regards to various billing methods was further analyzed. RESULTS Serological CMV tests were performed in 3,800 (19.5%) out of 19,511 pregnant women. 2,081 (54.8%) of these women were CMV seronegative. Among those, seroconversion rate of 0.37-1.42% was identified. A proportion of 2,710 (14.7%) of all 18,460 women with statutory health insurance made use of the CMV testing as an individual health service. CONCLUSIONS The low uptake of CMV serological testing in the study population covered indicates low risk awareness among pregnant women and their healthcare professionals. Presented seronegativity rates and routine seroconversion rate, demonstrate importance to improve intervention strategy to prevent feto-maternal CMV transmission.
Collapse
Affiliation(s)
- Hannah Greye
- Malformation Monitoring Centre Saxony-Anhalt, Medical Faculty Otto-von-Guericke-University, Leipziger Straße 44, D-39120, Magdeburg, Germany
| | - Thomas Wex
- Medical Laboratory for Clinical Chemistry, Microbiology, Infectious Diseases and Genetics "Prof. Schenk/Dr. Ansorge & Colleagues", Schwiesaustraße 11, D-39124, Magdeburg, Germany
| | - Elina Taneva
- Medical Laboratory for Clinical Chemistry, Microbiology, Infectious Diseases and Genetics "Prof. Schenk/Dr. Ansorge & Colleagues", Schwiesaustraße 11, D-39124, Magdeburg, Germany
| | - Anke Redlich
- Department of Obstetrics and Gynaecology, University Hospital Magdeburg, Gerhart-Hauptmann-Strasse 35, D-39108, Magdeburg, Germany
| | - Serban-Dan Costa
- Department of Obstetrics and Gynaecology, University Hospital Magdeburg, Gerhart-Hauptmann-Strasse 35, D-39108, Magdeburg, Germany
| | - Anke Rissmann
- Malformation Monitoring Centre Saxony-Anhalt, Medical Faculty Otto-von-Guericke-University, Leipziger Straße 44, D-39120, Magdeburg, Germany.
| |
Collapse
|
25
|
Moström M, Yu S, Tran D, Saccoccio F, Versoza CJ, Malouli D, Mirza A, Valencia S, Gilbert M, Blair R, Hansen S, Barry P, Früh K, Jensen JD, Pfeifer SP, Kowalik TF, Permar SR, Kaur A. Protective effect of pre-existing natural immunity in a nonhuman primate reinfection model of congenital cytomegalovirus infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.10.536057. [PMID: 37090643 PMCID: PMC10120644 DOI: 10.1101/2023.04.10.536057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Congenital cytomegalovirus (cCMV) is the leading infectious cause of neurologic defects in newborns with particularly severe sequelae in the setting of primary CMV infection in the first trimester of pregnancy. The majority of cCMV cases worldwide occur after non-primary infection in CMV-seropositive women; yet the extent to which pre-existing natural CMV-specific immunity protects against CMV reinfection or reactivation during pregnancy remains ill-defined. We previously reported on a novel nonhuman primate model of cCMV in rhesus macaques where 100% placental transmission and 83% fetal loss were seen in CD4 + T lymphocyte-depleted rhesus CMV (RhCMV)-seronegative dams after primary RhCMV infection. To investigate the protective effect of preconception maternal immunity, we performed reinfection studies in CD4+ T lymphocyte-depleted RhCMV-seropositive dams inoculated in late first / early second trimester gestation with RhCMV strains 180.92 ( n =2), or RhCMV UCD52 and FL-RhCMVΔRh13.1/SIV gag , a wild-type-like RhCMV clone with SIV gag inserted as an immunological marker ( n =3). An early transient increase in circulating monocytes followed by boosting of the pre-existing RhCMV-specific CD8+ T lymphocyte and antibody response was observed in the reinfected dams but not in control CD4+ T lymphocyte-depleted dams. Emergence of SIV Gag-specific CD8+ T lymphocyte responses in macaques inoculated with the FL-RhCMVΔRh13.1/SIV gag virus confirmed reinfection. Placental transmission was detected in only one of five reinfected dams and there were no adverse fetal sequelae. Viral whole genome, short-read, deep sequencing analysis confirmed transmission of both reinfection RhCMV strains across the placenta with ∼30% corresponding to FL-RhCMVΔRh13.1/SIV gag and ∼70% to RhCMV UCD52, consistent with the mixed human CMV infections reported in infants with cCMV. Our data showing reduced placental transmission and absence of fetal loss after non-primary as opposed to primary infection in CD4+ T lymphocyte-depleted dams indicates that preconception maternal CMV-specific CD8+ T lymphocyte and/or humoral immunity can protect against cCMV infection. Author Summary Globally, pregnancies in CMV-seropositive women account for the majority of cases of congenital CMV infection but the immune responses needed for protection against placental transmission in mothers with non-primary infection remains unknown. Recently, we developed a nonhuman primate model of primary rhesus CMV (RhCMV) infection in which placental transmission and fetal loss occurred in RhCMV-seronegative CD4+ T lymphocyte-depleted macaques. By conducting similar studies in RhCMV-seropositive dams, we demonstrated the protective effect of pre-existing natural CMV-specific CD8+ T lymphocytes and humoral immunity against congenital CMV after reinfection. A 5-fold reduction in congenital transmission and complete protection against fetal loss was observed in dams with pre-existing immunity compared to primary CMV in this model. Our study is the first formal demonstration in a relevant model of human congenital CMV that natural pre-existing CMV-specific maternal immunity can limit congenital CMV transmission and its sequelae. The nonhuman primate model of non-primary congenital CMV will be especially relevant to studying immune requirements of a maternal vaccine for women in high CMV seroprevalence areas at risk of repeated CMV reinfections during pregnancy.
Collapse
Affiliation(s)
- Matilda Moström
- Tulane National Primate Research Center, Tulane University, Covington LA
| | - Shan Yu
- Tulane National Primate Research Center, Tulane University, Covington LA
| | - Dollnovan Tran
- Tulane National Primate Research Center, Tulane University, Covington LA
| | | | - Cyril J. Versoza
- Center for Evolution & Medicine, School of Life Sciences, Arizona State University, Tempe, AZ
| | | | - Anne Mirza
- University of Massachusetts Chan Medical School, Worcester, MA
| | - Sarah Valencia
- Duke Human Vaccine Institute, Duke University, Durham, NC
| | - Margaret Gilbert
- Tulane National Primate Research Center, Tulane University, Covington LA
| | - Robert Blair
- Tulane National Primate Research Center, Tulane University, Covington LA
| | - Scott Hansen
- Oregon Health and Sciences University, Beaverton, OR
| | | | - Klaus Früh
- Oregon Health and Sciences University, Beaverton, OR
| | - Jeffrey D. Jensen
- Center for Evolution & Medicine, School of Life Sciences, Arizona State University, Tempe, AZ
| | - Susanne P. Pfeifer
- Center for Evolution & Medicine, School of Life Sciences, Arizona State University, Tempe, AZ
| | | | - Sallie R. Permar
- Duke Human Vaccine Institute, Duke University, Durham, NC
- Weill Cornell Medicine, New York, NY
| | - Amitinder Kaur
- Tulane National Primate Research Center, Tulane University, Covington LA
| |
Collapse
|
26
|
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
|
27
|
Raymenants J, Geenen C, Budts L, Thibaut J, Thijssen M, De Mulder H, Gorissen S, Craessaerts B, Laenen L, Beuselinck K, Ombelet S, Keyaerts E, André E. Indoor air surveillance and factors associated with respiratory pathogen detection in community settings in Belgium. Nat Commun 2023; 14:1332. [PMID: 36898982 PMCID: PMC10005919 DOI: 10.1038/s41467-023-36986-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 02/27/2023] [Indexed: 03/12/2023] Open
Abstract
Currently, the real-life impact of indoor climate, human behaviour, ventilation and air filtration on respiratory pathogen detection and concentration are poorly understood. This hinders the interpretability of bioaerosol quantification in indoor air to surveil respiratory pathogens and transmission risk. We tested 341 indoor air samples from 21 community settings in Belgium for 29 respiratory pathogens using qPCR. On average, 3.9 pathogens were positive per sample and 85.3% of samples tested positive for at least one. Pathogen detection and concentration varied significantly by pathogen, month, and age group in generalised linear (mixed) models and generalised estimating equations. High CO2 and low natural ventilation were independent risk factors for detection. The odds ratio for detection was 1.09 (95% CI 1.03-1.15) per 100 parts per million (ppm) increase in CO2, and 0.88 (95% CI 0.80-0.97) per stepwise increase in natural ventilation (on a Likert scale). CO2 concentration and portable air filtration were independently associated with pathogen concentration. Each 100ppm increase in CO2 was associated with a qPCR Ct value decrease of 0.08 (95% CI -0.12 to -0.04), and portable air filtration with a 0.58 (95% CI 0.25-0.91) increase. The effects of occupancy, sampling duration, mask wearing, vocalisation, temperature, humidity and mechanical ventilation were not significant. Our results support the importance of ventilation and air filtration to reduce transmission.
Collapse
Affiliation(s)
- Joren Raymenants
- Laboratory of Clinical Microbiology, KU Leuven, Herestraat 49, 3000, Leuven, Belgium.
- Department of General Internal Medicine, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium.
| | - Caspar Geenen
- Laboratory of Clinical Microbiology, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Lore Budts
- Department of Laboratory Medicine, National Reference Center of Respiratory Pathogens, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Jonathan Thibaut
- Laboratory of Clinical Microbiology, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Marijn Thijssen
- Laboratory of Clinical and Epidemiological Virology (Rega Institute), KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Hannelore De Mulder
- Laboratory of Clinical Microbiology, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Sarah Gorissen
- Laboratory of Clinical Microbiology, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Bastiaan Craessaerts
- Department of Laboratory Medicine, National Reference Center of Respiratory Pathogens, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Lies Laenen
- Laboratory of Clinical Microbiology, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
- Department of Laboratory Medicine, National Reference Center of Respiratory Pathogens, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Kurt Beuselinck
- Department of Laboratory Medicine, National Reference Center of Respiratory Pathogens, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Sien Ombelet
- Department of Laboratory Medicine, National Reference Center of Respiratory Pathogens, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Els Keyaerts
- Department of Laboratory Medicine, National Reference Center of Respiratory Pathogens, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Emmanuel André
- Laboratory of Clinical Microbiology, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
- Department of Laboratory Medicine, National Reference Center of Respiratory Pathogens, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| |
Collapse
|
28
|
To EE. Cell and Tissue Specific Metabolism of Nucleoside and Nucleotide Drugs: Case Studies and Implications for Precision Medicine. Drug Metab Dispos 2023; 51:360-368. [PMID: 36446610 DOI: 10.1124/dmd.122.000856] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 10/31/2022] [Accepted: 11/16/2022] [Indexed: 12/05/2022] Open
Abstract
Many clinically used antiviral drugs are nucleoside or nucleotide analog drugs, which have a unique mechanism of action that requires intracellular phosphorylation. This dependence on intracellular activation presents novel challenges for the discovery and development of nucleoside/nucleotide analog drugs. Contrary to many small molecule drug development programs that rely on plasma pharmacokinetics and systemic exposures, the precise mechanisms that result in efficacious intracellular nucleoside triphosphate concentrations must be understood in the process of nucleoside/nucleotide drug development. The importance is highlighted here, using the following as case studies: the herpes treatment acyclovir, the cytomegalovirus therapy ganciclovir, and human immunodeficiency virus (HIV) treatments based on tenofovir, which are also in use for HIV prophylaxis. For each drug, the specificity of metabolism that results in its activation in different cells or tissues is discussed, and the implications explored. Acyclovir's dependence on a viral enzyme for activation provides selective pressure for resistance mutations. Ganciclovir is also dependent on a viral enzyme for activation, and suicide gene therapy capitalizes on that for a novel oncology treatment. The tissue of most relevance for tenofovir activation depends on its use as treatment or as prophylaxis, and the pharmacogenomics and drug-drug interactions in those tissues must be considered. Finally, differential metabolism of different tenofovir prodrugs and its effects on toxicity risk are explored. Taken together, these examples highlight the importance of understanding tissue specific metabolism for optimal use of nucleoside/nucleotide drugs in the clinic. SIGNIFICANCE STATEMENT: Nucleoside and nucleotide analogue drugs are cornerstones in current antiviral therapy and prevention efforts that require intracellular phosphorylation for activity. Understanding their cell and tissue specific metabolism enables their rational, precision use for maximum efficacy.
Collapse
Affiliation(s)
- Elaine E To
- Gilead Sciences, Inc., Foster City, California, USA
| |
Collapse
|
29
|
Prevalence of congenital cytomegalovirus infection according to the type of maternal infection in Japan. J Infect Chemother 2023; 29:485-489. [PMID: 36716861 DOI: 10.1016/j.jiac.2023.01.020] [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: 08/23/2022] [Revised: 01/25/2023] [Accepted: 01/26/2023] [Indexed: 01/29/2023]
Abstract
INTRODUCTION Variable rates of cytomegalovirus (CMV) seropositivity in mothers from different individual's background may translate to distinct epidemiological patterns of congenital CMV infection. METHODS The prospective cohort study was conducted in Japan to evaluate the prevalence of vertical transmission rate according to the type of maternal infection. Post hoc power as a follow-up analysis was evaluated to compare the statistical power with other studies from France, Finland and Brazil. One thousand one hundred sixty-three pregnant women were measured IgG, IgM and IgG avidity index. The urine samples of neonates of these women were evaluated using polymerase chain reaction to diagnose the vertical transmission. RESULTS The prevalence of congenital CMV infection in the study population was 0.4%. The proportions of patients with primary and nonprimary infections were 60% and 40%, respectively, with a maternal seroprevalence of 82.5%. The rate of vertical transmission among the seronegative pregnant women before pregnancy was statistically higher than that among the seropositive pregnant women before pregnancy (p < 0.05), with a study power of 52.7%. The same difference was observed in France and Finland for maternal seroprevalence of 61% and 72% and statistical power of 56.9% and 66.7%, respectively. CONCLUSION The maternal seroprevalence of the present study conducted in Japan was much higher than that of studies in France and Finland. Nevertheless, seronegative pregnant women had a higher risk of vertical transmission before pregnancy.
Collapse
|
30
|
Targeted Protein-Specific Multi-Epitope-Based Vaccine Designing against Human Cytomegalovirus by Using Immunoinformatics Approaches. Vaccines (Basel) 2023; 11:vaccines11020203. [PMID: 36851082 PMCID: PMC9959080 DOI: 10.3390/vaccines11020203] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/10/2023] [Accepted: 01/11/2023] [Indexed: 01/19/2023] Open
Abstract
Cytomegaloviruses are emerging pathogenic agents known to cause congenital disorders in humans. In this study, immune epitopes (CTL, B cell and HTL) were screened for highly antigenic target proteins of the Human Cytomegalovirus. These shortlisted epitopes were then joined together through suitable linkers to construct multi epitope-based vaccine constructs (MEVCs). The functionality of each vaccine construct was evaluated through tertiary vaccine structure modelling and validations. Furthermore, physio-chemical properties including allergenicity, antigenicity molecular weight and many others were also predicted. The vaccine designs were also docked with the human TLR-4 receptor to demonstrate the receptor specific affinity and formed interactions. The vaccine peptides sequences were also subjected to codon optimization to confirm the potential vaccines expression in E. coli hosts. Additionally, all the MEVCs were also evaluated for immune response (IgG and IgM) induction. However, further in vivo tests are needed to ensure the efficacy of these vaccine designs.
Collapse
|
31
|
Aoki H, Bitnun A, Kitano T. The cost-effectiveness of maternal and neonatal screening for congenital cytomegalovirus infection in Japan. J Med Virol 2023; 95:e28391. [PMID: 36484373 DOI: 10.1002/jmv.28391] [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: 08/08/2022] [Revised: 11/08/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022]
Abstract
Congenital cytomegalovirus infection is the most common congenital infection. Using a decision tree model, cost-effectiveness of maternal screening with subsequent prenatal valacyclovir treatment and newborn screening with neonatal valganciclovir treatment was evaluated. The incremental cost-effectiveness ratio (ICER) was calculated for (1) universal maternal antibody screening with prenatal valacyclovir treatment compared to targeted newborn screening, and (2) universal newborn screening with postnatal valganciclovir treatment compared to targeted newborn screening. We performed a one-way sensitivity analysis. Compared to targeted newborn screening, the ICERs for universal newborn screening and maternal screening were 2 966 296 Japanese Yen (JPY) (21 188 USD) and 1 026 984 JPY (7336 USD), respectively. In all scenarios in the one-way sensitivity analysis, the ICERs of the maternal screening and the universal newborn screening strategies were less than three gross domestic product per capita compared with the targeted newborn screening strategy. Both maternal and universal newborn screening strategies may be cost-effective than a targeted newborn screening program. The potential utility of the maternal screening with valacyclovir treatment strategy, while potentially cost effective in regions with lower baseline seroprevalence rates, requires further study as the modeling was based on limited evidence.
Collapse
Affiliation(s)
- Hirosato Aoki
- Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan.,Department of Neonatology, Kanagawa Children's Medical Center, Yokohama, Kanagawa, Japan
| | - Ari Bitnun
- Department of Paediatrics, Division of Infectious Diseases, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Taito Kitano
- Department of Pediatrics, Nara Medical University, Kashihara, Nara, Japan.,Department of Health Policy and Management, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
32
|
Barlinn R, Dudman SG, Rollag H, Trogstad L, Lindstrøm JC, Magnus P. Maternal cytomegalovirus infection and delayed language development in children at 3 years of age-a nested case-control study in a large population-based pregnancy cohort. PLoS One 2022; 17:e0278623. [PMID: 36455052 PMCID: PMC9714838 DOI: 10.1371/journal.pone.0278623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 11/18/2022] [Indexed: 12/05/2022] Open
Abstract
INTRODUCTION Maternal cytomegalovirus (CMV) infection in pregnancy may result in vertical transmission of CMV to the child. Long-term effects of congenital CMV infection include visual, cognitive as well as neurological impairment. The aim of this study was to estimate the odds ratios for CMV seropositivity and seroconversion in mothers, with and without delayed language development in 3 year old children, nested within a large cohort. MATERIAL AND METHODS The Norwegian Mother, Father and Child Cohort Study (MoBa) is a prospective population-based pregnancy cohort that includes 95 200 mothers and 114 500 children. Blood samples were obtained from mothers during pregnancy weeks 17 or 18 in pregnancy and after birth. We included 300 women from MoBa with children suffering from delayed language development at three years of age, based on validated questionnaires. Within the cohort, 1350 randomly selected women were included as controls to perform a nested case-control study. The cases and controls were tested for CMV IgG antibodies by an enzyme-linked immunosorbent assay. RESULTS Among mothers of cases, 63.2% were CMV-IgG positive in the sample at birth, as compared to 55.9% among controls; OR 1.36, (95% CI; 1.05 to 1.76). Also, among case mothers, 8/118 (6.8%) initially seronegative cases, seroconverted. Among initially seronegative controls, seroconversion occurred in 23/618 (3.7%) mothers. The OR for seroconversion in cases as compared to control mothers was 1.88 (CI; 0.82 to 4.31), thus not statistically significant different. CONCLUSION This study shows a higher risk of delayed language development at three years of age in children born by mothers seropositive for CMV, compared to children born from seronegative mothers.
Collapse
Affiliation(s)
- Regine Barlinn
- Division for Infection Control and Environmental Health, Department of Microbiology, Oslo University Hospital, Norwegian Institute of Public Health, Oslo, Norway
- * E-mail:
| | - Susanne G. Dudman
- Department of Microbiology, Oslo University Hospital, and University of Oslo, Oslo, Norway
| | - Halvor Rollag
- Department of Microbiology, Oslo University Hospital, and University of Oslo, Oslo, Norway
| | - Lill Trogstad
- Division for Infection Control and Environmental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Jonas C. Lindstrøm
- Division for Infection Control and Environmental Health, Department of Methods Development and Analytics, Norwegian Institute of Public Health, Oslo, Norway
| | - Per Magnus
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
- University of Oslo, Oslo, Norway
| |
Collapse
|
33
|
Carneiro VCDS, Pereira JG, de Paula VS. Family Herpesviridae and neuroinfections: current status and research in progress. Mem Inst Oswaldo Cruz 2022; 117:e220200. [PMID: 36417627 PMCID: PMC9677594 DOI: 10.1590/0074-02760220200] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/10/2022] [Indexed: 11/22/2022] Open
Abstract
This article addresses the relationship between human herpesviruses (HHVs) and neuroinfections. Alphaherpesviruses, betaherpesviruses and gammaherpesviruses are neurotropic viruses that establish latency and exhibit reactivation capacity. Encephalitis and meningitis are common in cases of HHV. The condition promoted by HHV infection is a purported trigger for certain neurodegenerative diseases. Ongoing studies have identified an association between HSV-1 and the occurrence of Alzheimer's disease, multiple sclerosis and infections by HHV-6 and Epstein-Barr virus. In this review, we highlight the importance of research investigating the role of herpesviruses in the pathogenesis of diseases that affect the nervous system and describe other studies in progress.
Collapse
Affiliation(s)
| | | | - Vanessa Salete de Paula
- Fundação Oswaldo Cruz-Fiocruz, Instituto Oswaldo Cruz, Laboratório de Virologia Molecular, Rio de Janeiro, RJ, Brasil,+ Corresponding author:
| |
Collapse
|
34
|
Almishaal AA. Knowledge of cytomegalovirus infection among women in Saudi Arabia: A cross-sectional study. PLoS One 2022; 17:e0274863. [PMID: 36173957 PMCID: PMC9522269 DOI: 10.1371/journal.pone.0274863] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 09/07/2022] [Indexed: 12/02/2022] Open
Abstract
Background Cytomegalovirus (CMV) is the most common intrauterine congenital infection in humans. Worldwide seropositive rates of CMV are considerably high among women of childbearing age. There is currently no optimal drug treatment nor a vaccine for congenital CMV infection and therefore the best available program to date of prevention is practicing standard hygienic measures. The success of this program relies on women’s knowledge of CMV modes of transmissions, and risk and preventative behaviors. Objective The current study aims to assess the awareness and knowledge of CMV infection among women in Saudi Arabia. Method In this cross-sectional study, an online self-administered questionnaire was distributed to women 18 years of age or older residing in all regions of Saudi Arabia. The questionnaire included questions to assess awareness of CMV, knowledge of symptoms, transmission, and preventative measures of CMV infection. Results Out of the 1004 women who completed the questionnaire, self-reported knowledge of CMV was considerably low with only 82 women (8.17%) having heard of CMV infection. Most women reported learning about CMV from the internet and university. In binary logistic analyses, women pursuing studies in healthcare and those working in health professions, as well as those with undergraduate and graduate college degrees were significantly associated with higher knowledge of CMV. Urban area of residence was significantly associated with lower levels of knowledge of CMV. Among women who reported they had heard of CMV, their knowledge of CMV modes of transmission, symptoms, and preventative measures was considerably low. Regarding the transmission route, 23% reported no knowledge of modes of transmission, 59.75% reported mother-to-child transmission, 48.78% reported sexual intercourse, and 45% reported contact with body fluids of an infected person. Regarding knowledge of symptoms of congenital CMV, mental retardation and death were the most commonly reported clinical presentations. Conclusion The current study showed that the overall knowledge of CMV is very low among women in Saudi Arabia. Working in the healthcare field and higher education levels were significantly associated with better knowledge of CMV. It is crucial that women are provided with information regarding CMV-associated complications and preventative measures against mother-to-fetus transmission of CMV.
Collapse
Affiliation(s)
- Ali A. Almishaal
- College of Applied Medical Sciences, University of Hail, Hail, Saudi Arabia
- * E-mail:
| |
Collapse
|
35
|
Zdziarski P, Gamian A. High Monocyte Count Associated with Human Cytomegalovirus Replication In Vivo and Glucocorticoid Therapy May Be a Hallmark of Disease. Int J Mol Sci 2022; 23:ijms23179595. [PMID: 36076989 PMCID: PMC9455616 DOI: 10.3390/ijms23179595] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/16/2022] [Accepted: 08/19/2022] [Indexed: 11/24/2022] Open
Abstract
Cytomegalovirus (CMV) syndrome and infectious disease are defined as pathogen detection with appropriate clinical symptoms, but there are not pathognomonic signs of CMV disease. Although the prodrome of acute minor viral infections leukopenia (lymphopenia and neutropenia) is noted with onset of fever, followed by monocytosis, the role of monocytosis in CMV disease has not been described. Furthermore, under influence of corticosteroid therapy, CMV reactivation and monocytosis are described, but without a strict relationship with steroids dose. In the study, the monocyte level was investigated during the CMV infectious process. Regrettably, a non-selected group of 160 patients with high CMV viremia showed high dispersion of monocyte level and comparable with the median value for healthy subjects. Therefore, we investigated monocyte level in CMV-infected patients in relation to the logarithmic phase of the infectious process. Samples from patients with active CMV replication (exponential growth of CMV viremia) were tested. Significant monocytosis (above 1200/µL) during the logarithmic phase of CMV infection (with exponent between 3.23 and 5.77) was observed. Increased count and percentage of monocytes correlated with viral replication in several clinical situations except when there was a rapid recovery without relapse. Furthermore, glucocorticoids equivalent to 10 and 20 mg of dexamethasone during a 2–3-week period caused monocytosis—significant increase (to 1604 and 2214/µL, respectively). Conclusion: In light of the logarithmic increase of viral load, high monocytosis is a hallmark of CMV replication. In the COVID-19 era, presence of high virus level, especially part of virome (CMV) in the molecular technique, is not sufficient for the definition of either proven or probable CMV replication at any site. These preliminary observations merit additional studies to establish whether this clinical response is mediated by monocyte production or by decrease of differentiation to macrophages.
Collapse
Affiliation(s)
- Przemyslaw Zdziarski
- Lower Silesian Oncology, Pulmonology and Hematology Center, P.O. Box 1818, 50-385 Wroclaw, Poland
- Correspondence:
| | - Andrzej Gamian
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland
| |
Collapse
|
36
|
Modestly protective cytomegalovirus vaccination of young children effectively prevents congenital infection at the population level. Vaccine 2022; 40:5179-5188. [PMID: 35907677 DOI: 10.1016/j.vaccine.2022.07.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 11/21/2022]
Abstract
A vaccine to prevent congenital cytomegalovirus infection (cCMV) is a public health priority. cCMV results from maternal primary or non-primary CMV infection (reinfection, or reactivation of chronic infection) during pregnancy. Young children are a major source of transmission to pregnant women because they shed CMV at high viral loads for prolonged periods. CMV vaccines evaluated in clinical trials so far have demonstrated only approximately 50% efficacy against maternal primary infection. None of these have been approved, as higher levels of vaccine efficacy are assumed to be required to substantially reduce cCMV prevalence. Here, we designed a mathematical model to capture the relationship between viral shedding by young children and maternal CMV infections during pregnancy. Using this model, we were able to quantify the impact of CMV post-infection immunity on protecting against reinfection and viral shedding. There was a 36% reduction in the risk of infection to a seropositive person with post-infection immunity (reinfection) versus a seronegative person without this immunity (primary infection), given the same exposure. Viral shedding following reinfection was only 34% the quantity of that following primary infection. Our model also predicted that a vaccine that confers the equivalent of post-infection immunity, when given to young children, would markedly reduce both CMV transmission to pregnant women and the prevalence of cCMV. Thus, we predict that existing vaccine candidates that have been shown to be only modestly protective may in fact be highly effective at preventing cCMV by interrupting child-to-mother transmission.
Collapse
|
37
|
Elliott RP, Freeman BP, Meier JL, El-Herte R. Acute Cytomegalovirus Illness in an Immunocompetent Adult Causing Intravascular Hemolysis and Suspected Hemophagocytic Lymphohistiocytosis. Case Rep Infect Dis 2022; 2022:7949471. [PMID: 35847599 PMCID: PMC9286925 DOI: 10.1155/2022/7949471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 06/15/2022] [Indexed: 11/29/2022] Open
Abstract
Background Primary cytomegalovirus (CMV) infection of the immunocompetent host usually produces little-to-no illness. Occasionally, the infection results in mononucleosis syndrome, protracted fever, hepatitis, tissue-invasive disease, or Guillain-Barré syndrome. Hemolytic anemia and hemophagocytic lymphohistiocytosis (HLH) are rare complications that have not been reported to co-occur. Having hemolytic anemia in conjunction with more common findings of fever and hepatitis complicates the diagnosis of HLH. Case Presentation. A 34-year-old male with previously good health presented with a prolonged febrile illness, jaundice, and anemia. An extensive work-up during hospitalization revealed intravascular hemolytic anemia, leukopenia, hepatosplenomegaly, and biopsy evidence of extensive lymphohistiocytic infiltration of the liver with microgranulomata and sinusoidal hemophagocytosis. Soluble CD25 level was mildly elevated at 1200.3 pg/mL and the HScore calculation (fever, bicytopenia, hepatosplenomegaly, aspartate aminotransaminase 99 IU/L, ferritin 1570 ng/mL, fibrinogen 488 mg/dL, and triglycerides 173 mg/dL) suggested a moderate probability of reactive HLH. Primary CMV infection was diagnosed based on CMV IgM positivity, low CMV IgG avidity index, and low-grade CMV DNAemia. The CMV antigen was not detected in the liver biopsy, and the bone marrow biopsy was unremarkable. The illness began to improve before he received oral valganciclovir for 5 days, and he was in good health 10 months later. Conclusion Acute CMV illness in an immunocompetent adult can present with hemolytic anemia and clinicopathologic abnormalities consistent with a form fruste of HLH. The illness is likely due to an excessive or unbalanced immune response that may self-correct.
Collapse
Affiliation(s)
- Ross P. Elliott
- Internal Medicine Residency Program, MercyOne Medical Center and Clinics, Des Moines, IA 50314, USA
| | | | - Jeffery L. Meier
- Iowa City Veterans Affairs Health Care System, Iowa City, IA 52246, USA
- Division of Infectious Diseases, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Rima El-Herte
- Division of Infectious Diseases, Department of Medicine, Creighton, University School of Medicine and CHI Health, Omaha, NE 68124, USA
| |
Collapse
|
38
|
Balegamire SJ, McClymont E, Croteau A, Dodin P, Gantt S, Besharati AA, Renaud C, Mâsse B, Boucoiran I. Prevalence, incidence, and risk factors associated with cytomegalovirus infection in healthcare and childcare worker: a systematic review and meta-analysis. Syst Rev 2022; 11:131. [PMID: 35754052 PMCID: PMC9235282 DOI: 10.1186/s13643-022-02004-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 06/11/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Cytomegalovirus (CMV) is transmitted by direct contact with body fluids from infected individuals. Transmission of CMV in households, particularly those with young children, contributes significantly to CMV infection in the general population. However, little is known about the contribution of occupational healthcare or childcare exposure to risk of CMV infection. OBJECTIVES To determine CMV seroprevalence, incidence of primary infection, and associated risk factors in healthcare and childcare workers. METHODS Six electronic databases were searched systematically for publications on CMV infection in healthcare and childcare workers until March 7, 2022. Two authors independently evaluated the literature for quality and inclusion in our analyses. The pooled results for seroprevalence, incidence, and relative risk (RR) were determined using a random effects model. Heterogeneity among studies was quantified and further investigated in subgroup analysis and meta-regression. Publication bias was assessed using funnel plot. Statistical analyses were preformed using R version 4.05. RESULTS Forty-eight articles were included in this meta-analysis (quality assessment: 18 good, 14 fair, and 16 poor). Pooled CMV seroprevalence was 59.3% (95% CI: 49.8-68.6) among childcare workers and 49.5% (95% CI: 40.3-58.7) among healthcare workers, and pooled incidences of primary CMV infection per 100 person-years were respectively 7.4 (95% CI: 3.9-11.8) and 3.1 (95% CI: 1.3-5.6). RR for primary infection compared to controls were 3.4 (95% CI: 1.3-8.8) and 1.3 (95% CI: 0.6-2.7) for healthcare and childcare workers, respectively. The odds of CMV seropositivity were 1.6 (95% CI: 1.2-2.3) times higher for childcare workers compared to controls, but not significantly different between healthcare workers and controls (0.9; 95% CI: 0.6-1.2). CMV seropositivity in both groups was significantly associated with having one or more children residing at home, marital status, ethnicity, and age. CONCLUSIONS Childcare workers, but not healthcare workers, have an increased risk of prevalent and incident CMV infection, a risk that is further increased with the presence of at least one child living at home. These findings suggest that enforcing simple, conventional hygienic measures in childcare settings could help reduce transmission of CMV, and that special precautionary measures for preventing CMV infection may not be required for pregnant healthcare workers. SYSTEMATIC REVIEW REGISTRATION PROSPERO CRD42020139756.
Collapse
Affiliation(s)
- Safari Joseph Balegamire
- Department of Social and Preventive Medicine, École de Santé Publique de Université de Montréal, Montreal, QC, Canada. .,Women and Children's Infectious Diseases Center, CHU Sainte-Justine Research Center, Montreal, QC, Canada.
| | - Elisabeth McClymont
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Agathe Croteau
- National Institute of Public Health of Québec, Québec City, QC, Canada
| | - Philippe Dodin
- Women and Children's Infectious Diseases Center, CHU Sainte-Justine Research Center, Montreal, QC, Canada
| | - Soren Gantt
- Women and Children's Infectious Diseases Center, CHU Sainte-Justine Research Center, Montreal, QC, Canada.,Department of Microbiology, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Amir Abbas Besharati
- Women and Children's Infectious Diseases Center, CHU Sainte-Justine Research Center, Montreal, QC, Canada
| | - Christian Renaud
- Women and Children's Infectious Diseases Center, CHU Sainte-Justine Research Center, Montreal, QC, Canada.,Department of Microbiology, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Benoît Mâsse
- Department of Social and Preventive Medicine, École de Santé Publique de Université de Montréal, Montreal, QC, Canada.,Applied Clinical Research Unit, CHU Sainte Justine Research Center, Montreal, QC, Canada
| | - Isabelle Boucoiran
- Department of Social and Preventive Medicine, École de Santé Publique de Université de Montréal, Montreal, QC, Canada.,Women and Children's Infectious Diseases Center, CHU Sainte-Justine Research Center, Montreal, QC, Canada.,Division of Maternofetal Medicine, Department of Obstetrics and Gynecology, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
39
|
Fisher SA, Miller ES, Yee LM, Grobman WA, Premkumar A. Universal First-Trimester Cytomegalovirus Screening and Valaciclovir Prophylaxis in Pregnant Persons: A Cost-Effectiveness Analysis. Am J Obstet Gynecol MFM 2022; 4:100676. [PMID: 35714861 DOI: 10.1016/j.ajogmf.2022.100676] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Recent studies suggest a possible benefit of valaciclovir prophylaxis to prevent vertical transmission after a positive serologic screen for primary maternal cytomegalovirus infection during pregnancy, although its cost-effectiveness remains uncertain. OBJECTIVE We sought to determine the circumstances under which universal first-trimester maternal serologic screening for maternal cytomegalovirus infection, with valaciclovir prophylaxis to prevent congenital cytomegalovirus, is cost-effective. STUDY DESIGN We performed a decision analysis from the perspective of the pregnant person to assess whether universal maternal screening in the first trimester, with subsequent valaciclovir prophylaxis (8g/day from time of positive serologic screen for primary maternal cytomegalovirus infection through 21 weeks' gestation) for those who are acutely infected, is cost-effective compared to usual care (i.e., no routine serologic screening, but amniocentesis if mid-trimester sonographic findings suggest cytomegalovirus). For baseline estimates, we assumed a 35% risk of congenital cytomegalovirus after primary maternal infection and a 71% risk reduction with valaciclovir. We varied valaciclovir's efficacy to identify whether and at what threshold universal screening would be estimated to be cost-effective, compared to usual care. Monte Carlo analyses were performed. A willingness-to-pay threshold of $100,000/quality-adjusted life year was used to define cost-effectiveness. RESULTS Under base-case estimates, first-trimester universal screening and valaciclovir prophylaxis for seropositive pregnant persons with acute cytomegalovirus infection is not cost-effective, with a cost of $137,854 per maternal QALY, but results in 14 fewer cytomegalovirus-affected children per 100,000 pregnancies compared to usual care. In one-way sensitivity analysis, universal screening and treatment is estimated to be the cost-effective strategy if the incidence of primary maternal cytomegalovirus infection exceeds 2.6%, baseline risk of vertical transmission of cytomegalovirus without prophylaxis is greater than 36.8%, and the risk reduction of vertical transmission of cytomegalovirus with valaciclovir prophylaxis exceeds 75.9%. In Monte Carlo analyses, first-trimester universal serologic screening with valaciclovir prophylaxis is estimated to be the cost-effective strategy in 46.8% of runs. CONCLUSION Universal first-trimester serologic screening with valaciclovir prophylaxis is not the cost-effective strategy for antenatal management of cytomegalovirus under the base-case estimates. Although universal screening is cost-effective in certain circumstances when the efficacy of valaciclovir exceeds the base case, that result is not robust to variation of estimates across their reasonable ranges. These data can inform future studies to evaluate screening and treatment to prevent congenital CMV.
Collapse
Affiliation(s)
- Stephanie A Fisher
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.
| | - Emily S Miller
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.
| | - Lynn M Yee
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.
| | - William A Grobman
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, The Ohio State University College of Medicine, Cleveland, Ohio.
| | - Ashish Premkumar
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois; Department of Obstetrics and Gynecology, John H. Stroger, Jr. Hospital of Cook County, Chicago, Illinois.
| |
Collapse
|
40
|
Savedchuk S, Raslan R, Nystrom S, Sparks MA. Emerging Viral Infections and the Potential Impact on Hypertension, Cardiovascular Disease, and Kidney Disease. Circ Res 2022; 130:1618-1641. [PMID: 35549373 DOI: 10.1161/circresaha.122.320873] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Viruses are ubiquitous in the environment and continue to have a profound impact on human health and disease. The COVID-19 pandemic has highlighted this with impressive morbidity and mortality affecting the world's population. Importantly, the link between viruses and hypertension, cardiovascular disease, and kidney disease has resulted in a renewed focus and attention on this potential relationship. The virus responsible for COVID-19, SARS-CoV-2, has a direct link to one of the major enzymatic regulatory systems connected to blood pressure control and hypertension pathogenesis, the renin-angiotensin system. This is because the entry point for SARS-CoV-2 is the ACE2 (angiotensin-converting enzyme 2) protein. ACE2 is one of the main enzymes responsible for dampening the primary effector peptide Ang II (angiotensin II), metabolizing it to Ang-(1-7). A myriad of clinical questions has since emerged and are covered in this review. Several other viruses have been linked to hypertension, cardiovascular disease, and kidney health. Importantly, patients with high-risk apolipoprotein L1 (APOL1) alleles are at risk for developing the kidney lesion of collapsing glomerulopathy after viral infection. This review will highlight several emerging viruses and their potential unique tropisms for the kidney and cardiovascular system. We focus on SARS-CoV-2 as this body of literature in regards to cardiovascular disease has advanced significantly since the COVID-19 pandemic.
Collapse
Affiliation(s)
- Solomiia Savedchuk
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC (S.S., S.N., M.A.S.)
| | - Rasha Raslan
- Internal Medicine, Virginia Commonwealth University, Richmond (R.R.)
| | - Sarah Nystrom
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC (S.S., S.N., M.A.S.)
| | - Matthew A Sparks
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC (S.S., S.N., M.A.S.)
- Renal Section, Durham VA Health Care System, NC (M.A.S.)
| |
Collapse
|
41
|
Coste Mazeau P, Jacquet C, Muller C, Courant M, El Hamel C, Chianea T, Hantz S, Alain S. Potential of Anti-CMV Immunoglobulin Cytotect CP® In Vitro and Ex Vivo in a First-Trimester Placenta Model. Microorganisms 2022; 10:microorganisms10040694. [PMID: 35456746 PMCID: PMC9030298 DOI: 10.3390/microorganisms10040694] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/18/2022] [Accepted: 03/19/2022] [Indexed: 12/04/2022] Open
Abstract
Background: Congenital CMV infection is the leading cause of neonatal neurological deficit. We herein studied in vitro and ex vivo the potential of the hyperimmune globulin Cytotect CP® (Biotest, Germany) for congenital infection prevention and treatment. Methods: In vitro neutralization assays were conducted in fibroblasts and retinal epithelial cells on the CMV strains TB40/E and VHL/E to determine the 50% and 90% neutralizing doses (ND50 and ND90). The toxicity was assessed by measuring LDH release. Ex vivo assays were conducted in first-trimester villi explants with the TB40/E strain, namely, neutralization assays, the prevention of villi infection, and the inhibition of viral replication in infected villi. Viability was assessed by β-HCG quantification in supernatants. Results: The in vitro neutralization tests showed that Cytotect CP®® inhibits the development of infection foci (DN50: 0.011–0.014 U/mL for VHL/E and 0.032–0.033 U/mL for TB40E) without any toxicity. In the ex vivo neutralization assays, the DN50 were 0.011 U/mL on day 7 and 0.093 U/mL on day 14. For the prevention of villi infection, the EC50 was 0.024 U/mL on day 7. Cytotect-CP® did not inhibit viral growth in infected villi. No impact on villi viability was observed. Conclusions: These results sustained that Cytotect CP® has the potential to prevent CMV congenital infection.
Collapse
Affiliation(s)
- Perrine Coste Mazeau
- RESINFIT, UMR1092, University of Limoges, 2 Rue du Pr Descottes, 87000 Limoges, France; (C.J.); (C.M.); (S.H.)
- National Institute of Health and Medical Research INSERM, UMR 1092, 2 Rue du Pr Descottes, 87000 Limoges, France;
- National Reference Center for Herpesviruses, Virology Department, CHU Limoges, 2 Rue Martin Luther King, 87000 Limoges, France
- Gynecology and Obstetrics Department, CHU Limoges, 87000 Limoges, France
- Correspondence: (P.C.M.); (S.A.); Tel.: +33-5-5505-6164 (P.C.M.); +33-5-5505-6724 (S.A.); Fax: +33-5-5505-6722 (P.C.M. & S.A.)
| | - Chloé Jacquet
- RESINFIT, UMR1092, University of Limoges, 2 Rue du Pr Descottes, 87000 Limoges, France; (C.J.); (C.M.); (S.H.)
- National Institute of Health and Medical Research INSERM, UMR 1092, 2 Rue du Pr Descottes, 87000 Limoges, France;
- National Reference Center for Herpesviruses, Virology Department, CHU Limoges, 2 Rue Martin Luther King, 87000 Limoges, France
| | - Clotilde Muller
- RESINFIT, UMR1092, University of Limoges, 2 Rue du Pr Descottes, 87000 Limoges, France; (C.J.); (C.M.); (S.H.)
- National Institute of Health and Medical Research INSERM, UMR 1092, 2 Rue du Pr Descottes, 87000 Limoges, France;
| | - Mathis Courant
- National Institute of Health and Medical Research INSERM, UMR 1092, 2 Rue du Pr Descottes, 87000 Limoges, France;
| | - Chahrazed El Hamel
- Mother and Child Biobank (CB-HME), Pediatric Department, Hôpital de la Mère et de l’Enfant, CHU Limoges, 87000 Limoges, France;
| | - Thierry Chianea
- Department of Biochemistry and Molecular Genetics, CHU Limoges, 87000 Limoges, France;
| | - Sébastien Hantz
- RESINFIT, UMR1092, University of Limoges, 2 Rue du Pr Descottes, 87000 Limoges, France; (C.J.); (C.M.); (S.H.)
- National Institute of Health and Medical Research INSERM, UMR 1092, 2 Rue du Pr Descottes, 87000 Limoges, France;
- National Reference Center for Herpesviruses, Virology Department, CHU Limoges, 2 Rue Martin Luther King, 87000 Limoges, France
| | - Sophie Alain
- RESINFIT, UMR1092, University of Limoges, 2 Rue du Pr Descottes, 87000 Limoges, France; (C.J.); (C.M.); (S.H.)
- National Institute of Health and Medical Research INSERM, UMR 1092, 2 Rue du Pr Descottes, 87000 Limoges, France;
- National Reference Center for Herpesviruses, Virology Department, CHU Limoges, 2 Rue Martin Luther King, 87000 Limoges, France
- Correspondence: (P.C.M.); (S.A.); Tel.: +33-5-5505-6164 (P.C.M.); +33-5-5505-6724 (S.A.); Fax: +33-5-5505-6722 (P.C.M. & S.A.)
| |
Collapse
|
42
|
Kschonsak M, Johnson MC, Schelling R, Green EM, Rougé L, Ho H, Patel N, Kilic C, Kraft E, Arthur CP, Rohou AL, Comps-Agrar L, Martinez-Martin N, Perez L, Payandeh J, Ciferri C. Structural basis for HCMV Pentamer receptor recognition and antibody neutralization. SCIENCE ADVANCES 2022; 8:eabm2536. [PMID: 35275719 PMCID: PMC8916737 DOI: 10.1126/sciadv.abm2536] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 01/25/2022] [Indexed: 06/14/2023]
Abstract
Human cytomegalovirus (HCMV) represents the viral leading cause of congenital birth defects and uses the gH/gL/UL128-130-131A complex (Pentamer) to enter different cell types, including epithelial and endothelial cells. Upon infection, Pentamer elicits the most potent neutralizing response against HCMV, representing a key vaccine candidate. Despite its relevance, the structural basis for Pentamer receptor recognition and antibody neutralization is largely unknown. Here, we determine the structures of Pentamer bound to neuropilin 2 (NRP2) and a set of potent neutralizing antibodies against HCMV. Moreover, we identify thrombomodulin (THBD) as a functional HCMV receptor and determine the structures of the Pentamer-THBD complex. Unexpectedly, both NRP2 and THBD also promote dimerization of Pentamer. Our results provide a framework for understanding HCMV receptor engagement, cell entry, antibody neutralization, and outline strategies for antiviral therapies against HCMV.
Collapse
Affiliation(s)
- Marc Kschonsak
- Department of Structural Biology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Matthew C. Johnson
- Department of Structural Biology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Rachel Schelling
- University of Lausanne (UNIL), Lausanne University Hospital (CHUV), Department of Medicine, Division of Immunology and Allergy, Center for Human Immunology (CHIL), Lausanne, Switzerland
| | - Evan M. Green
- Department of Structural Biology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Lionel Rougé
- Department of Structural Biology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Hoangdung Ho
- Department of Structural Biology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Nidhi Patel
- Department of Structural Biology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Cem Kilic
- Department of Biochemical and Cellular Pharmacology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Edward Kraft
- Department of Biomolecular Resources, Genentech Inc., South San Francisco, CA 94080, USA
| | - Christopher P. Arthur
- Department of Structural Biology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Alexis L. Rohou
- Department of Structural Biology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Laetitia Comps-Agrar
- Department of Biochemical and Cellular Pharmacology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Nadia Martinez-Martin
- Department of Microchemistry, Proteomics and Lipidomics, Genentech Inc, South San Francisco, CA 94080, USA
| | - Laurent Perez
- University of Lausanne (UNIL), Lausanne University Hospital (CHUV), Department of Medicine, Division of Immunology and Allergy, Center for Human Immunology (CHIL), Lausanne, Switzerland
| | - Jian Payandeh
- Department of Structural Biology, Genentech Inc., South San Francisco, CA 94080, USA
| | - Claudio Ciferri
- Department of Structural Biology, Genentech Inc., South San Francisco, CA 94080, USA
| |
Collapse
|
43
|
Geris JM, Spector LG, Roesler M, Hernandez-Alvarado N, Blackstad M, Nelson HH, Schleiss MR. High prevalence of asymptomatic CMV shedding in healthy children attending the minnesota state fair. J Clin Virol 2022; 148:105102. [PMID: 35158280 PMCID: PMC8918022 DOI: 10.1016/j.jcv.2022.105102] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 02/01/2022] [Accepted: 02/05/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND Young children in the household are a known risk factor for maternal CMV infection and consequently, congenital infection in infants. However, little is known about viral shedding in pre-school aged children. OBJECTIVES To estimate the prevalence of CMV DNA shedding and CMV antibodies among healthy children and their mothers. STUDY DESIGN A study of children ages 0 through 5 years was undertaken at the 2019 Minnesota State Fair. Children and their mothers were assessed for CMV shedding by procurement of a saliva swab for CMV PCR testing. An optional finger-stick for capillary blood was used to assess CMV antibodies. RESULTS A total of 109 children and 85 mothers were enrolled. The prevalence of CMV saliva shedding among children (mean age 3.1 years, SE=0.16) and their mothers was 12/109 (11.0%) and 1/85 (1.2%), respectively. The prevalence of CMV DNA among children peaked at 3 years of age (26%) while the mean viral load was greatest at one year of age (236,693 IU/mL). CMV IgG antibodies among those who agreed to a finger-stick were detected in 16/35 mothers (45.7%) and 0/7 children (0%). Mothers of children aged 5 years or greater had the highest seroprevalence (61.5%). CONCLUSIONS The prevalence of CMV salivary shedding in this unselected sample of young children was approximately 11.0%. The overall maternal seroprevalence in our sample was <50%, suggesting these women are at risk for acquisition of a primary CMV infection in subsequent pregnancies.
Collapse
Affiliation(s)
- Jennifer M Geris
- Division of Epidemiology & Clinical Research, Department of Pediatrics, University of Minnesota, Minneapolis MN, USA; Institute for Molecular Virology, University of Minnesota, Minneapolis MN, USA.
| | - Logan G Spector
- Division of Epidemiology & Clinical Research, Department of Pediatrics, University of Minnesota, Minneapolis MN, USA
| | - Michelle Roesler
- Division of Epidemiology & Clinical Research, Department of Pediatrics, University of Minnesota, Minneapolis MN, USA
| | - Nelmary Hernandez-Alvarado
- Division of Pediatric Infectious Diseases and Immunology, Department of Pediatrics, University of Minnesota, Minneapolis MN, USA
| | - Mark Blackstad
- Division of Pediatric Infectious Diseases and Immunology, Department of Pediatrics, University of Minnesota, Minneapolis MN, USA
| | - Heather H Nelson
- Division of Epidemiology & Community Health, University of Minnesota School of Public Health, Minneapolis MN, USA
| | - Mark R Schleiss
- Institute for Molecular Virology, University of Minnesota, Minneapolis MN, USA; Division of Pediatric Infectious Diseases and Immunology, Department of Pediatrics, University of Minnesota, Minneapolis MN, USA
| |
Collapse
|
44
|
Congenital Cytomegalovirus Infections Mother-Newborn Pair Study in Southern Ethiopia. CANADIAN JOURNAL OF INFECTIOUS DISEASES AND MEDICAL MICROBIOLOGY 2021; 2021:4646743. [PMID: 35003406 PMCID: PMC8739911 DOI: 10.1155/2021/4646743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 12/08/2021] [Accepted: 12/21/2021] [Indexed: 11/17/2022]
Abstract
Introduction. Congenital cytomegalovirus (cCMV) is a common cause of neurodevelopmental delays and sensorineural hearing loss of infants, yet the prevalence of cCMV and the associated factors in Ethiopia are not studied. Hence, this study was to assess the prevalence and associated factors of cCMV in Southern Ethiopia. Methodology. A mother-newborn pair cross-sectional study was conducted at Hawassa University Comprehensive and Specialized Hospital, Ethiopia. Newborn’s saliva sample was tested for cCMV using Alethia CMV molecular assay. Mothers’ serum was tested serologically for anti-CMV IgM and IgG by EUROIMMUN ELISA. Pregnant women responded to a questionnaire about their previous and current obstetric history and sociodemographic characteristics. The chi-square (χ2) test and independent-sample t-test were used to determine the associations between infections and possible risk factors; then, potential variables were screened for multivariable analysis. Results. A total of 593 mother-newborn pairs were assessed. CMV was detected in 14 of 593 newborn saliva swabs (2.4%; 95% CI 1.2–3.7). As assessed by CMV IgM-positive results, maternal CMV seropositivity was 8.3% (49/593); thus, the rate of mother-to-child transmission of CMV was 28% (14/49) among CMV IgM-positive women. Congenital CMV infection was significantly associated with maternal exposure through nursery school children in the household, women sharing a feeding cup with children, and any of the detected curable STIs during pregnancy. Birth weight was negatively associated with CMV infection. Maternal age, gravidity, level of education, and sharing of children feeding utensils were not associated with cCMV infection. Conclusion. A high rate of cCMV infection in the absence of awareness demands further in-depth investigation in Ethiopia. Thus, policymakers must take appropriate action through the antenatal care system for prevention strategies and put in place a constant health education and awareness creation of pregnant women about the causes of infection and hygienic measures.
Collapse
|
45
|
Huang X, Sun P, Qin Y, Wang XJ, Wang M, Lin Y, Zhou R, Hu W, Liu Q, Yu X, Qin A. Disulfiram attenuates MCMV-Induced pneumonia by inhibition of NF-κB/NLRP3 signaling pathway in immunocompromised mice. Int Immunopharmacol 2021; 103:108453. [PMID: 34959186 DOI: 10.1016/j.intimp.2021.108453] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 11/05/2022]
Abstract
Cytomegalovirus (CMV) pneumonia in immunocompromised individuals is associated with damaging hyperinflammation and leads to high morbidity and mortality. It is urgently needed to develop new strategies to treat CMV-induced pneumonia. As disulfiram (DSF) reportedly inhibits inflammatory responses in different disease models, its therapeutic effects in CMV-induced pneumonia are proposed. In this study, we demonstrated that DSF effectively attenuated pulmonary injury and improved survival in murine CMV (MCMV) pneumonia model. DSF treatment inhibited lung inflammatory responses, e.g. reducing pro-inflammatory cytokines, upregulating anti-inflammatory cytokine, and lowering the accumulation of leukocytes in the lung. Similar to the in vivo results, DSF attenuated inflammatory responses and modulated NF-κB/NLRP3 inflammasome activation in MCMV-infected BMDMs. Furthermore, DSF reduced pulmonary fibrosis and viral loads in MCMV pneumonia mice and BMDMs. The mechanism of anti-inflammatory effects of DSF may due to its regulating NF-κB signaling and NLRP3 inflammasome activation. Collectively, our results suggest that DSF-mediated anti-hyperinflammatory effects have potentials for therapy of human CMV pneumonia.
Collapse
Affiliation(s)
- Xiaotao Huang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Ping Sun
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Yuyan Qin
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Xiao-Juan Wang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Mengyi Wang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Yongtong Lin
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Ruiqing Zhou
- Department of Hematology, Guangzhou First People's Hospital, 510180. Guangzhou, China
| | - Wenhui Hu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Qifa Liu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Xiyong Yu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China.
| | - Aiping Qin
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China.
| |
Collapse
|
46
|
Wang S, Xu X, Sun C, Zhang J, He X, Zhang Z, Huang H, Yan J, Jin W, Mao G. Sulphated glucuronomannan tetramer and hexamer from Sargassum thunbergii exhibit anti-human cytomegalovirus activity by blocking viral entry. Carbohydr Polym 2021; 273:118510. [PMID: 34560939 DOI: 10.1016/j.carbpol.2021.118510] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/21/2021] [Accepted: 07/27/2021] [Indexed: 11/19/2022]
Abstract
Human cytomegalovirus (HCMV) remains a major public health burden worldwide. The anti-HCMV activity of glucuronomannan oligosaccharides (Gs) and sulphated glucuronomannan oligosaccharides (SGs) was investigated. Among these Gs and SGs, G4S1 and G6S1 (higher sulphated glucuronomannan tetramer and hexamer) showed satisfactory anti-HCMV activity starting at 50 μg/mL and 10 μg/mL, respectively. The results of the morphology, western blotting, qPCR and TCID50 assay showed that they prevented lytic cytopathic changes, inhibited the expression of IE1/2 and UL44, and reduced the UL123 copy number and virus titre significantly. It was interesting to note that degree of sulphation and polymerization was more important for anti-HCMV activity. Moreover, the anti-HCMV activities of G4S1 and G6S1 were stable when stored at 4 °C, -20 °C, and -80 °C for at least three months and mainly occurred in the early stage of HCMV infection through the negative charge of the sulphate groups and the interaction between SGs and the host cells.
Collapse
Affiliation(s)
- Sanying Wang
- Zhejiang Provincial Key Lab of Geriatrics & Geriatrics Institute of Zhejiang Province, Department of Geriatrics, Zhejiang Hospital, Hangzhou 310030, PR China
| | - Xiaogang Xu
- Zhejiang Provincial Key Lab of Geriatrics & Geriatrics Institute of Zhejiang Province, Department of Geriatrics, Zhejiang Hospital, Hangzhou 310030, PR China
| | - Chuan Sun
- Zhejiang Provincial Key Lab of Geriatrics & Geriatrics Institute of Zhejiang Province, Department of Geriatrics, Zhejiang Hospital, Hangzhou 310030, PR China
| | - Jing Zhang
- Zhejiang Provincial Key Lab of Geriatrics & Geriatrics Institute of Zhejiang Province, Department of Geriatrics, Zhejiang Hospital, Hangzhou 310030, PR China
| | - Xinyue He
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, PR China
| | - Zhongshan Zhang
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, Huzhou University, Huzhou 313000, PR China
| | - Hong Huang
- Zhejiang Provincial Key Lab of Geriatrics & Geriatrics Institute of Zhejiang Province, Department of Geriatrics, Zhejiang Hospital, Hangzhou 310030, PR China
| | - Jing Yan
- Zhejiang Provincial Key Lab of Geriatrics & Geriatrics Institute of Zhejiang Province, Department of Geriatrics, Zhejiang Hospital, Hangzhou 310030, PR China.
| | - Weihua Jin
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, PR China.
| | - Genxiang Mao
- Zhejiang Provincial Key Lab of Geriatrics & Geriatrics Institute of Zhejiang Province, Department of Geriatrics, Zhejiang Hospital, Hangzhou 310030, PR China.
| |
Collapse
|
47
|
Goetzl L, Stephens AJ, Schlesinger Y, Darbinian N, Merabova N, Hillel M, Hirsch AJ, Streblow DN, Frias AE, Roberts VHJ, Haese NN, Mani A, Eldar-Yedidia Y. Fetal Central Nervous System Derived Extracellular Vesicles: Potential for Non-invasive Tracking of Viral Mediated Fetal Brain Injury. FRONTIERS IN VIROLOGY (LAUSANNE, SWITZERLAND) 2021; 1:782863. [PMID: 40012720 PMCID: PMC11864790 DOI: 10.3389/fviro.2021.782863] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
Introduction Extracellular vesicles derived from the fetal central nervous system (FCNSEs) can be purified from maternal serum or plasma using the protein Contactin-2/TAG1that is expressed almost exclusively by developing neurons in the hippocampus, cerebral cortex and cerebellum. We hypothesized that fetal CNSEs could be used to non-invasively detect and quantify viral mediated in-utero brain injury in the first trimester. Materials and Methods First trimester maternal samples were collected from a human clinical population infected with primary cytomegalovirus (CMV) and a non-human primate model of Zika (ZIKV) infection. In the CMV cohort, a nested case control study was performed comparing pregnancies with and without fetal infection. Cases of fetal infection were further subdivided into those with and without adverse neurologic outcome. ZIKV samples were collected serially following maternal inoculation or saline. All ZIKV cases had histopathologic findings on necropsy. Serum was precipitated with ExoQuick solution and FCEs were isolated with biotinylated anti-Contactin-2/TAG1 antibody-streptavidin matrix immunoabsorption. FCE Synaptopodin (SYNPO) and Neurogranin (NG) protein levels were measured using standard ELISA kits and normalized to the exosome marker CD81. Results Fetal CNSE SYNPO and NG were significantly reduced in cases of first trimester fetal CMV infection compared to those with infection limited to the mother but could not discriminate between fetal infection with and without adverse neurologic outcome. Following ZIKV inoculation, fetal CNSE SYNPO was reduced by 48 h and significantly reduced by day 4. Discussion These data are the first to suggest that first trimester non-invasive diagnosis of fetal viral infection is possible. Fetal CNSEs have the potential to augment clinical and pre-clinical studies of perinatal viral infection. Serial sampling may be needed to discriminate between fetuses that are responding to treatment and/or recovering due to innate defenses and those that have ongoing neuronal injury. If confirmed, this technology may advance the paradigm of first trimester prenatal diagnosis and change the calculus for the cost benefit of CMV surveillance programs in pregnancy.
Collapse
Affiliation(s)
- Laura Goetzl
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Angela J. Stephens
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | | | - Nune Darbinian
- Center for Neural Repair and Rehabilitation, Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Nana Merabova
- Department of Family Medicine, Medical College of Wisconsin-Prevea Health, Green Bay, WI, United States
| | | | - Alec J. Hirsch
- The Vaccine and Gene Institute, Oregon Health and Science University, Beaverton, OR, United States
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, OR, United States
| | - Daniel N. Streblow
- The Vaccine and Gene Institute, Oregon Health and Science University, Beaverton, OR, United States
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, OR, United States
| | - Antonio E. Frias
- Department of Obstetrics and Gynecology, Oregon Health and Science University, Portland, OR, United States
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, OR, United States
| | - Victoria H. J. Roberts
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, OR, United States
| | - Nicole N. Haese
- The Vaccine and Gene Institute, Oregon Health and Science University, Beaverton, OR, United States
- Division of Pathobiology and Immunology, Oregon National Primate Research Center, Beaverton, OR, United States
| | - Arunmani Mani
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | | |
Collapse
|
48
|
Pathogenesis of wild-type-like rhesus cytomegalovirus strains following oral exposure of immune-competent rhesus macaques. J Virol 2021; 96:e0165321. [PMID: 34788083 DOI: 10.1128/jvi.01653-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Rhesus cytomegalovirus (RhCMV) infection of rhesus macaques (Macaca mulatta) is a valuable nonhuman primate model of human CMV (HCMV) persistence and pathogenesis. In vivo studies predominantly use tissue culture-adapted variants of RhCMV that contain multiple genetic mutations compared to wild-type (WT) RhCMV. In many studies, animals have been inoculated by non-natural routes (e.g., subcutaneous, intravenous) that do not recapitulate disease progression via the normative route of mucosal exposure. Accordingly, the natural history of RhCMV would be more accurately reproduced by infecting macaques with strains of RhCMV that reflect the WT genome using natural routes of mucosal transmission. Herein, we tested two WT-like RhCMV strains, UCD52 and UCD59, and demonstrated that systemic infection and frequent, high-titer viral shedding in bodily fluids occurred following oral inoculation. RhCMV disseminated to a broad range of tissues, including the central nervous system and reproductive organs. Commonly infected tissues included the thymus, spleen, lymph nodes, kidneys, bladder, and salivary glands. Histological examination revealed prominent nodular hyperplasia in spleens and variable levels of lymphoid lymphofollicular hyperplasia in lymph nodes. One of six inoculated animals had limited viral dissemination and shedding, with commensurately weak antibody responses to RhCMV antigens. These data suggest that long-term RhCMV infection parameters might be restricted by local innate factors and/or de novo host immune responses in a minority of primary infections. Together, we have established an oral RhCMV infection model that mimics natural HCMV infection. The virological and immunological parameters characterized in this study will greatly inform HCMV vaccine designs for human immunization. IMPORTANCE Human cytomegalovirus (HCMV) is globally ubiquitous with high seroprevalence rates in all communities. HCMV infections can occur vertically following mother-to-fetus transmission across the placenta and horizontally following shedding of virus in bodily fluids in HCMV infected hosts and subsequent exposure of susceptible individuals to virus-laden fluids. Intrauterine HCMV has long been recognized as an infectious threat to fetal growth and development. Since vertical HCMV infections occur following horizontal HCMV transmission to the pregnant mother, the nonhuman primate model of HCMV pathogenesis was used to characterize the virological and immunological parameters of infection following primary mucosal exposures to rhesus cytomegalovirus.
Collapse
|
49
|
Zenebe MH, Mekonnen Z, Loha E, Padalko E. Seroprevalence and associated factors of maternal cytomegalovirus in Southern Ethiopia: a cross-sectional study. BMJ Open 2021; 11:e051390. [PMID: 34675017 PMCID: PMC8532544 DOI: 10.1136/bmjopen-2021-051390] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVES The aim of this study was to assess the seroprevalence and associated factors of cytomegalovirus (CMV) among pregnant women in Southern Ethiopia. DESIGN Cross-sectional study. SETTING The study was conducted in Hawassa University comprehensive and specialised hospital. Hawassa, Southern Ethiopia. PARTICIPANTS A total of 600 consecutive pregnant women attending the delivery ward were recruited for the study from August to October 2020. OUTCOME MEASURES The study assessed the rate of maternal anti-CMV IgG and IgM antibodies. The association of obstetric history, sociodemographic and behavioural characteristics with seropositivity of CMV was also evaluated based on the collected data using structured questioners. RESULTS Seropositivity for CMV IgM antibodies was 8.2% (49/600) (95% CI 6% to 10.5%), whereas the CMV IgG was 88.7% (532/600), (95% CI 89.5% to 94.0%). Seroprevalence of CMV IgM was higher in women of older age, currently unmarried, having nursery schooled children and with any of the detected curable sexually transmitted infections, while seroprevalence of CMV IgG was significantly associated only with women having nursery schooled children. Seroprevalence was not significantly associated with previous adverse pregnancy outcome, gravidity, being a child daycare occupant mother and newborn birth weight. CONCLUSION In the present study, we identified a high rate of CMV IgM and CMV IgG seroprevalence among pregnant women in Southern Ethiopia. Given that there is no existing CMV diagnosis, special attention should be designed to pregnant women in parallel to the existing antenatal care facility. Besides, training healthcare professionals will support awareness conception among pregnant women concerning the sequels of CMV infection during pregnancy.
Collapse
Affiliation(s)
- Mengistu Hailemariam Zenebe
- Medical Laboratory Sciences, Hawassa University College of Medicine and Health Sciences, Hawassa, South Ethiopia, Ethiopia
- Department of Diagnostic Sciences, Ghent University Faculty of Medicine and Health Sciences, Gent, Belgium
- Medical Laboratory Sciences, Jimma University Institute of Health, Jimma, Ethiopia
| | - Zeleke Mekonnen
- Medical Laboratory Sciences, Jimma University Institute of Health, Jimma, Ethiopia
| | - Eskindir Loha
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
- Chr Michelson Institute, Bergen, Norway
| | - Elizaveta Padalko
- Department of Diagnostic Sciences, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
50
|
Weil C, Bilavsky E, Sinha A, Chodick G, Goodman E, Wang WV, Calhoun SR, Marks MA. Epidemiology of cytomegalovirus infection in pregnancy in Israel: Real-world data from a large healthcare organization. J Med Virol 2021; 94:713-719. [PMID: 34665462 DOI: 10.1002/jmv.27403] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 09/30/2021] [Accepted: 10/16/2021] [Indexed: 11/09/2022]
Abstract
Congenital cytomegalovirus infection (cCMVi) is the leading cause of nonhereditary sensorineural hearing loss among newborns. Women newly acquiring cytomegalovirus infection (CMVi) during pregnancy have the highest risk of vertical transmission. This study aimed to describe the epidemiology of CMVi in pregnancy in a large healthcare database. A retrospective cohort study was performed using the Maccabi Healthcare Services database (Israel). Women aged 18-44 years old on July 1, 2013 with no record of pregnancy in the prior 6 months were followed through December 31, 2017 for first pregnancy occurrence. Pregnancy outcomes (live birth, spontaneous/therapeutic abortions, stillbirth, and uncertain outcomes) were captured. CMV test results were obtained to assess serostatus at the start of pregnancy (SoP) and primary CMV infection (CMVi) during pregnancy. Associations of demographic and reproductive factors with pCMVi were investigated (multivariable logistic regression). The study included 84 699 pregnant women (median age = 31 years; interquartile range = 28-35). Live birth, fetal loss, and uncertain pregnancy outcomes accounted for 76.8%, 18.2%, and 5.0%, respectively. The seroprevalence of CMV at the start of pregnancy in this cohort was 63.4% (95% confidence interval [CI]: 63.1-63.7). Among seronegative women with available test results (n = 10 657), CMVi incidence was 14.5 per 1000 (95% CI = 12.2-16.7). In multivariate logistic regression models adjusting for maternal age, CMVi was significantly associated with having one or more prior live births (odds ratio [OR]: 3.8 [95% CI: 2.6-5.4]) and having a child less than 6 years of age (OR: 4.3 [95%CI: 3.0-6.1]). One in three pregnant women in Israel is at risk for primary CMVi. This study demonstrates that real-world electronic healthcare data can be leveraged to support clinical management and development of interventions for congenital CMV by identifying women at high risk for CMVi during pregnancy.
Collapse
Affiliation(s)
- Clara Weil
- Maccabi Institute for Research and Innovation (Maccabitech), Maccabi Healthcare Services, Tel Aviv, Israel
| | - Efraim Bilavsky
- Schneider Children's Medical Center, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Gabriel Chodick
- Maccabi Institute for Research and Innovation (Maccabitech), Maccabi Healthcare Services, Tel Aviv, Israel.,Schneider Children's Medical Center, Petah Tikva, Israel
| | | | | | | | | |
Collapse
|