1
|
Garabedian C, Sibiude J, Anselem O, Attie-Bittach T, Bertholdt C, Blanc J, Dap M, de Mézerac I, Fischer C, Girault A, Guerby P, Le Gouez A, Madar H, Quibel T, Tardy V, Stirnemann J, Vialard F, Vivanti A, Sananès N, Verspyck E. [Fetal death: Expert consensus from the College of French Gynecologists and Obstetricians]. GYNECOLOGIE, OBSTETRIQUE, FERTILITE & SENOLOGIE 2024; 52:549-611. [PMID: 39153884 DOI: 10.1016/j.gofs.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/19/2024]
Abstract
Fetal death is defined as the spontaneous cessation of cardiac activity after fourteen weeks of amenorrhea. In France, the prevalence of fetal death after 22 weeks is between 3.2 and 4.4/1000 births. Regarding the prevention of fetal death in the general population, it is not recommended to counsel for rest and not to prescribe vitamin A, vitamin D nor micronutrient supplementation for the sole purpose of reducing the risk of fetal death (Weak recommendations; Low quality of evidence). It is not recommended to prescribe aspirin (Weak recommendation; Very low quality of evidence). It is recommended to offer vaccination against influenza in epidemic periods and against SARS-CoV-2 (Strong recommendations; Low quality of evidence). It is not recommended to systematically look for nuchal cord encirclements during prenatal screening ultrasounds (Strong Recommendation; Low Quality of Evidence) and not to perform systematic antepartum monitoring by cardiotocography (Weak Recommendation; Very Low Quality of Evidence). It is not recommended to ask women to perform an active fetal movement count to reduce the risk of fetal death (Strong Recommendation; High Quality of Evidence). Regarding evaluation in the event of fetal death, it is suggested that an external fetal examination be systematically offered (Expert opinion). It is recommended that a fetopathological and anatomopathological examination of the placenta be carried out to participate in cause identification (Strong Recommendation. Moderate quality of evidence). It is recommended that chromosomal analysis by microarray testing be performed rather than conventional karyotype, in order to be able to identify a potentially causal anomaly more frequently (Strong Recommendation, moderate quality of evidence); to this end, it is suggested that postnatal sampling of the placental fetal surface for genetic purposes be preferred (Expert Opinion). It is suggested to test for antiphospholipid antibodies and systematically perform a Kleihauer test and a test for irregular agglutinins (Expert opinion). It is suggested to offer a summary consultation, with the aim of assessing the physical and psychological status of the parents, reporting the results, discussing the cause and providing information on monitoring for a subsequent pregnancy (Expert opinion). Regarding announcement and support, it is suggested to announce fetal death without ambiguity, using simple words and adapting to each situation, and then to support couples with empathy in the various stages of their care (Expert opinion). Regarding management, it is suggested that, in the absence of a situation at risk of disseminated intravascular coagulation or maternal vitality, the patient's wishes should be taken into account when determining the time between the diagnosis of fetal death and induction of birth. Returning home is possible if it's the patient wish (Expert opinion). In all situations excluding maternal life-threatening emergencies, the preferred mode of delivery is vaginal delivery, regardless the history of cesarean section(s) history (Expert opinion). In the event of fetal death, it is recommended that mifepristone 200mg be prescribed at least 24hours before induction, to reduce the delay between induction and delivery (Low recommendation. Low quality of evidence). There are insufficient data in the literature to make a recommendation regarding the route of administration (vaginal or oral) of misoprostol, neither the type of prostaglandin to reduce induction-delivery time or maternal morbidity. It is suggested that perimedullary analgesia be introduced at the start of induction if the patient asks, regardless of gestational age. It is suggested to prescribe cabergoline immediately in the postpartum period in order to avoid lactation, whatever the gestational age, after discussing the side effects of the treatment with the patient (Expert opinion). The risk of recurrence of fetal death after unexplained fetal death does not appear to be increased in subsequent pregnancies, and data from the literature are insufficient to make a recommendation on the prescription of aspirin. In the event of a history of fetal death due to vascular issues, low-dose aspirin is recommended to reduce perinatal morbidity, and should not be combined with heparin therapy (Low recommendation, very low quality of evidence). It is suggested not to recommend an optimal delay before initiating another pregnancy just because of the history of fetal death. It is suggested that the woman and co-parent be informed of the possibility of psychological support. Fetal heart rate monitoring is not indicated solely because of a history of fetal death. It is suggested that delivery not be systematically induced. However, induction can be considered depending on the context and parental request. The gestational age will be discussed, taking into account the benefits and risks, especially before 39 weeks. If a cause of fetal death is identified, management will be adapted on a case-by-case basis (expert opinion). In the event of fetal death occurring in a twin pregnancy, it is suggested that the surviving twin be evaluated as soon as the diagnosis of fetal death is made. In the case of dichorionic pregnancy, it is suggested to offer ultrasound monitoring on a monthly basis. It is suggested not to deliver prematurely following fetal death of a twin. If fetal death occurs in a monochorionic twin pregnancy, it is suggested to contact the referral competence center, in order to urgently look for signs of acute fetal anemia on ultrasound in the surviving twin, and to carry out weekly ultrasound monitoring for the first month. It is suggested not to induce birth immediately.
Collapse
Affiliation(s)
| | - Jeanne Sibiude
- Service de gynécologie-obstétrique, hôpital Trousseau, AP-HP, Paris, France
| | - Olivia Anselem
- Maternité Port-Royal, groupe hospitalier Paris Centre, AP-HP, 75014 Paris, France
| | | | - Charline Bertholdt
- Pôle de gynécologie-obstétrique, pôle laboratoires, CHRU de Nancy, université de Lorraine, 54000 Nancy, France
| | - Julie Blanc
- Service de gynécologie-obstétrique, hôpital Nord, hôpitaux universitaires de Marseille, AP-HM, Marseille, France
| | - Matthieu Dap
- Pôle de gynécologie-obstétrique, pôle laboratoires, CHRU de Nancy, université de Lorraine, 54000 Nancy, France
| | | | - Catherine Fischer
- Service d'anesthésie, maternité Port-Royal, groupe hospitalier Paris Centre, AP-HP, Paris, France
| | - Aude Girault
- Maternité Port-Royal, groupe hospitalier Paris Centre, AP-HP, 75014 Paris, France
| | - Paul Guerby
- Service de gynécologie-obstétrique, CHU de Toulouse, Toulouse, France
| | - Agnès Le Gouez
- Service d'anesthésie, hôpital Antoine-Béclère, AP-HP, université Paris Saclay, Clamart, France
| | - Hugo Madar
- Service de gynécologie-obstétrique, CHU de Bordeaux, 33000 Bordeaux, France
| | - Thibaud Quibel
- Service de gynécologie-obstétrique, CHI de Poissy Saint-Germain-en-Laye, Poissy, France
| | - Véronique Tardy
- Direction des plateaux médicotechniques, hospices civils de Lyon, Lyon, France; Département de biochimie biologie moléculaire, université Claude-Bernard Lyon, Lyon, France
| | - Julien Stirnemann
- Service de gynécologie-obstétrique, hôpital Necker, AP-HP, Paris, France
| | - François Vialard
- Département de génétique, CHI de Poissy Saint-Germain-en-Laye, Poissy, France
| | - Alexandre Vivanti
- Service de gynécologie-obstétrique, DMU santé des femmes et des nouveau-nés, hôpital Antoine-Béclère, AP-HP, université Paris Saclay, Clamart, France
| | - Nicolas Sananès
- Service de gynécologie-obstétrique, hôpital américain, Neuilly-sur-Seine, France
| | - Eric Verspyck
- Service de gynécologie-obstétrique, CHU Charles-Nicolle, Rouen, France
| |
Collapse
|
2
|
Rubino G, Yörük E. Immunosenescence, immunotolerance and rejection: clinical aspects in solid organ transplantation. Transpl Immunol 2024; 86:102068. [PMID: 38844001 DOI: 10.1016/j.trim.2024.102068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 05/27/2024] [Accepted: 06/01/2024] [Indexed: 07/21/2024]
Abstract
As a consequence of increased lifespan and rising number of elderly individuals developing end-stage organ disease, the higher demand for organs along with a growing availability for organs from older donors pose new challenges for transplantation. During aging, dynamic adaptations in the functionality and structure of the biological systems occur. Consistently, immunosenescence (IS) accounts for polydysfunctions within the lymphocyte subsets, and the onset of a basal but persistent systemic inflammation characterized by elevated levels of pro-inflammatory mediators. There is an emerging consensus about a causative link between such hallmarks and increased susceptibility to morbidities and mortality, however the role of IS in solid organ transplantation (SOT) remains loosely addressed. Dissecting the immune-architecture of immunologically-privileged sites may prompt novel insights to extend allograft survival. A deeper comprehension of IS in SOT might unveil key standpoints for the clinical management of transplanted patients.
Collapse
Affiliation(s)
- Graziella Rubino
- University Hospital Tübingen, Department of Tropical Medicine, Wilhelmstraße 27, 72074 Tübingen, Germany; Institute for Transfusion Medicine, University Ulm and Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, 89081 Ulm, Germany.
| | - Efdal Yörük
- Berit Klinik, Gastrointestinal Center, Florastrasse 1, 9403 Goldach, Switzerland; University Hospital Tübingen, Department of Ophthalmology, Elfriede-Alhorn-Straße 7, 72076 Tübingen, Germany
| |
Collapse
|
3
|
Pesch MH, Mowers J, Huynh A, Schleiss MR. Intrauterine Fetal Demise, Spontaneous Abortion and Congenital Cytomegalovirus: A Systematic Review of the Incidence and Histopathologic Features. Viruses 2024; 16:1552. [PMID: 39459885 PMCID: PMC11512218 DOI: 10.3390/v16101552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
The objective was to review the existing literature reporting on spontaneous abortion (SA) and intrauterine fetal demise (IUFD) associated with cytomegalovirus (CMV) infection. A review using standardized terminology such as 'intrauterine fetal death', 'congenital cytomegalovirus' and 'CMV' was performed using PubMed and Embase (Medline) using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) methodology. Twenty-one studies met inclusion criteria. CMV was identified as a potential or likely factor in a median of 7.1% of SA or IUFD in study cohorts. Of the studies, 11 used fetal remains, 18 used placenta, 6 used serum, and 1 used post-mortem dried blood spot as specimens for testing for CMV. Features commonly observed were fetal thrombotic vasculopathy, hydrops fetalis and chronic villitis. CMV is frequently noted in studies evaluating viral etiologies of SA or IUFD. Large population-based studies are needed to estimate the incidence of CMV-associated SA or IUFD. CMV and congenital CMV should be included on the differential diagnosis in all cases of SA or IUFD of unknown etiology.
Collapse
Affiliation(s)
- Megan H. Pesch
- Division of Developmental and Behavioral Pediatrics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jonathan Mowers
- Division of Pathology, Ascension Hospital Providence, Southfield, MI 48075, USA;
| | - Anh Huynh
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA;
| | - Mark R. Schleiss
- Division of Pediatric Infectious Diseases, University of Minnesota, Minneapolis, MN 55455, USA;
| |
Collapse
|
4
|
Howard A, Nishikawa JK, Sánchez PJ. "Minimally symptomatic" congenital cytomegalovirus infection: latest data and emerging concepts. Curr Opin Pediatr 2024; 36:480-488. [PMID: 38747205 DOI: 10.1097/mop.0000000000001364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
PURPOSE OF REVIEW Universal and targeted screening of newborns for congenital cytomegalovirus (CMV) infection is increasing globally. Questions remain concerning the management of infants who have been identified with congenital CMV infection, especially those with "minimally symptomatic" or clinically inapparent infection. Our objective is to discuss current management of CMV-infected neonates with a focus on less affected infants with or without sensorineural hearing loss (SNHL). RECENT FINDINGS Valganciclovir is being prescribed increasingly in neonates with congenital CMV infection for improvement in hearing outcomes through 2 years of age. Treatment initiated in the first month of age is recommended for clinically apparent disease. A recent study showed hearing improvement at 18-22 months of age when therapy was initiated at age 1-3 months in infants with clinically inapparent CMV infection and isolated SNHL. SUMMARY Antiviral therapy with either ganciclovir or valganciclovir has shown moderate benefit in prevention of hearing deterioration among infants with clinically apparent CMV infection or isolated SNHL. Sustainability of benefit beyond 2 years of age remains unknown. At present, infants with clinically inapparent CMV infection (normal complete evaluation including hearing) should not receive antiviral therapy. All CMV-infected infants require close audiological and neurodevelopmental follow-up.
Collapse
Affiliation(s)
- Ashley Howard
- Department of Pediatric Infectious Diseases and Immunology, Connecticut Children's Hospital, Hartford, Connecticut
- Department of Pediatrics, University of Connecticut, Farmington, Connecticut
| | - Javier K Nishikawa
- Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Pablo J Sánchez
- Department of Pediatrics, Divisions of Neonatology and Pediatric Infectious Diseases, Nationwide Children's Hospital, Abigail Wexner Research Institute at Nationwide Children's Hospital, Center for Perinatal Research, Ohio Perinatal Research Network, The Ohio State University College of Medicine, Columbus, Ohio, USA
| |
Collapse
|
5
|
Egilmezer E, Hamilton ST, Lauw G, Follett J, Sonntag E, Schütz M, Marschall M, Rawlinson WD. Human Cytomegalovirus Dysregulates Cellular Dual-Specificity Tyrosine Phosphorylation-Regulated Kinases and Sonic Hedgehog Pathway Proteins in Neural Astrocyte and Placental Models. Viruses 2024; 16:918. [PMID: 38932210 PMCID: PMC11209403 DOI: 10.3390/v16060918] [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: 01/12/2024] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
Human cytomegalovirus (CMV) infection is the leading non-genetic cause of congenital malformation in developed countries, causing significant fetal injury, and in some cases fetal death. The pathogenetic mechanisms through which this host-specific virus infects then damages both the placenta and the fetal brain are currently ill-defined. We investigated the CMV modulation of key signaling pathway proteins for these organs including dual-specificity tyrosine phosphorylation-regulated kinases (DYRK) and Sonic Hedgehog (SHH) pathway proteins using human first trimester placental trophoblast (TEV-1) cells, primary human astrocyte (NHA) brain cells, and CMV-infected human placental tissue. Immunofluorescence demonstrated the accumulation and re-localization of SHH proteins in CMV-infected TEV-1 cells with Gli2, Ulk3, and Shh re-localizing to the CMV cytoplasmic virion assembly complex (VAC). In CMV-infected NHA cells, DYRK1A re-localized to the VAC and DYRK1B re-localized to the CMV nuclear replication compartments, and the SHH proteins re-localized with a similar pattern as was observed in TEV-1 cells. Western blot analysis in CMV-infected TEV-1 cells showed the upregulated expression of Rb, Ulk3, and Shh, but not Gli2. In CMV-infected NHA cells, there was an upregulation of DYRK1A, DYRK1B, Gli2, Rb, Ulk3, and Shh. These in vitro monoculture findings are consistent with patterns of protein upregulation and re-localization observed in naturally infected placental tissue and CMV-infected ex vivo placental explant histocultures. This study reveals CMV-induced changes in proteins critical for fetal development, and identifies new potential targets for CMV therapeutic development.
Collapse
Affiliation(s)
- Ece Egilmezer
- Serology and Virology Division, Microbiology, NSW Health Pathology, Prince of Wales Hospital, Sydney 2031, Australia; (E.E.)
- School of Clinical Medicine, University of New South Wales, Kensington 2052, Australia
| | - Stuart T. Hamilton
- Serology and Virology Division, Microbiology, NSW Health Pathology, Prince of Wales Hospital, Sydney 2031, Australia; (E.E.)
- School of Clinical Medicine, University of New South Wales, Kensington 2052, Australia
| | - Glen Lauw
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney 2033, Australia
| | - Jasmine Follett
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney 2033, Australia
| | - Eric Sonntag
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg, 91054 Erlangen, Germany (M.M.)
| | - Martin Schütz
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg, 91054 Erlangen, Germany (M.M.)
| | - Manfred Marschall
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg, 91054 Erlangen, Germany (M.M.)
| | - William D. Rawlinson
- Serology and Virology Division, Microbiology, NSW Health Pathology, Prince of Wales Hospital, Sydney 2031, Australia; (E.E.)
- School of Clinical Medicine, University of New South Wales, Kensington 2052, Australia
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney 2033, Australia
| |
Collapse
|
6
|
Grosse SD, Fleming P, Pesch MH, Rawlinson WD. Estimates of congenital cytomegalovirus-attributable infant mortality in high-income countries: A review. Rev Med Virol 2024; 34:e2502. [PMID: 38282398 PMCID: PMC10878676 DOI: 10.1002/rmv.2502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 11/15/2023] [Accepted: 12/11/2023] [Indexed: 01/30/2024]
Abstract
As many as 5%-10% of infants with symptomatic congenital cytomegalovirus (cCMV) disease, or 0.4%-0.8% of all liveborn infants with cCMV infection, die in early infancy in high-income countries. However, estimates are uncertain due to several potential biases that can result from data limitations and study designs. First, infants with cCMV infections who die prior to diagnosis, which usually occurs at 1-4 weeks after birth, may be excluded from both the count of deaths and the denominator of cCMV births, resulting in left truncation and immortal time biases. These 'biases' are features of the data and do not reflect bias on the part of researchers, but understanding the potential existence of threats to validity can help with interpretation of findings. Left truncation of infant deaths occurring prior to diagnosis of cCMV can result in understatement of the burden of infant deaths due to cCMV. Conversely, overestimation of infant deaths associated with symptomatic cCMV may occur in clinical case series owing to greater representation of relatively severely affected infants owing to ascertainment and referral biases. In this review, we summarise the characteristics of 26 studies that reported estimates of cCMV-associated infant deaths, including potential biases or limitations to which those estimates may have been subject. We discuss study designs whose implementation might generate improved estimates of infant deaths attributable to cCMV. More complete estimates of the overall public health impact of cCMV could inform current and future screening, prevention, and vaccine research.
Collapse
Affiliation(s)
- Scott D. Grosse
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Patrick Fleming
- Frank H. Netter MD School of Medicine, Quinnipiac University, Hamden, Connecticut, USA
| | - Megan H. Pesch
- Division of Developmental and Behavioral Pediatrics, Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - William D. Rawlinson
- Serology and Virology Division, NSW Health Pathology, Prince of Wales Hospital, Sydney, New South Wales, Australia
- School of Biomedical Sciences & School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
7
|
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: 5] [Impact Index Per Article: 5.0] [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
|
8
|
McMillen CM, Boyles DA, Kostadinov SG, Hoehl RM, Schwarz MM, Albe JR, Demers MJ, Hartman AL. Congenital Rift Valley fever in Sprague Dawley rats is associated with diffuse infection and pathology of the placenta. PLoS Negl Trop Dis 2022; 16:e0010898. [PMID: 36315601 PMCID: PMC9648853 DOI: 10.1371/journal.pntd.0010898] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 11/10/2022] [Accepted: 10/17/2022] [Indexed: 11/12/2022] Open
Abstract
Rift Valley fever (RVF) is a disease of animals and humans associated with abortions in ruminants and late-gestation miscarriages in women. Here, we use a rat model of congenital RVF to identify tropisms, pathologies, and immune responses in the placenta during vertical transmission. Infection of late-gestation pregnant rats resulted in vertical transmission to the placenta and widespread infection throughout the decidua, basal zone, and labyrinth zone. Some pups from infected dams appeared normal while others had gross signs of teratogenicity including death. Histopathological lesions were detected in placenta from pups regardless of teratogenicity, while teratogenic pups had widespread hemorrhage throughout multiple placenta layers. Teratogenic events were associated with significant increases in placental pro-inflammatory cytokines, type I interferons, and chemokines. RVFV displays a high degree of tropism for all placental tissue layers and the degree of hemorrhage and inflammatory mediator production is highest in placenta from pups with adverse outcomes. Given the potential for RVFV to emerge in new locations and the recent evidence of emerging viruses, like Zika and SARS-CoV-2, to undergo vertical transmission, this study provides essential understanding regarding the mechanisms by which RVFV crosses the placenta barrier. Rift Valley fever virus (RVFV) infections cause human health and economical burdens given its ability to induce high rates of abortions in ruminants and possible contributions towards late-term miscarriages in women. In this study, we have identified important structures in the placenta targeted by this emerging bunyavirus. Inflammation was associated with more severe fetal outcomes such as death and fetal deformities. The striking similarities between the pathologies of the placenta in the rat model of congenital RVF and those observed in naturally infected ruminants highlight the utility of this rodent model. These findings may be further translated towards understanding the mechanisms involved in vertical transmission of RVFV in humans.
Collapse
Affiliation(s)
- Cynthia M. McMillen
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, United States of America
| | - Devin A. Boyles
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Stefan G. Kostadinov
- Department of Pathology, Magee Women’s Hospital of the University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States of America
| | - Ryan M. Hoehl
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Madeline M. Schwarz
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, United States of America
| | - Joseph R. Albe
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, United States of America
| | - Matthew J. Demers
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Amy L. Hartman
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
9
|
Herpes Simplex Virus Type 2 (HSV-2) and Cytomegalovirus (CMV) among Women with Macerated Stillbirth: A Cross-Sectional Hospital-Based Study from Mwanza, Tanzania. J Pregnancy 2022; 2022:2156835. [PMID: 36120505 PMCID: PMC9477628 DOI: 10.1155/2022/2156835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 08/10/2022] [Accepted: 08/18/2022] [Indexed: 11/18/2022] Open
Abstract
Background Stillbirth adversely affects pregnancy outcomes in low- and middle-income countries (LMICs). Viral infections have been implicated as one of the causes of stillbirths. Despite high rates of stillbirths and high viral prevalence in LMICs, there is limited information regarding their association. This study investigated the magnitude of herpes simplex 2 virus (HSV-2) and human cytomegalovirus (HCMV) among women with macerated stillbirth. Methods A cross-sectional hospital-based study was conducted involving 279 women with macerated stillbirth between July and August 2018 at different health facilities in Mwanza, Tanzania. Detection of HSV-2 was done by immunochromatographic test while that of HCMV was done using enzyme-linked immunosorbent assay (ELISA). Descriptive data analysis was done using STATA version 13. Results A total of 28 (10.04%, 95% CI: 6.8-13.9) tested positive for HSV-2 IgG antibodies with only 4 (1.43%, 95% CL: 0.3-2.8) testing positive for HSV-2 IgM antibodies. HCMV IgG antibodies were detected in 131 (77.98%, 95% CI: 71-84) of 168 women tested. By multivariate logistic regulation analysis, advanced age (OR: 0.93, 95% CI: 0.87-0.99, p = 0.025) was significantly associated with negative HSV-2 IgG antibodies. By log multinomial regression analysis, only urban residence (RRR.4.43: 95% CI 1.53-12.80, p = 0.006) independently predicted HCMV IgG seropositivity among women with stillbirth. Twenty-one (30.9%) of women with positive HCMV IgG antibodies had low avidity index (<40%) indicating recent infection. Conclusion Significant proportion of women with macerated stillbirth residing in urban and with low age have HCMV and HSV antibodies, respectively. This calls for the need to consider introducing screening of these infections in the Tanzanian antenatal package and further studies to explore the role of these viruses in causing stillbirth in Tanzania.
Collapse
|
10
|
Abstract
Over a century of research has focused on improving our understanding of congenital cytomegalovirus (cCMV), yet it remains the most common congenital infection in the United States, affecting 3 to 6 per 1000 live born infants each year. Pregnancies affected by cCMV are at a heightened risk of spontaneous abortion and intrauterine fetal demise. Neonates born with cCMV are also at substantial risk for long-term neurodevelopmental sequelae and disability, including sensorineural hearing loss, even those born without clinically apparent disease. Considerable progress has been made in recent years in study of the epidemiology and transmission of cCMV, developing better diagnostic strategies, implementing newborn screening programs, improving therapeutics, and launching vaccine trials. In this article, we review recent developments in the understanding of the virology and immunobiology of cytomegalovirus. We further discuss how this knowledge informs our understanding of the pathophysiology of cCMV and directs strategies aimed at improving outcomes and quality of life for congenitally infected children. We also provide an update on the epidemiology of cCMV in the United States, evolving scientific understanding of maternal-fetal transmission, enhanced screening approaches, and recognition of neonatal and long-term sequelae. Finally, we review the current landscape of pediatric cCMV research and provide recommendations for novel and high-priority areas for future investigation.
Collapse
Affiliation(s)
- Megan H. Pesch
- University of Michigan and CS Mott Children’s Hospital, Division of Developmental and Behavioral Pediatrics, Department of Pediatrics, Ann Arbor, Michigan
| | - Mark R. Schleiss
- Department of Pediatrics, University of Minnesota Medical School, Division of Pediatric Infectious Diseases
| |
Collapse
|
11
|
Tsakiridis I, Giouleka S, Mamopoulos A, Athanasiadis A, Dagklis T. Investigation and management of stillbirth: a descriptive review of major guidelines. J Perinat Med 2022; 50:796-813. [PMID: 35213798 DOI: 10.1515/jpm-2021-0403] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 02/01/2022] [Indexed: 11/15/2022]
Abstract
Stillbirth is a common and devastating pregnancy complication. The aim of this study was to review and compare the recommendations of the most recently published guidelines on the investigation and management of this adverse outcome. A descriptive review of guidelines from the American College of Obstetricians and Gynecologists (ACOG), the Royal College of Obstetricians and Gynecologists (RCOG), the Perinatal Society of Australia and New Zealand (PSANZ), the Society of Obstetricians and Gynecologists of Canada (SOGC) on stillbirth was carried out. Regarding investigation, there is consensus that medical history and postmortem examination are crucial and that determining the etiology may improve care in a subsequent pregnancy. All guidelines recommend histopathological examination of the placenta, genetic analysis and microbiology of fetal and placental tissues, offering less invasive techniques when autopsy is declined and a Kleihauer test to detect large feto-maternal hemorrhage, whereas they discourage routine screening for inherited thrombophilias. RCOG and SOGC also recommend a complete blood count, coagulopathies' testing, anti-Ro and anti-La antibodies' measurement in cases of hydrops and parental karyotyping. Discrepancies exist among the reviewed guidelines on the definition of stillbirth and the usefulness of thyroid function tests and maternal viral screening. Moreover, only ACOG and RCOG discuss the management of stillbirth. They agree that, in the absence of coagulopathies, expectant management should be considered and encourage vaginal birth, but they suggest different labor induction protocols and different management in subsequent pregnancies. It is important to develop consistent international practice protocols, in order to allow effective determination of the underlying causes and optimal management of stillbirths, while identifying the gaps in the current literature may highlight the need for future research.
Collapse
Affiliation(s)
- Ioannis Tsakiridis
- Third Department of Obstetrics and Gynaecology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Sonia Giouleka
- Third Department of Obstetrics and Gynaecology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Apostolos Mamopoulos
- Third Department of Obstetrics and Gynaecology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Apostolos Athanasiadis
- Third Department of Obstetrics and Gynaecology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Themistoklis Dagklis
- Third Department of Obstetrics and Gynaecology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
12
|
Oumarou Hama H, Aboudharam G, Barbieri R, Lepidi H, Drancourt M. Immunohistochemical diagnosis of human infectious diseases: a review. Diagn Pathol 2022; 17:17. [PMID: 35094696 PMCID: PMC8801197 DOI: 10.1186/s13000-022-01197-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 01/18/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Immunohistochemistry (IHC) using monoclonal and polyclonal antibodies is a useful diagnostic method for detecting pathogen antigens in fixed tissues, complementing the direct diagnosis of infectious diseases by PCR and culture on fresh tissues. It was first implemented in a seminal publication by Albert Coons in 1941. MAIN BODY Of 14,198 publications retrieved from the PubMed, Google, Google Scholar and Science Direct databases up to December 2021, 230 were selected for a review of IHC techniques, protocols and results. The methodological evolutions of IHC and its application to the diagnosis of infectious diseases, more specifically lice-borne diseases, sexually transmitted diseases and skin infections, were critically examined. A total of 59 different pathogens have been detected once in 22 different tissues and organs; and yet non-cultured, fastidious and intracellular pathogens accounted for the vast majority of pathogens detected by IHC. Auto-IHC, incorporating patient serum as the primary antibody, applied to diseased heart valves surgically collected from blood culture-negative endocarditis patients, detected unidentified Gram-positive cocci and microorganisms which were subsequently identified as Coxiella burnetii, Bartonella quintana, Bartonella henselae and Tropheryma whipplei. The application of IHC to ancient tissues dated between the ends of the Ptolemaic period to over 70 years ago, have also contributed to paleomicrobiology diagnoses. CONCLUSION IHC plays an important role in diagnostic of infectious diseases in tissue samples. Paleo-auto-IHC derived from auto-IHC, is under development for detecting non-identified pathogens from ancient specimens.
Collapse
Affiliation(s)
- Hamadou Oumarou Hama
- IHU Méditerranée Infection, Marseille, France
- Aix-Marseille-Univ., IRD, MEPHI, IHU Méditerranée Infection, Marseille, France
| | - Gérard Aboudharam
- Aix-Marseille-Univ., IRD, MEPHI, IHU Méditerranée Infection, Marseille, France
- Aix-Marseille-Univ., Ecole de Médecine Dentaire, Marseille, France
| | - Rémi Barbieri
- IHU Méditerranée Infection, Marseille, France
- Aix-Marseille-Univ., IRD, MEPHI, IHU Méditerranée Infection, Marseille, France
| | - Hubert Lepidi
- Aix-Marseille-Univ., IRD, MEPHI, IHU Méditerranée Infection, Marseille, France
- Laboratoire d'Histologie, Faculté de Médecine, Université de la Méditerranée, Marseille, France
| | - Michel Drancourt
- IHU Méditerranée Infection, Marseille, France.
- Aix-Marseille-Univ., IRD, MEPHI, IHU Méditerranée Infection, Marseille, France.
| |
Collapse
|
13
|
Huang Z, Xia S, Mei S, Wen Y, Liu J, Dong C, Chen W, Yu P, Qu L, Luo Y, Zheng L. Integrated Analysis Reveals the Characteristics and Effects of SARS-CoV-2 Maternal–Fetal Transmission. Front Microbiol 2022; 13:813187. [PMID: 35154056 PMCID: PMC8828581 DOI: 10.3389/fmicb.2022.813187] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 01/04/2022] [Indexed: 01/14/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has caused a pandemic of coronavirus disease 2019 (COVID-19) and is threatening global health. SARS-CoV-2 spreads by air with a transmission rate of up to 15%, but the probability of its maternal–fetal transmission through the placenta is reported to be low at around 3.28%. However, it is still unclear that which tissues and developmental periods hold higher risks and what the underlying molecular mechanisms are. We conducted an integrated analysis of large-scale transcriptome and single-cell sequencing data to investigate the key factors that affect SARS-CoV-2 maternal–fetal transmission as well as the characteristics and effects of them. Our results showed that the abundance of cytomegalovirus (CMV) and Zika virus (ZIKV) infection-associated factors in the placenta were higher than their primarily infected tissues, while the expression levels of SARS-CoV-2 binding receptor angiotensin-converting enzyme II (ACE2) were similar between lung and placenta. By contrast, an important SARS-CoV-2 infection-associated factor, type II transmembrane serine protease (TMPRSS2), was poorly expressed in placenta. Further scRNA-Seq analysis revealed that ACE2 and TMPRSS2 were co-expressed in very few trophoblastic cells. Interestingly, during the embryonic development stages, the abundance of ACE2 and TMPRSS2 was much higher in multiple embryonic tissues than in the placenta. Based on our present analysis, the intestine in 20th week of embryonic development was at a high risk of SARS-CoV-2 infection. Additionally, we found that during the fetal development, ACE2 and TMPRSS2 were enriched in pathogen infection-associated pathways and may involve in the biological processes related to T-cell activation. In conclusion, our present study suggests that though the placenta provides a good physical barrier against SARS-CoV-2 infection for healthy fetal development, multiple embryonic tissues are under risks of the virus infection, which may be adversely affected once infected prenatally. Therefore, it is necessary to enhance maternal care to prevent the potential impact and harm of SARS-CoV-2 maternal–fetal transmission.
Collapse
Affiliation(s)
- Ziliang Huang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Shuting Xia
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shiqiang Mei
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yanzi Wen
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jialiu Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Chengzhi Dong
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Wenxin Chen
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Peijie Yu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Lianghu Qu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yanmin Luo
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Yanmin Luo,
| | - Lingling Zheng
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Lingling Zheng,
| |
Collapse
|
14
|
Glynn SM, Yang YJ, Thomas C, Friedlander RL, Cagino KA, Matthews KC, Riley LE, Baergen RN, Prabhu M. SARS-CoV-2 and Placental Pathology: Malperfusion Patterns Are Dependent on Timing of Infection During Pregnancy. Am J Surg Pathol 2022; 46:51-57. [PMID: 34310367 PMCID: PMC8662940 DOI: 10.1097/pas.0000000000001772] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The extent to which severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection at different points in the pregnancy timeline may affect maternal and fetal outcomes remains unknown. We sought to characterize the impact of SARS-CoV-2 infection proximate and remote from delivery on placental pathology. We performed a secondary analysis of placental pathology from a prospective cohort of universally tested SARS-CoV-2 positive women >20 weeks gestation at 1 institution. Subjects were categorized as having acute or nonacute SARS-CoV-2 based on infection <14 or ≥14 days from delivery admission, respectively, determined by nasopharyngeal swab, symptom history, and serologies, when available. A subset of SARS-CoV-2 negative women represented negative controls. Placental pathology was available for 90/97 (92.8%) of SARS-CoV-2 positive women, of which 26 were from women with acute SARS-CoV-2 infection and 64 were from women with nonacute SARS-CoV-2. Fetal vascular malperfusion lesions were significantly more frequent among the acute SARS-CoV-2 group compared with the nonacute SARS-CoV-2 group (53.8% vs. 18.8%; P=0.002), while frequency of maternal vascular malperfusion lesions did not differ by timing of infection (30.8% vs. 29.7%; P>0.99). When including 188 SARS-CoV-2 negative placentas, significant differences in frequency of fetal vascular malperfusion lesions remained between acute, nonacute and control cases (53.8% vs. 18.8% vs. 13.2%, respectively; P<0.001). No differences were noted in obstetric or neonatal outcomes between acutely and nonacutely infected women. Our findings indicate timing of infection in relation to delivery may alter placental pathology, with potential clinical implications for risk of thromboembolic events and impact on fetal health.
Collapse
Affiliation(s)
| | - Yawei J. Yang
- Department of Pathology and Laboratory Medicine, New York-Presbyterian Hospital/Weill Cornell Medicine
| | | | | | | | | | - Laura E. Riley
- Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY
| | - Rebecca N. Baergen
- Department of Pathology and Laboratory Medicine, New York-Presbyterian Hospital/Weill Cornell Medicine
| | - Malavika Prabhu
- Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY
| |
Collapse
|
15
|
Song X, Li Q, Diao J, Li J, Li Y, Zhang S, Chen L, Wei J, Shu J, Liu Y, Sun M, Sheng X, Wang T, Qin J. Association Between First-Trimester Maternal Cytomegalovirus Infection and Stillbirth: A Prospective Cohort Study. Front Pediatr 2022; 10:803568. [PMID: 35372174 PMCID: PMC8970618 DOI: 10.3389/fped.2022.803568] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 02/23/2022] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Given that the time lag between cytomegalovirus (CMV) screening and diagnosed testing, a better knowledge of the association between pregnant women with CMV screening test positive and stillbirth in an epidemiological perspective was required to assist people being counseled reframe their pregnancy and birth plans based on the magnitude of the risk. METHODS This study recruited 44048 eligible pregnant women from March 13, 2013 to December 31, 2019. Serological tests including CMV-specific IgM and IgG, and IgG avidity index were used to screen for maternal CMV infection and were measured by automated chemiluminescence immunoassay. The association was assessed using the inverse probability of group-weighted multivariate-adjusted log-binomial models. RESULTS A total of 540 infants ended with a stillbirth (12.3 per 1000 pregnancies), and 2472 pregnancies with maternal CMV infection were screened out (56.1 per 1000 pregnancies) among all eligible pregnancies. In the comparison analysis, 326 infants ended with a stillbirth (86.6 per 1000 pregnancies) in the maternal CMV infection group compared with 214 infants (7.8 per 1000 pregnancies) in the group where mothers were not infected with CMV (RR 12.17; 95% CI 9.43-15.71). After excluding the pregnancies of stillbirth with birth defects, a strong association between the two groups was still observed (RR 9.38; 95% CI 6.92-12.70). CONCLUSION Our findings quantified the risk of a woman having a baby with stillbirth if she had a positive serologic CMV screening test in her first trimester, and supported the value of using CMV serologic tests as part of regular testing in pregnant women. TRIAL REGISTRATION Registered in Chinese Clinical Trial Registry Center; registration number, ChiCTR1800016635; registration date, 06/14/2018 (Retrospectively registered); URL of trial registry record, https://www.chictr.org.cn/showproj.aspx?proj=28300.
Collapse
Affiliation(s)
- Xinli Song
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Qiongxuan Li
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Jingyi Diao
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Jinqi Li
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Yihuan Li
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Senmao Zhang
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Letao Chen
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Jianhui Wei
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Jing Shu
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Yiping Liu
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Mengting Sun
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Xiaoqi Sheng
- National Health Committee (NHC) Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China
| | - Tingting Wang
- National Health Committee (NHC) Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China
| | - Jiabi Qin
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China.,National Health Committee (NHC) Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China.,Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Hunan Provincial Key Laboratory of Clinical Epidemiology, Changsha, China
| |
Collapse
|
16
|
Biology and pathology of the uterine microenvironment and its natural killer cells. Cell Mol Immunol 2021; 18:2101-2113. [PMID: 34426671 PMCID: PMC8429689 DOI: 10.1038/s41423-021-00739-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/12/2021] [Indexed: 02/07/2023] Open
Abstract
Tissues are the new frontier of discoveries in immunology. Cells of the immune system are an integral part of tissue physiology and immunity. Determining how immune cells inhabit, housekeep, and defend gut, lung, brain, liver, uterus, and other organs helps revealing the intimate details of tissue physiology and may offer new therapeutic targets to treat pathologies. The uterine microenvironment modulates the development and function of innate lymphoid cells [ILC, largely represented by natural killer (NK) cells], macrophages, T cells, and dendritic cells. These immune cells, in turn, contribute to tissue homeostasis. Regulated by ovarian hormones, the human uterine mucosa (endometrium) undergoes ~400 monthly cycles of breakdown and regeneration from menarche to menopause, with its fibroblasts, glands, blood vessels, and immune cells remodeling the tissue into the transient decidua. Even more transformative changes occur upon blastocyst implantation. Before the placenta is formed, the endometrial glands feed the embryo by histiotrophic nutrition while the uterine spiral arteries are stripped of their endothelial layer and smooth muscle actin. This arterial remodeling is carried out by invading fetal trophoblast and maternal immune cells, chiefly uterine NK (uNK) cells, which also assist fetal growth. The transformed arteries no longer respond to maternal stimuli and meet the increasing demands of the growing fetus. This review focuses on how the everchanging uterine microenvironment affects uNK cells and how uNK cells regulate homeostasis of the decidua, placenta development, and fetal growth. Determining these pathways will help understand the causes of major pregnancy complications.
Collapse
|
17
|
Tomac J, Mazor M, Lisnić B, Golemac M, Kveštak D, Bralić M, Bilić Zulle L, Brinkmann MM, Dölken L, Reinert LS, Paludan SR, Krmpotić A, Jonjić S, Juranić Lisnić V. Viral infection of the ovaries compromises pregnancy and reveals innate immune mechanisms protecting fertility. Immunity 2021; 54:1478-1493.e6. [PMID: 34015257 DOI: 10.1016/j.immuni.2021.04.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 11/29/2020] [Accepted: 04/16/2021] [Indexed: 12/16/2022]
Abstract
Viral infections during pregnancy are a considerable cause of adverse outcomes and birth defects, and the underlying mechanisms are poorly understood. Among those, cytomegalovirus (CMV) infection stands out as the most common intrauterine infection in humans, putatively causing early pregnancy loss. We employed murine CMV as a model to study the consequences of viral infection on pregnancy outcome and fertility maintenance. Even though pregnant mice successfully controlled CMV infection, we observed highly selective, strong infection of corpus luteum (CL) cells in their ovaries. High infection densities indicated complete failure of immune control in CL cells, resulting in progesterone insufficiency and pregnancy loss. An abundance of gap junctions, absence of vasculature, strong type I interferon (IFN) responses, and interaction of innate immune cells fully protected the ovarian follicles from viral infection. Our work provides fundamental insights into the effect of CMV infection on pregnancy loss and mechanisms protecting fertility.
Collapse
Affiliation(s)
- Jelena Tomac
- Department of Histology and Embryology, University of Rijeka, Faculty of Medicine, B. Branchetta 20, 51000 Rijeka, Croatia
| | - Marija Mazor
- Center for Proteomics, University of Rijeka, Faculty of Medicine, B. Branchetta 20, 51000 Rijeka, Croatia
| | - Berislav Lisnić
- Department of Histology and Embryology, University of Rijeka, Faculty of Medicine, B. Branchetta 20, 51000 Rijeka, Croatia; Center for Proteomics, University of Rijeka, Faculty of Medicine, B. Branchetta 20, 51000 Rijeka, Croatia
| | - Mijo Golemac
- Department of Histology and Embryology, University of Rijeka, Faculty of Medicine, B. Branchetta 20, 51000 Rijeka, Croatia
| | - Daria Kveštak
- Department of Histology and Embryology, University of Rijeka, Faculty of Medicine, B. Branchetta 20, 51000 Rijeka, Croatia
| | - Marina Bralić
- Department of Histology and Embryology, University of Rijeka, Faculty of Medicine, B. Branchetta 20, 51000 Rijeka, Croatia
| | - Lidija Bilić Zulle
- Clinical Hospital Rijeka, University of Rijeka, B. Branchetta 20, 51000 Rijeka, Croatia
| | - Melanie M Brinkmann
- Viral Immune Modulation Research Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; Institute of Genetics, Technische Universität Braunschweig, 38106 Braunschweig, Germany
| | - Lars Dölken
- Institute for Virology and Immunobiology, Julius-Maximilians-Universität Würzburg, 97080 Würzburg, Germany; Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Centre for Infection Research, 97080 Würzburg, Germany
| | - Line S Reinert
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Soren R Paludan
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Astrid Krmpotić
- Department of Histology and Embryology, University of Rijeka, Faculty of Medicine, B. Branchetta 20, 51000 Rijeka, Croatia
| | - Stipan Jonjić
- Department of Histology and Embryology, University of Rijeka, Faculty of Medicine, B. Branchetta 20, 51000 Rijeka, Croatia; Center for Proteomics, University of Rijeka, Faculty of Medicine, B. Branchetta 20, 51000 Rijeka, Croatia.
| | - Vanda Juranić Lisnić
- Department of Histology and Embryology, University of Rijeka, Faculty of Medicine, B. Branchetta 20, 51000 Rijeka, Croatia; Center for Proteomics, University of Rijeka, Faculty of Medicine, B. Branchetta 20, 51000 Rijeka, Croatia.
| |
Collapse
|
18
|
Adibi JJ, Layden AJ, Birru RL, Miragaia A, Xun X, Smith MC, Yin Q, Millenson ME, O’Connor TG, Barrett ES, Snyder NW, Peddada S, Mitchell RT. First trimester mechanisms of gestational sac placental and foetal teratogenicity: a framework for birth cohort studies. Hum Reprod Update 2021; 27:747-770. [PMID: 33675653 PMCID: PMC8222765 DOI: 10.1093/humupd/dmaa063] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 12/18/2020] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND The function of the gestational sac (GS) and the placenta in the closely related processes of embryogenesis and teratogenicity in the first trimester has been minimally described. The prevailing assumption is that direct teratogenic effects are mediated by the critical extraembryonic organ, the placenta, which either blocks or transfers exposures to the foetus. Placental transfer is a dominant mechanism, but there are other paradigms by which the placenta can mediate teratogenic effects. Knowledge of these paradigms and first trimester human developmental biology can be useful to the epidemiologist in the conduct of biomarker-based studies of both maternal and child health. OBJECTIVE AND RATIONALE Our aim is to provide a causal framework for modelling the teratogenic effects of first trimester exposures on child health outcomes mediated by the GS and placenta using biomarker data collected in the first trimester. We initially present first trimester human developmental biology for the sake of informing and strengthening epidemiologic approaches. We then propose analytic approaches of modelling placental mechanisms by way of causal diagrams using classical non-embryolethal teratogens (diethylstilboestrol [DES], folic acid deficiency and cytomegalovirus [CMV]) as illustrative examples. We extend this framework to two chronic exposures of particular current interest, phthalates and maternal adiposity. SEARCH METHODS Information on teratogens was identified by a non-systematic, narrative review. For each teratogen, we included papers that answered the five following questions: (i) why were these exposures declared teratogens? (ii) is there a consensus on biologic mechanism? (iii) is there reported evidence of a placental mechanism? (iv) can we construct a theoretical model of a placental mechanism? and (v) can this knowledge inform future work on measurement and modelling of placental-foetal teratogenesis? We prioritized literature specific to human development, the organogenesis window in the first trimester and non-embryolethal mechanisms. OUTCOMES As a result of our review of the literature on five exposures considered harmful in the first trimester, we developed four analytic strategies to address first trimester placental mechanisms in birth cohort studies: placental transfer and direct effects on the foetus (DES and maternal adiposity), indirect effects through targeted placental molecular pathways (DES and phthalates), pre-placental effects through disruptions in embryonic and extraembryonic tissue layer differentiation (folic acid deficiency), and multi-step mechanisms that involve maternal, placental and foetal immune function and inflammation (DES and CMV). WIDER IMPLICATIONS The significance of this review is to offer a causal approach to classify the large number of potentially harmful exposures in pregnancy when the exposure occurs in the first trimester. Our review will facilitate future research by advancing knowledge of the first trimester mechanisms necessary for researchers to effectively associate environmental exposures with child health outcomes.
Collapse
Affiliation(s)
- Jennifer J Adibi
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alexander J Layden
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rahel L Birru
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alexandra Miragaia
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiaoshuang Xun
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Megan C Smith
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Qing Yin
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Thomas G O’Connor
- Department of Psychiatry, University of Rochester Medical Center, Rochester, NY, USA
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
- Department of Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, NY, USA
| | - Emily S Barrett
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ, USA
| | - Nathaniel W Snyder
- Department of Microbiology and Immunology, Center for Metabolic Disease Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Shyamal Peddada
- Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rod T Mitchell
- MRC Centre for Reproductive Health, The University of Edinburgh, Queens Medical Research Institute, Edinburgh, UK
| |
Collapse
|
19
|
Berkebile ZW, Putri DS, Abrahante JE, Seelig DM, Schleiss MR, Bierle CJ. The Placental Response to Guinea Pig Cytomegalovirus Depends Upon the Timing of Maternal Infection. Front Immunol 2021; 12:686415. [PMID: 34211475 PMCID: PMC8239309 DOI: 10.3389/fimmu.2021.686415] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 05/25/2021] [Indexed: 11/16/2022] Open
Abstract
Human cytomegalovirus (HCMV) infects the placenta, and these placental infections can cause fetal injury and/or demise. The timing of maternal HCMV infection during pregnancy is a determinant of fetal outcomes, but how development affects the placenta's susceptibility to infection, the likelihood of placental injury post-infection, and the frequency of transplacental HCMV transmission remains unclear. In this study, guinea pig cytomegalovirus (GPCMV) was used to model primary maternal infection and compare the effects of infection at two different times on the placenta. When guinea pigs were infected with GPCMV at either 21- or 35-days gestation (dGA), maternal and placental viral loads, as determined by droplet digital PCR, were not significantly affected by the timing of maternal infection. However, when the transcriptomes of gestational age-matched GPCMV-infected and control placentas were compared, significant infection-associated changes in gene expression were only observed after maternal infection at 35 dGA. Notably, transcripts associated with immune activation (e.g. Cxcl10, Ido1, Tgtp1, and Tlr8) were upregulated in the infected placenta. A GPCMV-specific in situ hybridization assay detected rare infected cells in the main placenta after maternal infection at either time, and maternal infection at 35 dGA also caused large areas of GPCMV-infected cells in the junctional zone. As GPCMV infection after mid-gestation is known to cause high rates of stillbirth and/or fetal growth restriction, our results suggest that the placenta becomes sensitized to infection-associated injury late in gestation, conferring an increased risk of adverse pregnancy outcomes after cytomegalovirus infection.
Collapse
Affiliation(s)
- Zachary W. Berkebile
- Department of Pediatrics, Division of Pediatric Infectious Diseases, University of Minnesota, Minneapolis, MN, United States
| | - Dira S. Putri
- Department of Pediatrics, Division of Pediatric Infectious Diseases, University of Minnesota, Minneapolis, MN, United States
| | - Juan E. Abrahante
- Informatics Institute, University of Minnesota, Minneapolis, MN, United States
| | - Davis M. Seelig
- Department of Veterinary Clinical Sciences, University of Minnesota, Minneapolis, MN, United States
| | - Mark R. Schleiss
- Department of Pediatrics, Division of Pediatric Infectious Diseases, University of Minnesota, Minneapolis, MN, United States
| | - Craig J. Bierle
- Department of Pediatrics, Division of Pediatric Infectious Diseases, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
20
|
Ríos-Barnés M, Fortuny C, Alarcón A, Noguera-Julian A. Renal Involvement in Congenital Cytomegalovirus Infection: A Systematic Review. Microorganisms 2021; 9:1304. [PMID: 34203932 PMCID: PMC8232607 DOI: 10.3390/microorganisms9061304] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/03/2021] [Accepted: 06/12/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Congenital cytomegalovirus (cCMV) infection is the most frequent mother-to-child transmitted infection worldwide and a prevalent cause of neonatal disease and long-term morbidity. The kidney is a target organ for CMV, which replicates in renal tubules and is excreted in large quantities in urine for years in children with cCMV infection. Nonetheless, kidney disease has rarely been reported in cCMV-infected patients. OBJECTIVE We aimed to describe the available data on renal involvement in patients with cCMV infection at the pathologic, functional, anatomical, and/or clinical levels. METHODS A systematic search was performed in the MEDLINE/PubMed, SCOPUS, and Cochrane databases. Studies describing any renal involvement in fetuses or neonates aged ≤3 weeks at diagnosis of microbiologically confirmed cCMV infection were eligible. RESULTS Twenty-four articles were included, with a very low level of evidence. Pathologic findings in autopsy studies universally described CMV typical inclusion bodies in tubular cells. No functional studies were identified. cCMV infection was not associated with an increased risk of kidney malformations. Congenital nephrotic syndrome was the most common clinical condition associated with cCMV, but a causal relationship cannot be established. CONCLUSIONS Typical pathological features of cCMV infection are very common in renal tissue, but they do not seem to entail significant consequences at the anatomical or clinical levels.
Collapse
Affiliation(s)
- María Ríos-Barnés
- Malalties Infeccioses i Resposta Inflamatòria Sistèmica en Pediatria, Unitat d’Infeccions, Servei de Pediatria, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain; (M.R.-B.); (C.F.)
| | - Clàudia Fortuny
- Malalties Infeccioses i Resposta Inflamatòria Sistèmica en Pediatria, Unitat d’Infeccions, Servei de Pediatria, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain; (M.R.-B.); (C.F.)
- Departament de Pediatria, Universitat de Barcelona, 08950 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
- Red de Investigación Translacional en Infectología Pediátrica (RITIP), 28046 Madrid, Spain
| | - Ana Alarcón
- Departament de Pediatria, Universitat de Barcelona, 08950 Barcelona, Spain;
- Cervell Neonatal, Servei de Neonatologia, Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | - Antoni Noguera-Julian
- Malalties Infeccioses i Resposta Inflamatòria Sistèmica en Pediatria, Unitat d’Infeccions, Servei de Pediatria, Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain; (M.R.-B.); (C.F.)
- Departament de Pediatria, Universitat de Barcelona, 08950 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
- Red de Investigación Translacional en Infectología Pediátrica (RITIP), 28046 Madrid, Spain
| |
Collapse
|
21
|
Abstract
OBJECTIVES To provide an investigation protocol to help health care providers determine the cause of a fetal death. OPTIONS Consideration has been given to protocols for the investigation of fetal death that are currently available in Canada and in other countries. OUTCOMES Identification of possible causes of stillbirth and their relationship to future pregnancies. EVIDENCE Articles related to the etiology of fetal death were identified in a search of PubMed (June 2006 to September 2018), the Cochrane Library, and investigation protocols from the American College of Obstetricians and Gynecologists, the International Stillbirth Alliance Collaborative for Improving Classification of Perinatal Deaths, the Royal College of Obstetricians and Gynaecologists, the Queensland clinical guidelines, and the Reproductive Care Program of Nova Scotia. BENEFITS To provide better advice for women regarding possible causes of fetal death and implications for future pregnancies. VALIDATION The evidence obtained was reviewed and evaluated by the Maternal-Fetal Medicine Committee and the Clinical Practice Obstetrics Committee of the Society of Obstetricians and Gynaecologists of Canada. The level of evidence and quality of the recommendation made was described using the Evaluation of Evidence criteria of the Canadian Task Force on Preventive Health Care. RECOMMENDATIONS
Collapse
|
22
|
Bengtsson BOS, van Houten JP. Severe Intrauterine Growth Restriction, Thrombocytopenia, and Direct Hyperbilirubinemia in a 26-week Premature Infant. Neoreviews 2021; 22:e68-e72. [PMID: 33386318 DOI: 10.1542/neo.22-1-e68] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Bengt-Ola S Bengtsson
- Pediatrix Medical Group, Department of Pediatrics, Neonatal Intensive Care Unit, Community Memorial Hospital of San Buenaventura, Ventura, CA.,Western University of Health Sciences, College of Osteopathic Medicine of the Pacific, Pomona, CA
| | - John P van Houten
- Pediatrix Medical Group, Department of Pediatrics, Neonatal Intensive Care Unit, Community Memorial Hospital of San Buenaventura, Ventura, CA.,Western University of Health Sciences, College of Osteopathic Medicine of the Pacific, Pomona, CA
| |
Collapse
|
23
|
Boppana SB, Britt WJ. Recent Approaches and Strategies in the Generation of Anti-human Cytomegalovirus Vaccines. Methods Mol Biol 2021; 2244:403-463. [PMID: 33555597 DOI: 10.1007/978-1-0716-1111-1_19] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Human cytomegalovirus is the largest human herpesvirus and shares many core features of other herpesviruses such as tightly regulated gene expression during genome replication and latency as well as the establishment of lifelong persistence following infection. In contrast to stereotypic clinical syndromes associated with alpha-herpesvirus infections, almost all primary HCMV infections are asymptomatic and acquired early in life in most populations in the world. Although asymptomatic in most individuals, HCMV is a major cause of disease in hosts with deficits in adaptive and innate immunity such as infants who are infected in utero and allograft recipients following transplantation. Congenital HCMV is a commonly acquired infection in the developing fetus that can result in a number of neurodevelopmental abnormalities. Similarly, HCMV is a major cause of disease in allograft recipients in the immediate and late posttransplant period and is thought to be a major contributor to chronic allograft rejection. Even though HCMV induces robust innate and adaptive immune responses, it also encodes a vast array of immune evasion functions that are thought aid in its persistence. Immune correlates of protective immunity that prevent or modify intrauterine HCMV infection remain incompletely defined but are thought to consist primarily of adaptive responses in the pregnant mother, thus making congenital HCMV a potentially vaccine modifiable disease. Similarly, HCMV infection in allograft recipients is often more severe in recipients without preexisting adaptive immunity to HCMV. Thus, there has been a considerable effort to modify HCMV specific immunity in transplant recipient either through active immunization or passive transfer of adaptive effector functions. Although efforts to develop an efficacious vaccine and/or passive immunotherapy to limit HCMV disease have been underway for nearly six decades, most have met with limited success at best. In contrast to previous efforts, current HCMV vaccine development has relied on observations of unique properties of HCMV in hopes of reproducing immune responses that at a minimum will be similar to that following natural infection. However, more recent findings have suggested that immunity following naturally acquired HCMV infection may have limited protective activity and almost certainly, is not sterilizing. Such observations suggest that either the induction of natural immunity must be specifically tailored to generate protective activity or alternatively, that providing targeted passive immunity to susceptible populations could be prove to be more efficacious.
Collapse
Affiliation(s)
- Suresh B Boppana
- Departments of Pediatrics, The University of Alabama at Birmingham, Birmingham, AL, USA.,Departments of Microbiology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - William J Britt
- Departments of Pediatrics, The University of Alabama at Birmingham, Birmingham, AL, USA. .,Departments of Microbiology, The University of Alabama at Birmingham, Birmingham, AL, USA. .,Departments of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
24
|
Fisher MA, Lloyd ML. A Review of Murine Cytomegalovirus as a Model for Human Cytomegalovirus Disease-Do Mice Lie? Int J Mol Sci 2020; 22:ijms22010214. [PMID: 33379272 PMCID: PMC7795257 DOI: 10.3390/ijms22010214] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 12/10/2020] [Accepted: 12/17/2020] [Indexed: 12/12/2022] Open
Abstract
Since murine cytomegalovirus (MCMV) was first described in 1954, it has been used to model human cytomegalovirus (HCMV) diseases. MCMV is a natural pathogen of mice that is present in wild mice populations and has been associated with diseases such as myocarditis. The species-specific nature of HCMV restricts most research to cell culture-based studies or to the investigation of non-invasive clinical samples, which may not be ideal for the study of disseminated disease. Initial MCMV research used a salivary gland-propagated virus administered via different routes of inoculation into a variety of mouse strains. This revealed that the genetic background of the laboratory mice affected the severity of disease and altered the extent of subsequent pathology. The advent of genetically modified mice and viruses has allowed new aspects of disease to be modeled and the opportunistic nature of HCMV infection to be confirmed. This review describes the different ways that MCMV has been used to model HCMV diseases and explores the continuing difficulty faced by researchers attempting to model HCMV congenital cytomegalovirus disease using the mouse model.
Collapse
Affiliation(s)
- Michelle A. Fisher
- Division of Infection and Immunity, School of Biomedical Sciences, The University of Western Australia, Nedlands 6009, Australia;
| | - Megan L. Lloyd
- Division of Infection and Immunity, School of Biomedical Sciences, The University of Western Australia, Nedlands 6009, Australia;
- Marshall Centre for Infectious Diseases Research and Training, Division of Infection and Immunity, School of Biomedical Sciences, The University of Western Australia, Nedlands 6009, Australia
- Correspondence:
| |
Collapse
|
25
|
Putri DS, Berkebile ZW, Mustafa HJ, Fernández-Alarcón C, Abrahante JE, Schleiss MR, Bierle CJ. Cytomegalovirus infection elicits a conserved chemokine response from human and guinea pig amnion cells. Virology 2020; 548:93-100. [PMID: 32838950 DOI: 10.1016/j.virol.2020.06.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 02/06/2023]
Abstract
Human cytomegalovirus (HCMV) infects the chorioamnion, but whether these infections cause fetal membrane dysfunction remains poorly understood. We sought to assess whether guinea pig cytomegalovirus (GPCMV) infects amnion-derived cells in vitro, compare the inflammatory response of amnion cells to GPCMV and HCMV, and determine if GPCMV infects the amnion in vivo. We found that GPCMV replicates in primary guinea pig amnion derived cells and HPV16 E6/E7-transduced amniotic epithelial cells (AEC[E6/E7]s). HCMV and GPCMV infection of amnion cells increased the transcription of the chemokines CCL5/Ccl5, CXCL8/Cxcl8, and CXCL10/Cxcl10. Myd88-knockdown decreased Ccl5 and Cxc8 transcription in GPCMV-infected AEC[E6/E7]s. GPCMV was detected in the guinea pig amnion after primary maternal infection, revealing that guinea pigs are an appropriate model to study fetal membrane physiology after cytomegalovirus infection. As inflammation is known to cause fetal membrane weakening, the amnion's response to cytomegalovirus infection may cause preterm birth and other adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Dira S Putri
- Department of Pediatrics, Division of Pediatric Infectious Diseases and Immunology, University of Minnesota, Minneapolis, MN, USA.
| | - Zachary W Berkebile
- Department of Pediatrics, Division of Pediatric Infectious Diseases and Immunology, University of Minnesota, Minneapolis, MN, USA.
| | - Hiba J Mustafa
- Department of Obstetrics, Gynecology and Women's Health, Division of Maternal-Fetal Medicine, University of Minnesota, Minneapolis, MN, USA.
| | - Claudia Fernández-Alarcón
- Department of Pediatrics, Division of Pediatric Infectious Diseases and Immunology, University of Minnesota, Minneapolis, MN, USA.
| | - Juan E Abrahante
- Informatics Institute, University of Minnesota, Minneapolis, MN, USA.
| | - Mark R Schleiss
- Department of Pediatrics, Division of Pediatric Infectious Diseases and Immunology, University of Minnesota, Minneapolis, MN, USA.
| | - Craig J Bierle
- Department of Pediatrics, Division of Pediatric Infectious Diseases and Immunology, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
26
|
Billette de Villemeur A, Tattevin P, Salmi LR. Hygiene promotion might be better than serological screening to deal with Cytomegalovirus infection during pregnancy: a methodological appraisal and decision analysis. BMC Infect Dis 2020; 20:418. [PMID: 32546244 PMCID: PMC7298945 DOI: 10.1186/s12879-020-05139-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 06/05/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Cytomegalovirus infection is the most frequent viral congenital infection, with possible consequences such as deafness, or psychomotor retardation. In 2016, the French High Council of Public Health was mandated to update recommendations regarding prevention of cytomegalovirus infection in pregnant women. We summarize a critical appraisal of knowledge and deterministic decision analysis comparing the current no-screening situation to serological screening during pregnancy, and to hygiene promotion. METHODS Screening was defined as systematic serological testing, during the first trimester, with repeated tests as needed, to all pregnant women. Outcomes were: 1) severe sequela: intellectual deficiency with IQ ≤ 50 or hearing impairment < 70 dB or sight impairment (≤ 3/10 at best eye); 2) moderate sequela: any level of intellectual, hearing or sight deficiency; and 3) death or termination of pregnancy. We simulated the one-year course of cytomegalovirus infection in a cohort of 800,000 pregnant women. We developed a deterministic decision model, using best and min-max estimates, extracted from systematic reviews or original studies. RESULTS Relevant data were scarce or imprecise. We estimated that 4352 maternal primary infections would result in 1741 foetal infections, and an unknown number of maternal reinfections would result in 1699 foetal infections. There would be 788 cytomegalovirus-related consequences, including 316 foetal deaths or terminations of pregnancy, and 424 moderate and 48 severe sequelae. Screening would result in a 1.66-fold increase of poor outcomes, mostly related to a 2.93-fold increase in deaths and terminations of pregnancy, not compensated by the decrease in severe symptomatic newborns. The promotion of hygiene would result in a 0.75-fold decrease of poor outcomes, related to both a decrease in severe sequelae among symptomatic newborns (RR = 0.75; min-max: 1.00-0.68), and in deaths and terminations of pregnancy (RR = 0.75; min-max: 0.97-0.68). CONCLUSIONS Prevention of cytomegalovirus infection during pregnancy should promote hygiene; serological screening should not be recommended.
Collapse
Affiliation(s)
| | - Pierre Tattevin
- CHU de Rennes, Service de maladies infectieuses et médecine tropicale, F-35000, Rennes, France
| | - Louis-Rachid Salmi
- CHU de Bordeaux, Pôle de santé publique, Service d'Information Médicale, F-33000, Bordeaux, France.
- Univ. Bordeaux, ISPED, Centre INSERM U1219-Bordeaux Population Health, F-33000, Bordeaux, France.
- INSERM, ISPED, Centre INSERM U1219-Bordeaux Population Health, F-33000, Bordeaux, France.
| |
Collapse
|
27
|
Jaiman S, Romero R, Pacora P, Jung E, Bhatti G, Yeo L, Kim YM, Kim B, Kim CJ, Kim JS, Qureshi F, Jacques SM, Erez O, Gomez-Lopez N, Hsu CD. Disorders of placental villous maturation in fetal death. J Perinat Med 2020; 0:/j/jpme.ahead-of-print/jpm-2020-0030/jpm-2020-0030.xml. [PMID: 32238609 PMCID: PMC8262362 DOI: 10.1515/jpm-2020-0030] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 02/21/2020] [Indexed: 12/22/2022]
Abstract
Objective The aims of this study were to ascertain the frequency of disorders of villous maturation in fetal death and to also delineate other placental histopathologic lesions in fetal death. Methods This was a retrospective observational cohort study of fetal deaths occurring among women between January 2004 and January 2016 at Hutzel Women's Hospital, Detroit, MI, USA. Cases comprised fetuses with death beyond 20 weeks' gestation. Fetal deaths with congenital anomalies and multiple gestations were excluded. Controls included pregnant women without medical/obstetrical complications and delivered singleton, term (37-42 weeks) neonate with 5-min Apgar score ≥7 and birthweight between the 10th and 90th percentiles. Results Ninety-two percent (132/143) of placentas with fetal death showed placental histologic lesions. Fetal deaths were associated with (1) higher frequency of disorders of villous maturation [44.0% (64/143) vs. 1.0% (4/405), P < 0.0001, prevalence ratio, 44.6; delayed villous maturation, 22% (31/143); accelerated villous maturation, 20% (28/143); and maturation arrest, 4% (5/143)]; (2) higher frequency of maternal vascular malperfusion lesions [75.5% (108/143) vs. 35.7% (337/944), P < 0.0001, prevalence ratio, 2.1] and fetal vascular malperfusion lesions [88.1% (126/143) vs. 19.7% (186/944), P < 0.0001, prevalence ratio, 4.5]; (3) higher frequency of placental histologic patterns suggestive of hypoxia [59.0% (85/143) vs. 9.3% (82/942), P < 0.0001, prevalence ratio, 6.8]; and (4) higher frequency of chronic inflammatory lesions [53.1% (76/143) vs. 29.9% (282/944), P < 0.001, prevalence ratio 1.8]. Conclusion This study demonstrates that placentas of women with fetal death were 44 times more likely to present disorders of villous maturation compared to placentas of those with normal pregnancy. This suggests that the burden of placental disorders of villous maturation lesions is substantial.
Collapse
Affiliation(s)
- Sunil Jaiman
- Perinatology 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, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Pathology, Hutzel Women’s Hospital, Wayne State University School of Medicine, Detroit, MI, USA
| | - Roberto Romero
- Perinatology 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, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
- Detroit Medical Center, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Florida International University, Miami, Florida, USA
| | - Percy Pacora
- Perinatology 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, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Eunjung Jung
- Perinatology 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, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Gaurav Bhatti
- Perinatology 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, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Lami Yeo
- Perinatology 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, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Yeon Mee Kim
- Perinatology 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, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Pathology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Bomi Kim
- Perinatology 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, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Pathology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Chong Jai Kim
- Perinatology 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, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Jung-Sun Kim
- Perinatology 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, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Pathology, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Republic of Korea
| | - Faisal Qureshi
- Perinatology 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, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Pathology, Hutzel Women’s Hospital, Wayne State University School of Medicine, Detroit, MI, USA
| | - Suzanne M. Jacques
- Perinatology 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, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Pathology, Hutzel Women’s Hospital, Wayne State University School of Medicine, Detroit, MI, USA
| | - Offer Erez
- Perinatology 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, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Soroka University Medical Center, School of Medicine, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Nardhy Gomez-Lopez
- Perinatology 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, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Chaur-Dong Hsu
- Perinatology 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, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
28
|
Differential Expression of PDGF Receptor-α in Human Placental Trophoblasts Leads to Different Entry Pathways by Human Cytomegalovirus Strains. Sci Rep 2020; 10:1082. [PMID: 31974453 PMCID: PMC6978357 DOI: 10.1038/s41598-020-57471-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 12/12/2019] [Indexed: 12/14/2022] Open
Abstract
Human cytomegalovirus (CMV) is the leading non-genetic cause of fetal malformation in developed countries. CMV placental infection is a pre-requisite for materno-fetal transmission of virus, and fetal infection. We investigated the roles of the viral pentameric complex gH/gL/pUL128-pUL131A, and cellular platelet-derived growth factor receptor-α (PDGFRα) for CMV infection in first trimester extravillous-derived (SGHPL-4) and villous-derived (HTR-8/SVneo) trophoblast cells. Infection with four CMV clinical and laboratory strains (Merlin, TB40E, Towne, AD169), and Merlin deletion mutants of UL128-, UL130-, and UL131A-genes, showed a cell type-dependent requirement of the viral pentameric complex for infection of trophoblast cells. The viral pentameric complex was essential for infection of villous trophoblasts, but non-essential for extravillous trophoblasts. Blocking of PDGFRα in extravillous trophoblasts, which naturally express PDGFRα, inhibited entry of pentameric complex-deficient CMV strains, but not the entry of pentameric positive CMV strains. Transient expression of PDGFRα in villous trophoblasts, which are naturally deficient in PDGFRα, promoted the entry of CMV strains lacking gH/gL/pUL128-pUL131A, but had no effect on entry of pentameric positive CMV strains. These results suggest PDGFRα is an important cell receptor for entry of CMV mutant strains lacking gH/gL/pUL128-pUL131A complexes in some placental cells, suggesting these entry pathways could be potential antiviral targets.
Collapse
|
29
|
Directive clinique No 394 - Investigation sur la mortinaissance. JOURNAL OF OBSTETRICS AND GYNAECOLOGY CANADA 2020; 42:100-108. [DOI: 10.1016/j.jogc.2019.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
30
|
Graham N, Heazell AEP. When the Fetus Goes Still and the Birth Is Tragic: The Role of the Placenta in Stillbirths. Obstet Gynecol Clin North Am 2019; 47:183-196. [PMID: 32008668 DOI: 10.1016/j.ogc.2019.10.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Because of the critical role that placental structure and function plays during pregnancy, abnormal placental structure and function is closely related to stillbirth: when an infant dies before birth. However, understanding the role of the placental and specific lesions is incomplete, in part because of the variation in definitions of lesions and in classifying causes of stillbirths. Nevertheless, placental abnormalities are seen more frequently in stillbirths than live births, with placental abruption, chorioamnionitis, and maternal vascular malperfusion most commonly reported. Critically, some placental lesions affect the management of subsequent pregnancies. Histopathological examination of the placenta is recommended following stillbirth.
Collapse
Affiliation(s)
- Nicole Graham
- Faculty of Biological, Medical and Human Sciences, Maternal and Fetal Health Research Centre, School of Medical Sciences, University of Manchester, Manchester Academic Health Science Centre, St. Mary's Hospital, Central Manchester University Hospitals NHS Foundation Trust, 5th Floor (Research), Oxford Road, Manchester M13 9WL, UK
| | - Alexander E P Heazell
- Faculty of Biological, Medical and Human Sciences, Maternal and Fetal Health Research Centre, School of Medical Sciences, University of Manchester, Manchester Academic Health Science Centre, St. Mary's Hospital, Central Manchester University Hospitals NHS Foundation Trust, 5th Floor (Research), Oxford Road, Manchester M13 9WL, UK.
| |
Collapse
|
31
|
Costa ML, de Moraes Nobrega G, Antolini-Tavares A. Key Infections in the Placenta. Obstet Gynecol Clin North Am 2019; 47:133-146. [PMID: 32008664 DOI: 10.1016/j.ogc.2019.10.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Congenital infections are an important cause of morbidity and mortality worldwide, especially in low-income settings. This review discusses the main pathways of infections and associated adverse maternal and fetal outcomes, considering the TORCH pathogens, including Zika virus; the acronym stands for Toxoplasma gondii infection, other (Listeria monocytogenes, Treponema pallidum, and parvovirus B19, among others, including Zika virus), rubella virus, cytomegalovirus, and herpes simplex viruses type 1 and type 2.
Collapse
Affiliation(s)
- Maria Laura Costa
- Department of Obstetrics and Gynecology, School of Medicine, University of Campinas, Rua Alexander Fleming 101, Campinas, São Paulo 13084-881, Brazil.
| | - Guilherme de Moraes Nobrega
- Department of Obstetrics and Gynecology, School of Medicine, University of Campinas, Rua Alexander Fleming 101, Campinas, São Paulo 13084-881, Brazil
| | - Arthur Antolini-Tavares
- Department of Pathological Anatomy, School of Medicine, University of Campinas, Rua Alexander Fleming 101, Campinas, São Paulo 13084-881, Brazil
| |
Collapse
|
32
|
Tabata T, Petitt M, Fang-Hoover J, Freed DC, Li F, An Z, Wang D, Fu TM, Pereira L. Neutralizing Monoclonal Antibodies Reduce Human Cytomegalovirus Infection and Spread in Developing Placentas. Vaccines (Basel) 2019; 7:vaccines7040135. [PMID: 31569508 PMCID: PMC6963214 DOI: 10.3390/vaccines7040135] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/16/2019] [Accepted: 09/22/2019] [Indexed: 12/18/2022] Open
Abstract
Congenital human cytomegalovirus (HCMV) infection is a leading cause of birth defects worldwide, yet the most effective strategies for preventing virus transmission during pregnancy are unknown. We measured the efficacy of human monoclonal antibodies (mAbs) to HCMV attachment/entry factors glycoprotein B (gB) and the pentameric complex, gH/gL-pUL128–131, in preventing infection and spread of a clinical strain in primary placental cells and explants of developing anchoring villi. A total of 109 explants from five first-trimester placentas were cultured, and infection was analyzed in over 400 cell columns containing ~120,000 cytotrophoblasts (CTBs). mAbs to gB and gH/gL, 3-25 and 3-16, respectively, neutralized infection in stromal fibroblasts and trophoblast progenitor cells. mAbs to pUL128-131 of the pentameric complex, 1-103 and 2-18, neutralized infection of amniotic epithelial cells better than mAbs 3-25 and 3-16 and hyperimmune globulin. Select mAbs neutralized infection of cell column CTBs, with mAb 2-18 most effective, followed by mAb 3-25. Treatment of anchoring villi with mAbs postinfection reduced spread in CTBs and impaired formation of virion assembly compartments, with mAb 2-18 achieving better suppression at lower concentrations. These results predict that antibodies generated by HCMV vaccines or used for passive immunization have the potential to reduce transplacental transmission and congenital disease.
Collapse
Affiliation(s)
- Takako Tabata
- Department of Cell and Tissue Biology, University of California, San Francisco, CA 94143, USA.
| | - Matthew Petitt
- Department of Cell and Tissue Biology, University of California, San Francisco, CA 94143, USA.
| | - June Fang-Hoover
- Department of Cell and Tissue Biology, University of California, San Francisco, CA 94143, USA.
| | | | | | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX 77030, USA.
| | - Dai Wang
- Merck & Co., Inc., Kenilworth, NJ 07033, USA.
| | - Tong-Ming Fu
- Merck & Co., Inc., Kenilworth, NJ 07033, USA.
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX 77030, USA.
| | - Lenore Pereira
- Department of Cell and Tissue Biology, University of California, San Francisco, CA 94143, USA.
| |
Collapse
|
33
|
Mathias CR, Joung SJS. Diagnostic challenges in congenital cytomegalovirus infection in pregnancy: A case report. Case Rep Womens Health 2019; 22:e00119. [PMID: 31192993 PMCID: PMC6510697 DOI: 10.1016/j.crwh.2019.e00119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/20/2019] [Accepted: 04/25/2019] [Indexed: 11/26/2022] Open
Abstract
Cytomegalovirus is the most common congenital viral infection. Infection can cause developmental delay, sensorineural deafness and fetal death. Fetal damage is more severe when infection occurs in the first trimester of pregnancy. Prenatal ultrasound findings may be cerebral, such as ventriculomegaly, microcephaly and periventricular leukomalacia, as well as non-cerebral, such as echogenic bowel, ascites and pericardial effusion. We present a case of congenital cytomegalovirus infection in which the only ultrasound sign noted at routine second-trimester scan was low-grade echogenic bowel, a soft marker, which progressed to severe disease in the third trimester, when further investigation was prompted, leading to the diagnosis. Patients need to be counselled regarding the possible perinatal prognosis. Ultrasound markers can often but not always predict severity and, hence, counselling can be a challenge. Conclusion: A meticulous anatomy survey in mid-trimester remains the norm and ultrasound soft markers should prompt comprehensive testing for viral infections in pregnancy. Fetal damage caused by infection with CMV is more severe when the infection occurs in the first trimester of pregnancy. CMV is the most common congenital viral infection causingdevelopmental delay, sensorineural deafness and fetal death. Ultrasound markers can often but not always predict the severity of congenital infection. A meticulous anatomy survey in mid-trimester remains the norm. Ultrasound soft markers should prompt comprehensive testing for viral infections in pregnancy.
Collapse
Affiliation(s)
- Caroline Ruth Mathias
- Department of Obstetrics and Gynaecology, Nepean Hospital, Kingswood 2747, NSW, Australia
| | - Steven Jin Sung Joung
- Department of Obstetrics and Gynaecology, Nepean Hospital, Kingswood 2747, NSW, Australia
| |
Collapse
|
34
|
Anfasa F, Goeijenbier M, Widagdo W, Siegers JY, Mumtaz N, Okba N, van Riel D, Rockx B, Koopmans MPG, Meijers JCM, Martina BEE. Zika Virus Infection Induces Elevation of Tissue Factor Production and Apoptosis on Human Umbilical Vein Endothelial Cells. Front Microbiol 2019; 10:817. [PMID: 31068911 PMCID: PMC6491739 DOI: 10.3389/fmicb.2019.00817] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 04/01/2019] [Indexed: 12/22/2022] Open
Abstract
Zika virus (ZIKV) infection is typically characterized by a mild disease presenting with fever, maculopapular rash, headache, fatigue, myalgia, and arthralgia. A recent animal study found that ZIKV-infected pregnant Ifnar -/-mice developed vascular damage in the placenta and reduced amount of fetal capillaries. Moreover, ZIKV infection causes segmental thrombosis in the umbilical cord of pregnant rhesus macaques. Furthermore, several case reports suggest that ZIKV infection cause coagulation disorders. These results suggest that ZIKV could cause an alteration in the host hemostatic response, however, the mechanism has not been investigated thus far. This paper aims to determine whether ZIKV infection on HUVECs induces apoptosis and elevation of tissue factor (TF) that leads to activation of secondary hemostasis. We infected HUVECs with two ZIKV strains and performed virus titration, immunostaining, and flow cytometry to confirm and quantify infection. We measured TF concentrations with flow cytometry and performed thrombin generation test (TGT) as a functional assay to assess secondary hemostasis. Furthermore, we determined the amount of cell death using flow cytometry. We also performed enzyme-linked immunosorbent assay (ELISA) to determine interleukin (IL)-6 and IL-8 production and conducted blocking experiments to associate these cytokines with TF expression. Both ZIKV strains infected and replicated to high titers in HUVECs. We found that infection induced elevation of TF expressions. We also showed that increased TF expression led to shortened TGT time. Moreover, the data revealed that infection induced apoptosis. In addition, there was a significant increase of IL-6 and IL-8 production in infected cells. Here we provide in vitro evidence that infection of HUVECs with ZIKV induces apoptosis and elevation of TF expression that leads to activation of secondary hemostasis.
Collapse
Affiliation(s)
- Fatih Anfasa
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands.,Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Marco Goeijenbier
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands.,Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Widagdo Widagdo
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Jurre Y Siegers
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Noreen Mumtaz
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Nisreen Okba
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Debby van Riel
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Barry Rockx
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Marion P G Koopmans
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Joost C M Meijers
- Department of Plasma Proteins, Sanquin Research, Amsterdam, Netherlands.,Department of Experimental Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Byron E E Martina
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, Netherlands.,Artemis One Health Research Institute, Delft, Netherlands
| |
Collapse
|
35
|
Huynh KT, van Zuylen WJ, Ford CE, Rawlinson WD. Selective modulation of Wnt-binding receptor tyrosine kinase ROR2 expression by human cytomegalovirus regulates trophoblast migration. J Gen Virol 2019; 100:99-104. [DOI: 10.1099/jgv.0.001179] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Kim T. Huynh
- 1School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Wendy J. van Zuylen
- 1School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
- 2Serology and Virology Division, NSW Health Pathology, Prince of Wales Hospital, Sydney, NSW, Australia
| | - Caroline E. Ford
- 3Gynaecological Cancer Research Group, School of Women’s and Children’s Health, University of New South Wales, Sydney, NSW, Australia
| | - William D. Rawlinson
- 4School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
- 3Gynaecological Cancer Research Group, School of Women’s and Children’s Health, University of New South Wales, Sydney, NSW, Australia
- 2Serology and Virology Division, NSW Health Pathology, Prince of Wales Hospital, Sydney, NSW, Australia
- 1School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
36
|
Yapar Eyi EG, Altuğ N. Aortic Arch Thrombosis Associated with Fetal Cytomegalovirus Viremia. AJP Rep 2019; 9:e23-e26. [PMID: 30723573 PMCID: PMC6358456 DOI: 10.1055/s-0038-1675631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 09/11/2018] [Indexed: 11/26/2022] Open
Abstract
Cytomegalovirus(CMV) associated thrombosis has been reported sporadically in the medical literature; however, its antenatal scenario has not been documented. We herein present the antenatal, Doppler's ultrasound and magnetic resonance angiographic features of thrombosis in the aortic arch showing extension toward the medial lumen of the brachiocephalic trunk with critical occlusion of the left common carotid artery and left subclavian artery in a term fetus to raise obstetricians'/ neonatologists'/pediatric cardiologists' awareness for the association between CMV viremia and intrauterine thrombosis that caused cerebral injury, neurodevelopmental impairment, and permanent sequela.
Collapse
Affiliation(s)
- Elif Gul Yapar Eyi
- Perinatology Subdivision, Zekai Tahir Burak Women's Health Education and Research Hospital, Ankara, Turkey
| | - Nahide Altuğ
- Pediatric Cardiology, Zekai Tahir Burak Women's Health Education and Research Hospital, Ankara, Turkey
| |
Collapse
|
37
|
A Native Human Monoclonal Antibody Targeting HCMV gB (AD-2 Site I). Int J Mol Sci 2018; 19:ijms19123982. [PMID: 30544903 PMCID: PMC6321246 DOI: 10.3390/ijms19123982] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 12/04/2018] [Accepted: 12/08/2018] [Indexed: 12/22/2022] Open
Abstract
Hyperimmune globulin (HIG) has shown efficacy against human cytomegalovirus (HCMV) for both transplant and congenital transmission indications. Replicating that activity with a monoclonal antibody (mAb) offers the potential for improved consistency in manufacturing, lower infusion volume, and improved pharmacokinetics, as well as reduced risk of off-target reactivity leading to toxicity. HCMV pathology is linked to its broad cell tropism. The glycoprotein B (gB) envelope protein is important for infections in all cell types. Within gB, the antigenic determinant (AD)-2 Site I is qualitatively more highly-conserved than any other region of the virus. TRL345, a high affinity (Kd = 50 pM) native human mAb to this site, has shown efficacy in neutralizing the infection of fibroblasts, endothelial and epithelial cells, as well as specialized placental cells including trophoblast progenitor cells. It has also been shown to block the infection of placental fragments grown ex vivo, and to reduce syncytial spread in fibroblasts in vitro. Manufacturing and toxicology preparation for filing an IND (investigational new drug) application with the US Food and Drug Administration (FDA) are expected to be completed in mid-2019.
Collapse
|
38
|
Manickam C, Shah SV, Lucar O, Ram DR, Reeves RK. Cytokine-Mediated Tissue Injury in Non-human Primate Models of Viral Infections. Front Immunol 2018; 9:2862. [PMID: 30568659 PMCID: PMC6290327 DOI: 10.3389/fimmu.2018.02862] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 11/20/2018] [Indexed: 12/12/2022] Open
Abstract
Viral infections trigger robust secretion of interferons and other antiviral cytokines by infected and bystander cells, which in turn can tune the immune response and may lead to viral clearance or immune suppression. However, aberrant or unrestricted cytokine responses can damage host tissues, leading to organ dysfunction, and even death. To understand the cytokine milieu and immune responses in infected host tissues, non-human primate (NHP) models have emerged as important tools. NHP have been used for decades to study human infections and have played significant roles in the development of vaccines, drug therapies and other immune treatment modalities, aided by an ability to control disease parameters, and unrestricted tissue access. In addition to the genetic and physiological similarities with humans, NHP have conserved immunologic properties with over 90% amino acid similarity for most cytokines. For example, human-like symptomology and acute respiratory syndrome is found in cynomolgus macaques infected with highly pathogenic avian influenza virus, antibody enhanced dengue disease is common in neotropical primates, and in NHP models of viral hepatitis cytokine-induced inflammation induces severe liver damage, fibrosis, and hepatocellular carcinoma recapitulates human disease. To regulate inflammation, anti-cytokine therapy studies in NHP are underway and will provide important insights for future human interventions. This review will provide a comprehensive outline of the cytokine-mediated exacerbation of disease and tissue damage in NHP models of viral infections and therapeutic strategies that can aid in prevention/treatment of the disease syndromes.
Collapse
Affiliation(s)
- Cordelia Manickam
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Spandan V. Shah
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Olivier Lucar
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Daniel R. Ram
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - R. Keith Reeves
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Ragon Institute of Massachusetts General Hospital, MIT and Harvard, Cambridge, MA, United States
| |
Collapse
|
39
|
Hamilton ST, Hutterer C, Egilmezer E, Steingruber M, Milbradt J, Marschall M, Rawlinson WD. Human cytomegalovirus utilises cellular dual-specificity tyrosine phosphorylation-regulated kinases during placental replication. Placenta 2018; 72-73:10-19. [PMID: 30501876 DOI: 10.1016/j.placenta.2018.10.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 09/28/2018] [Accepted: 10/12/2018] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Congenital cytomegalovirus (HCMV) infection may cause significant fetal malformation and in severe cases fetal and neonatal death. Fetal injury may be caused indirectly by the placental response to infection. Dual-specificity tyrosine phosphorylation-regulated kinases (DYRKs) have recently been identified as critical kinases for HCMV replication. In this study we provide first evidence that DYRK1A and DYRK1B are utilised during HCMV placental replication. METHODS DYRK expression was investigated in AD169- and Merlin-infected TEV-1 trophoblast cells, ex vivo placental explants and naturally infected clinical placentae by immunofluorescence, western blot, co-immunoprecipitation and RT-qPCR. RESULTS HCMV-infected placental cells showed accumulation and re-localisation of DYRK1A and DYRK1B protein to areas of cytoplasmic virion assembly complexes and nuclear viral replication compartments, respectively. This accumulation was a result of upregulated DYRK1A/B protein expression with HCMV inducing up to a 5.3-fold increase in DYRK1A and up to a 4.7-fold increase in DYRK1B protein, relative to mock-infected TEV-1 cells (p < 0.0001). Increased DYRK protein expression was correlated with DYRK1A/B mRNA upregulation, with HCMV-infected cells showing up to a 3.7-fold increase and 2.9-fold increase in DYRK1A and DYRK1B mRNA levels respectively (p < 0.05). Protein-protein interactions were detected between DYRK1A/1B complexes and HCMV immediate early IE2p86, early pp65 and pUL44 and late pp150 proteins. Treatment of HCMV-infected TEV-1 cells and placental explants with DYRK inhibitors significantly inhibited HCMV replication (p < 0.05) indicating these cellular kinases are required during HCMV placental replication. CONCLUSION HCMV modulates cellular DYRKs during placental replication which may have implications for congenital HCMV pathogenesis and represent promising antiviral targets.
Collapse
Affiliation(s)
- Stuart T Hamilton
- Serology and Virology Division, SEALS Microbiology, Prince of Wales Hospital, Sydney, Australia; School of Women's and Children's Health, University of New South Wales, Australia
| | - Corina Hutterer
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Germany
| | - Ece Egilmezer
- Serology and Virology Division, SEALS Microbiology, Prince of Wales Hospital, Sydney, Australia; School of Medical Sciences, University of New South Wales, Australia
| | - Mirjam Steingruber
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Germany
| | - Jens Milbradt
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Germany
| | - Manfred Marschall
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Germany
| | - William D Rawlinson
- Serology and Virology Division, SEALS Microbiology, Prince of Wales Hospital, Sydney, Australia; School of Women's and Children's Health, University of New South Wales, Australia; School of Medical Sciences, University of New South Wales, Australia; School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, Australia.
| |
Collapse
|
40
|
Abstract
In the United States, cytomegalovirus is the most common congenital viral infection and the number 1 cause of nonhereditary sensorineural hearing loss. The vast majority of infants may be asymptomatic, especially if cytomegalovirus is contracted later in the pregnancy, and some symptoms may have a delayed onset. Therefore, it is important for the pathologist to identify the common histologic findings to help confirm the diagnosis so the child can be followed for late sequelae. Histologic examination of the placenta is important in live births and in cases of intrauterine fetal demise. Chronic lymphoplasmacytic villitis and fibrotic, avascular villi are the most common findings. When present, Cowdry A intranuclear and basophilic intracytoplasmic inclusions are characteristic. Immunohistochemistry for cytomegalovirus can highlight these inclusions as well as the associated eosinophilic debris. In addition, polymerase chain reaction or viral culture on placental or fetal samples can be performed for confirmation.
Collapse
Affiliation(s)
- Kaleigh Lindholm
- From the Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora (Dr Lindholm); and the Department of Pathology, Denver Health Medical Center, Denver, Colorado (Dr O'Keefe)
| | | |
Collapse
|
41
|
Abstract
Why certain viruses cross the physical barrier of the human placenta but others do not is incompletely understood. Over the past 20 years, we have gained deeper knowledge of intrauterine infection and routes of viral transmission. This review focuses on human viruses that replicate in the placenta, infect the fetus, and cause birth defects, including rubella virus, varicella-zoster virus, parvovirus B19, human cytomegalovirus (CMV), Zika virus (ZIKV), and hepatitis E virus type 1. Detailed discussions include ( a) the architecture of the uterine-placental interface, ( b) studies of placental explants ex vivo that provide insights into the infection and spread of CMV and ZIKV to the fetal compartment and how these viruses undermine early development, and ( c) novel treatments and vaccines that limit viral replication and have the potential to reduce dissemination, vertical transmission and the occurrence of congenital disease.
Collapse
Affiliation(s)
- Lenore Pereira
- Department of Cell and Tissue Biology, School of Dentistry, University of California, San Francisco, California 94143, USA;
| |
Collapse
|
42
|
Shi TL, Huang LJ, Xiong YQ, Zhong YY, Yang JJ, Fu T, Lei XF, Chen Q. The risk of herpes simplex virus and human cytomegalovirus infection during pregnancy upon adverse pregnancy outcomes: A meta-analysis. J Clin Virol 2018; 104:48-55. [DOI: 10.1016/j.jcv.2018.04.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 04/13/2018] [Accepted: 04/24/2018] [Indexed: 12/12/2022]
|
43
|
Rawlinson WD, Palasanthiran P, Hall B, Al Yazidi L, Cannon MJ, Cottier C, van Zuylen WJ, Wilkinson M. Neonates with congenital Cytomegalovirus and hearing loss identified via the universal newborn hearing screening program. J Clin Virol 2018; 102:110-115. [PMID: 29571077 DOI: 10.1016/j.jcv.2018.03.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 03/11/2018] [Accepted: 03/14/2018] [Indexed: 11/27/2022]
Abstract
BACKGROUND Congenital cytomegalovirus (CMV) is the most common non-genetic cause of sensorineural hearing loss. Currently, there are no universal CMV screening programs for newborns or routine CMV testing of neonates with hearing loss in Australia, or elsewhere. OBJECTIVES This study was undertaken to determine the prevalence of congenital CMV infection in infants with hearing loss identified using routine resources via the Australian universal neonatal hearing screening (UNHS) program. STUDY DESIGN Infants who failed UNHS, referred for audiological testing and found to have permanent hearing loss were screened for CMV via PCR of urine and saliva. Congenital CMV was diagnosed if CMV was detected in infants ≤30 days of age, or using retrospective testing on stored new born screening cards, retrospective testing, or using clinical criteria if >30 days of age. The cohort was analyzed for time of testing and prevalence of congenital CMV determined. RESULTS The Audiology Department reviewed 1669 infants who failed UNHS between 2009 and 2016. Thirty percent (502/1669) had permanent hearing loss confirmed, of whom 336/502 were offered CMV testing. A definite (n = 11) or probable (n = 8) diagnosis of congenital CMV occurred in 19/323 (5.9%), of whom definite diagnoses were made in 4/19 on tests positive prior to 21 days of life, in 5/19 who were positive on neonatal blood screening card (NBSC) testing, in 2/19 who were positive on placental testing. In 8/19 probable diagnoses were made based on positive testing between ages 23-42 days and a consistent clinical syndrome in the absence of another cause for hearing loss after genetic and other testing. CMV testing mirrored the timing of audiological testing, with ∼40% completing audiology and CMV testing by 21 days, and 64% by 30 days. CONCLUSION This program, utilizing existing clinical services identified probable congenital CMV in ∼6% of a large cohort failing UNHS with permanent hearing loss, of whom more than half were definite diagnoses. No additional assets were required to those already existing in this tertiary referral pediatric centre, whilst providing useful and timely data for clinical and audiological management.
Collapse
Affiliation(s)
- William D Rawlinson
- Serology and Virology Division, NSW Health Pathology, Prince of Wales Hospital, Sydney, NSW, Australia; School of Medical Sciences & School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia.
| | - Pamela Palasanthiran
- Department of Immunology and Infectious Diseases, Sydney Children's Hospital & School of Women's and Child Health, University of New South Wales, Sydney, NSW, Australia
| | - Beverly Hall
- Serology and Virology Division, NSW Health Pathology, Prince of Wales Hospital, Sydney, NSW, Australia
| | - Laila Al Yazidi
- Department of Immunology and Infectious Diseases, Sydney Children's Hospital & School of Women's and Child Health, University of New South Wales, Sydney, NSW, Australia
| | - Michael J Cannon
- National Center on Birth Defects and Developmental Disabilities, US Centers for Disease Control and Prevention, Centers for Disease Control and Prevention, GA, USA
| | - Carolyn Cottier
- Hearing Support Services, Sydney Children's Hospital, Sydney, NSW, Australia
| | - Wendy J van Zuylen
- Serology and Virology Division, NSW Health Pathology, Prince of Wales Hospital, Sydney, NSW, Australia; School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Monica Wilkinson
- Audiology Department, Sydney Children's Hospital, Sydney, NSW, Australia
| |
Collapse
|
44
|
Update on treatment of cytomegalovirus infection in pregnancy and of the newborn with congenital cytomegalovirus. Curr Opin Infect Dis 2018; 29:615-624. [PMID: 27607910 DOI: 10.1097/qco.0000000000000317] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE OF REVIEW The purpose of this review is to assess the recent studies of therapy of pregnant women and neonates, aimed at preventing the consequences of congenital cytomegalovirus (CMV) infection. RECENT FINDINGS A recent randomized controlled trial of treatment of CMV during pregnancy with hyperimmune globulin did not show significant efficacy in prevention of foetal infection and morbidity, although there was a trend towards improvement with treatment. Trials of antiviral therapy of the mother during pregnancy have involved small numbers only, confounded by ethical and practical difficulties, and further studies are needed to demonstrate whether or not antivirals are useful and well tolerated in this setting.Antiviral treatment of neonatal CMV acquired congenitally has been studied in well controlled trials and the antiviral valganciclovir has shown efficacy in reducing the more severe outcomes. Trials are ongoing of the use of antivirals in less severe disease, although results are likely to take several years. SUMMARY Congenital CMV infection is the most frequent cause of congenital malformation in developed countries, with a symptomatic prevalence of 0.64% of all live births. Infection may result in neurodevelopmental delay, foetal or neonatal death, and most frequently, sensorineural hearing loss. Successful control of viral infections during pregnancy and in the newborn period is essential in reducing early and late morbidity and mortality. Control of congenital CMV infection may be via primary prevention methods such as reducing contact with the pathogen, improved hygiene - both for the pregnant mother and for the neonate, or secondary prevention via reduction of vertical transmission from mother to foetus and reduction in consequences of infection by treatment of infected pregnant women and infected neonates.
Collapse
|
45
|
Mulkey SB, Vezina G, Bulas DI, Khademian Z, Blask A, Kousa Y, Cristante C, Pesacreta L, du Plessis AJ, DeBiasi RL. Neuroimaging Findings in Normocephalic Newborns With Intrauterine Zika Virus Exposure. Pediatr Neurol 2018; 78:75-78. [PMID: 29167058 DOI: 10.1016/j.pediatrneurol.2017.10.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 10/13/2017] [Accepted: 10/14/2017] [Indexed: 12/01/2022]
Abstract
BACKGROUND Congenital Zika infection can result in a spectrum of neurological abnormalities in the newborn. Newborns exposed to Zika virus in utero often have neuroimaging as part of their clinical evaluation. METHODS Through the Congenital Zika Program at Children's National Health System in Washington DC, we performed fetal or neonatal neuroimaging, including magnetic resonance imaging and ultrasound, on over 70 fetuses or neonates with intrauterine Zika exposure. Novel findings on neonatal brain magnetic resonance imaging were observed in two instances. RESULTS Gadolinium-contrast magnetic resonance imaging showed enhancement of multiple cranial nerves at three days of age on one infant. Another infant underwent magnetic resonance imaging at 16 days of age and was shown to have a chronic ischemic cerebral infarction. This infant had previously normal fetal magnetic resonance imaging. CONCLUSION Cranial nerve enhancement and cerebral infarction may be among the expanding list of neurological findings in congenital Zika infection. Postnatal brain magnetic resonance imaging should be considered for newborns exposed to Zika virus in utero.
Collapse
Affiliation(s)
- Sarah B Mulkey
- Division of Fetal and Transitional Medicine, Children's National Health System, Washington, District of Columbia; Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia; Department of Neurology, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia.
| | - Gilbert Vezina
- Division of Radiology, Children's National Health System, Washington, District of Columbia
| | - Dorothy I Bulas
- Division of Fetal and Transitional Medicine, Children's National Health System, Washington, District of Columbia; Division of Radiology, Children's National Health System, Washington, District of Columbia
| | - Zarir Khademian
- Division of Radiology, Children's National Health System, Washington, District of Columbia
| | - Anna Blask
- Division of Radiology, Children's National Health System, Washington, District of Columbia
| | - Youssef Kousa
- Division of Neurology, Children's National Health System, Washington, District of Columbia
| | - Caitlin Cristante
- Division of Fetal and Transitional Medicine, Children's National Health System, Washington, District of Columbia
| | - Lindsay Pesacreta
- Division of Fetal and Transitional Medicine, Children's National Health System, Washington, District of Columbia
| | - Adre J du Plessis
- Division of Fetal and Transitional Medicine, Children's National Health System, Washington, District of Columbia; Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia; Department of Neurology, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Roberta L DeBiasi
- Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia; Division of Infectious Diseases, Children's National Health System, Washington, District of Columbia; Department of Microbiology, Immunology and Tropical Medicine, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| |
Collapse
|
46
|
Abstract
CONTEXT - Fetal vascular malperfusion, also known as fetal thrombotic vasculopathy, remains an underrecognized pathologic finding and should be noted during placental evaluation. OBJECTIVE - To review histologic findings, gain familiarity with the updated terminology, and to recognize important clinical associations with this entity. DATA SOURCES - University of Michigan cases, PubMed search, multiple review articles including recent placental workshop group consensus statement, and selected book chapters. CONCLUSIONS - Multiple histologic patterns of fetal vascular malperfusion have been described including thrombosis, avascular villi, villous stromal-vascular karyorrhexis, intramural fibrin thrombi, and stem villous vascular obliteration. Various underlying etiologies can be involved in fetal vascular malperfusion. Cord lesions including abnormal insertion, length, and coiling are important causes. Maternal vascular malperfusion such as preeclampsia, hypercoagulable states, lupus anticoagulant, and sometimes diabetes have been associated with this condition. Fetal cardiac dysfunction/malformations and severe fetal inflammatory response in the setting of ascending intrauterine infection have also been attributed to this important finding. Fetal vascular malperfusion has been implicated in several significant and sometimes devastating clinical associations; these include intrauterine growth restriction, poor perinatal outcome, fetal demise, and neurodevelopmental sequelae. A diagnostic challenge may be encountered in cases with prior intrauterine fetal death, since degenerative changes post demise result in a similar histomorphologic picture. The diffuse versus the focal nature of the lesions may help in the distinction.
Collapse
Affiliation(s)
- Amer Heider
- From the Department of Pathology Michigan Medicine, University of Michigan, Ann Arbor
| |
Collapse
|
47
|
Opsjøn BE, Nordbø SA, Vogt C. Unrecognized viral infections and chromosome abnormalities as a cause of fetal death - examination with fluorescence in situ hybridization, immunohistochemistry and polymerase chain reaction. APMIS 2017; 125:826-832. [PMID: 28737006 DOI: 10.1111/apm.12726] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 05/03/2017] [Indexed: 11/28/2022]
Abstract
Fifteen to 50% of fetal deaths remain unexplained after post-mortem examination depending on inclusion criteria and classification systems. Our aim was to examine a selection of unexplained fetal deaths in order to investigate whether any common chromosome aberrations or viral infections were present. Reports from 351 fetal autopsies performed at the Department of Pathology and Medical Genetics at St. Olavs University Hospital from 2001 through 2010 were reviewed. Of these, 105 fetal deaths were classified as unexplained. Tissue samples from 30 cases were further examined with fluorescence in situ hybridization (FISH) to detect abnormalities in chromosomes 13, 18, and 21. The samples were also examined with immunohistochemistry (IHC) and polymerase chain reaction (PCR) to detect infections with cytomegalovirus, parvovirus B19, herpes simplex virus 1 and 2, enterovirus, and parechovirus. In two cases, a possible trisomy 13 mosaicism was found. No viruses were detected. In our selection of 30 unexplained cases, possible trisomy 13 mosaicism was found in two cases, and no viruses were detected. High degree of maceration and missing placental examination often complicate the investigation of fetal death, and extensive ancillary examinations do not necessarily contribute to a more specific diagnosis.
Collapse
Affiliation(s)
- Bente Ediassen Opsjøn
- Department of Laboratory Medicine, Children's and Women's Health, Faculty of Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Svein Arne Nordbø
- Department of Laboratory Medicine, Children's and Women's Health, Faculty of Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Department of Medical Microbiology, St. Olavs Hospital, Trondheim, Norway
| | - Christina Vogt
- Department of Laboratory Medicine, Children's and Women's Health, Faculty of Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Department of Pathology and Medical Genetics, St. Olavs Hospital, Trondheim, Norway
| |
Collapse
|
48
|
Nijkamp J, Sebire N, Bouman K, Korteweg F, Erwich J, Gordijn S. Perinatal death investigations: What is current practice? Semin Fetal Neonatal Med 2017; 22:167-175. [PMID: 28325580 PMCID: PMC7118457 DOI: 10.1016/j.siny.2017.02.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Perinatal death (PD) is a devastating obstetric complication. Determination of cause of death helps in understanding why and how it occurs, and it is an indispensable aid to parents wanting to understand why their baby died and to determine the recurrence risk and management in subsequent pregnancy. Consequently, a perinatal death requires adequate diagnostic investigation. An important first step in the analysis of PD is to identify the case circumstances, including relevant details regarding maternal history, obstetric history and current pregnancy (complications are evaluated and recorded). In the next step, placental examination is suggested in all cases, together with molecular cytogenetic evaluation and fetal autopsy. Investigation for fetal-maternal hemorrhage by Kleihauer is also recommended as standard. In cases where parents do not consent to autopsy, alternative approaches such as minimally invasive postmortem examination, postmortem magnetic resonance imaging, and fetal photographs are good alternatives. After all investigations have been performed it is important to combine findings from the clinical review and investigations together, to identify the most probable cause of death and counsel the parents regarding their loss.
Collapse
Affiliation(s)
- J.W. Nijkamp
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands,Corresponding author. Department of Obstetrics and Gynecology, University Medical Centre Groningen, CB 21, P.O. box 30001, 9700 RB Groningen, The Netherlands.
| | - N.J. Sebire
- Department of Pediatric Pathology, Clinical Molecular Genetics, Great Ormond Street Hospital for Children and UCL Institute of Child Health, London, UK
| | - K. Bouman
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - F.J. Korteweg
- Department of Obstetrics and Gynecology, Martini Hospital, Groningen, The Netherlands
| | - J.J.H.M. Erwich
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - S.J. Gordijn
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
49
|
Pereira L, Tabata T, Petitt M, Fang-Hoover J. Congenital cytomegalovirus infection undermines early development and functions of the human placenta. Placenta 2017; 59 Suppl 1:S8-S16. [PMID: 28477968 DOI: 10.1016/j.placenta.2017.04.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 04/19/2017] [Accepted: 04/24/2017] [Indexed: 12/31/2022]
Abstract
Congenital human cytomegalovirus (HCMV) infection is a major viral cause of birth defects, including microcephaly, neurological deficits, loss of hearing and vision, and intrauterine growth restriction. Despite its public health significance, there is no approved treatment for congenital infection during pregnancy; existing antivirals have unacceptable toxicities. The mechanisms of HCMV-induced placental injury, reduced capacity for compensatory development and transmission to the fetus are poorly understood, limiting the development of alternative strategies for clinical management of the disease. Recently, self-renewing, multipotent trophoblast progenitor cells (TBPCs) were reported to reside in the chorion of the human placenta and differentiate into the mature trophoblast subtypes - transport syncytiotrophoblasts and invasive cytotrophoblasts - forming chorionic villi, the functional units of the placenta. HCMV infects TBPCs, reducing the population of progenitor cells and their functional capacity to self-renew, migrate and differentiate. Human TBPCs and chorionic villus explants from first trimester represent relevant models for evaluating efficacies of new antiviral agents in protecting and restoring growth of the developing placenta in response to adverse conditions. Correlating pathology from complications of congenital HCMV infection with impaired development in the tissue environment of anchoring villus explants and defects in TBPC differentiation may enable identification of molecular pathways that could serve as targets for intervention. Here we summarize studies that could open up novel avenues of research on potential therapeutics to sustain placental development, promote differentiation and improve function and pregnancy outcomes.
Collapse
Affiliation(s)
- Lenore Pereira
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, CA 94143, United States.
| | - Takako Tabata
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, CA 94143, United States
| | - Matthew Petitt
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, CA 94143, United States
| | - June Fang-Hoover
- Department of Cell and Tissue Biology, School of Dentistry, University of California San Francisco, San Francisco, CA 94143, United States
| |
Collapse
|
50
|
Expression of KIR2DS1 by decidual natural killer cells increases their ability to control placental HCMV infection. Proc Natl Acad Sci U S A 2016; 113:15072-15077. [PMID: 27956621 DOI: 10.1073/pnas.1617927114] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The combination of the activating killer cell Ig-like receptor 2DS1 (KIR2DS1) expressed by maternal decidual natural killer cells (dNK) and the presence of its ligand, the HLA-C allotype HLA-C2, expressed by fetal trophoblasts, reduces the risk of developing pregnancy complications. However, no molecular or cellular mechanism explains this genetic correlation. Here we demonstrate that KIR2DS1+ dNK acquired higher cytotoxic function than KIR2DS1- dNK when exposed to human cytomegalovirus (HCMV)-infected decidual stromal cells (DSC), particularly when DSCs express HLA-C2. Furthermore, dNK were unable to degranulate or secrete cytokines in response to HCMV-infected primary fetal extravillous trophoblasts. This emphasizes the immunological challenge to clear placental viral infections within the immune-privileged placenta. Activation of dNK through KIR2DS1/HLA-C2 interaction increases their ability to respond to placental HCMV infection and may limit subsequent virus-induced placental pathology. This mechanism is directly related to how KIR2DS1 expressed by dNK reduces development of severe pregnancy complications such as miscarriages and preterm delivery.
Collapse
|