1
|
Wang S, Mei Z, Chen J, Zhao K, Kong R, McClements L, Zhang H, Liao A, Liu C. Maternal Immune Activation: Implications for Congenital Heart Defects. Clin Rev Allergy Immunol 2025; 68:36. [PMID: 40175706 DOI: 10.1007/s12016-025-09049-y] [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] [Accepted: 03/22/2025] [Indexed: 04/04/2025]
Abstract
Congenital heart defects (CHD) are the most common major birth defects and one of the leading causes of death from congenital defects after birth. CHD can arise in pregnancy from the combination of genetic and non-genetic factors. The maternal immune activation (MIA) hypothesis is widely implicated in embryonic neurodevelopmental abnormalities. MIA has been found to be associated with the development of asthma, diabetes mellitus, and other diseases in the offspring. Given the important role of cardiac immune cells and cytokines in embryonic heart development, it is hypothesized that MIA may play a significant role in embryonic heart development. This review aims to stimulate further investigation into the relationship between MIA and CHD and to highlight the gaps in the knowledge. It evaluates the impact of MIA on CHD in the context of pregnancy complications, immune-related diseases, infections, and environmental and lifestyle factors. The review outlines the mechanisms by which immune cells and their secretome indirectly regulate the immuno-microenvironment of the embryonic heart by influencing placental development. Furthermore, the inflammatory cytokines cross the placenta to induce related reactions including oxidative stress in the embryonic heart directly. This review delineates the role of MIA in CHD and underscores the impact of maternal factors, especially immune factors, as well as the embryonic cardiac immuno-microenvironment, on embryonic heart development. This review extends our understanding of the importance of MIA in the pathogenesis of CHD and provides important insights into prenatal prevention and treatment strategies for this congenital condition.
Collapse
Affiliation(s)
- Sixing Wang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
- Second Clinical Department, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Zilin Mei
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Jin Chen
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Kai Zhao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Ruize Kong
- Department of Vascular Surgery, NHC Key Laboratory of Healthy Birth and Birth Defect Prevention in Western China First People'S Hospital of Yunnan Province, Kunming, PR China
| | - Lana McClements
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Huiping Zhang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China.
| | - Aihua Liao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China.
| | - Chunyan Liu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China.
| |
Collapse
|
2
|
Borrego-Ruiz A, Borrego JJ. Involvement of virus infections and antiviral agents in schizophrenia. Psychol Med 2025; 55:e73. [PMID: 40059820 PMCID: PMC12055031 DOI: 10.1017/s0033291725000467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 02/06/2025] [Accepted: 02/12/2025] [Indexed: 05/08/2025]
Abstract
BACKGROUND Schizophrenia is a chronic and complex mental disorder resulting from interactions between cumulative and synergistic genetic and environmental factors. Viral infection during the prenatal stage constitutes one of the most relevant risk factors for the development of schizophrenia later in adulthood. METHODS A narrative review was conducted to explore the link between viral infections and schizophrenia, as well as the neuropsychiatric effects of antiviral drugs, particularly in the context of this specific mental condition. Literature searches were performed using the PubMed, Scopus, and Web of Science databases. RESULTS Several viral infections, such as herpesviruses, influenza virus, Borna disease virus, and coronaviruses, can directly or indirectly disrupt normal fetal brain development by modifying gene expression in the maternal immune system, thereby contributing to the pathophysiological symptoms of schizophrenia. In addition, neuropsychiatric effects caused by antiviral drugs are frequent and represent significant adverse outcomes for viral treatment. CONCLUSIONS Epidemiological evidence suggests a potential relationship between viruses and schizophrenia. Increases in inflammatory cytokine levels and changes in the expression of key genes observed in several viral infections may constitute potential links between these viral infections and schizophrenia. Furthermore, antivirals may affect the central nervous system, although for most drugs, their mechanisms of action are still unclear, and a strong relationship between antivirals and schizophrenia has not yet been established.
Collapse
Affiliation(s)
- Alejandro Borrego-Ruiz
- Departamento de Psicología Social y de las Organizaciones, Universidad Nacional de Educación a Distancia (UNED), Madrid, Spain
| | - Juan J. Borrego
- Departamento de Microbiología, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA, Plataforma BIONAND, Málaga, Spain
| |
Collapse
|
3
|
Zheng X, Wang T, Hao H, D'Souza RR, Strickland MJ, Warren JL, Darrow LA, Chang HH. Influenza Activity and Preterm Birth in the Atlanta Metropolitan Area: A Time-Series Analysis from 2010 to 2017. Epidemiology 2025; 36:141-148. [PMID: 39588975 PMCID: PMC11785485 DOI: 10.1097/ede.0000000000001819] [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] [Indexed: 11/27/2024]
Abstract
BACKGROUND Annual influenza epidemics lead to a substantial public health burden, and pregnant people are vulnerable to severe outcomes. Influenza during pregnancy is hypothesized to increase the risk of adverse birth outcomes, but population-based epidemiologic evidence remains limited and inconsistent. METHODS We conducted a time-series analysis to estimate short-term associations between community-level seasonal influenza activity and daily counts of preterm births in Atlanta, United States from October 17, 2010 to July 10, 2017. We defined weekly influenza exposures four ways: (1) percent test-positive from virologic surveillance, (2) percent of patients with symptoms of influenza-like illness (ILI) in outpatient settings, (3) a composite measure of percent test-positive and ILI, and (4) influenza hospitalization rates. We used Poisson log-linear models to estimate associations, adjusting for time-varying confounders and ongoing at-risk pregnancies. We further examined associations by influenza type and exposure lags and effect modification by maternal characteristics. RESULTS We studied a total of 316,253 births. We found consistent positive associations between influenza activity and preterm birth across different exposure measures and exposure lags. An interquartile range increase in a composite measure of ILI activity and percent test-positive was associated with a 1.014 (95% confidence interval: 1.001-1.027) increase in preterm birth during the same week. In stratified analyses, associations were more pronounced among married, non-Black, and Hispanic pregnant people. CONCLUSION Periods of high influenza activity were associated with an increased risk of preterm birth.
Collapse
Affiliation(s)
- Xiaping Zheng
- From the Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University
| | - Tingyu Wang
- From the Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University
| | - Hua Hao
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University
| | - Rohan R D'Souza
- From the Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University
| | | | - Joshua L Warren
- Department of Biostatistics, School of Public Health, Yale University
| | | | - Howard H Chang
- From the Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University
| |
Collapse
|
4
|
Yalcinkaya A, Yalcinkaya R, Sardh F, Landegren N. Immune dynamics throughout life in relation to sex hormones and perspectives gained from gender-affirming hormone therapy. Front Immunol 2025; 15:1501364. [PMID: 39885993 PMCID: PMC11779622 DOI: 10.3389/fimmu.2024.1501364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 12/23/2024] [Indexed: 02/01/2025] Open
Abstract
Biological sex is closely associated with the properties and extent of the immune response, with males and females showing different susceptibilities to diseases and variations in immunity. Androgens, predominantly in males, generally suppress immune responses, while estrogens, more abundant in females, tend to enhance immunity. It is also established that sex hormones at least partially explain sex biases in different diseases, particularly autoimmune diseases in females. These differences are influenced by hormonal, genetic, and environmental factors, and vary throughout life stages. The advent of gender-affirming hormone therapy offers a novel opportunity to study the immunological effects of sex hormones. Despite the limited studies on this topic, available research has revealed that testosterone therapy in transgender men may suppress certain immune functions, such as type I interferon responses, while increasing inflammation markers like TNF-α. Transgender women on estrogen therapy also experience alterations in coagulation-related and inflammatory characteristics. Furthermore, other possible alterations in immune regulation can be inferred from the assessment of inflammatory and autoimmune markers in transgender individuals receiving hormone therapy. Understanding the complex interactions between sex hormones and the immune system, particularly through the unique perspective offered by gender-affirming hormone therapies, may facilitate the development of targeted therapies for infections and autoimmune diseases while also improving healthcare outcomes for transgender individuals. Here we review immune dynamics throughout life in both sexes and provide a summary of novel findings drawn from studies exploring gender-affirming hormone therapy.
Collapse
Affiliation(s)
- Ahmet Yalcinkaya
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- Department of Medical Biochemistry, Hacettepe University Faculty of Medicine, Ankara, Türkiye
| | - Rumeysa Yalcinkaya
- Department of Pediatric Infectious Diseases, Ankara Etlik City Hospital, Ankara, Türkiye
| | - Fabian Sardh
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- Center for Molecular Medicine, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Nils Landegren
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- Center for Molecular Medicine, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
5
|
Cao J, Li Y, Xue F, Sheng Z, Liu L, Zhang Y, Wang L, Zeng L, Jiang Y, Fan D, Li F, An J. Case study: May human norovirus infection be associated with premature delivery? Virol Sin 2024; 39:951-962. [PMID: 39490793 PMCID: PMC11738789 DOI: 10.1016/j.virs.2024.10.004] [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: 02/06/2024] [Accepted: 10/18/2024] [Indexed: 11/05/2024] Open
Abstract
Human norovirus (HuNoV) is the leading cause of acute gastroenteritis. The varying severity of chronic infection in patients with underlying immune deficiencies poses additional burdens on public health. However, the potential effects of HuNoV infection during pregnancy, a specific immune perturbed state, have been rarely reported. Recently, four cases of HuNoV-infected patients in the late stages of pregnancy were admitted to the Guangzhou Women and Children's Medical Center, and premature rupture of membranes as primary adverse outcome was observed in these cases. Samples of fetal accessory tissue were collected from two of these cases at delivery to explore the potential pathogenesis. Pathological analysis showed placental malperfusion in both maternal and fetal vascular, while a decrease in vessels was not observed in villi of placenta. There was obvious pathological change in the chorion of fetal membrane, accompanied by a tendency of Th-1 immune bias. Notably, aggregation of M2 macrophages was observed in the chorion of the fetal membrane, potentially recruited for tissue repair. Next-generation sequencing showed minimal changes in immune pathways within placenta tissue. A gene panel associated with immunosuppression was identified in the fetal membrane of HuNoV-infected women compared to those of normal parturient. Taken together, this study provides clues for the association between the HuNoV and premature delivery, which requires the attention of the clinicians.
Collapse
Affiliation(s)
- Jiaying Cao
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Department of Clinical Laboratory, The Fifth People's Hospital of Jinan, Jinan, 250022, China
| | - Yuetong Li
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Feiyang Xue
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Ziyang Sheng
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Libo Liu
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yingying Zhang
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Lele Wang
- Department of Obstetrics & Gynaecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510180, China
| | - Liang Zeng
- Department of Pathology, Guangzhou Women and Children Medical Center, Guangzhou Medical University, Guangzhou, 510180, China
| | - Yanmin Jiang
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Dongying Fan
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Fang Li
- Department of Obstetrics & Gynaecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510180, China.
| | - Jing An
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
6
|
Chronopoulos J, Pernet E, Tran KA, McGovern TK, Morozan A, Wang S, Tsai O, Makita K, Divangahi M, Martin JG. Pregnancy enhances antiviral immunity independent of type I IFN but dependent on IL-17-producing γδ + T cells in the nasal mucosa. SCIENCE ADVANCES 2024; 10:eado7087. [PMID: 39331716 PMCID: PMC11430450 DOI: 10.1126/sciadv.ado7087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 08/23/2024] [Indexed: 09/29/2024]
Abstract
Pregnancy is associated with profound changes in immunity. However, pregnancy-related respiratory immune adaptations in response to influenza infection and their impact on disease severity remain unclear. Here, we describe, in a preclinical model of mid-gestation pregnancy, a mechanism of enhanced host defense against influenza A virus (IAV) localized to the nasal cavity that limits viral replication and reduces the magnitude of intrapulmonary immune responses. Consequently, the pregnant mice show reduced pulmonary pathology and preserved airway function after IAV infection. The early restriction of viral replication is independent of type I interferon (IFN) but dependent on increased antimicrobial peptides (AMPs) driven by interleukin-17+ (IL-17+) γδ+ T cells within the nasal passages. This pathway of host defense against IAV infection in the upper airways during pregnancy restricts early viral infection and prevents virus dissemination into the lung supporting maternal fitness.
Collapse
Affiliation(s)
- Julia Chronopoulos
- Department of Medicine, McGill University, Montreal, Quebec, Canada
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Erwan Pernet
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Department of Medical Biology, Université du Québec à Trois-Rivières, Quebec, Canada
| | - Kim A. Tran
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Department of Pathology, McGill University, Montreal, Quebec, Canada
| | - Toby K. McGovern
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Arina Morozan
- Department of Medicine, McGill University, Montreal, Quebec, Canada
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Sadie Wang
- Department of Medicine, McGill University, Montreal, Quebec, Canada
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Oscar Tsai
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
| | - Kosuke Makita
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Maziar Divangahi
- Department of Medicine, McGill University, Montreal, Quebec, Canada
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Department of Pathology, McGill University, Montreal, Quebec, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
| | - James G. Martin
- Department of Medicine, McGill University, Montreal, Quebec, Canada
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
7
|
Lissa SM, Lapinski BA, Graf ME, Reda S, Debur MDC, Presibella M, Pereira LA, de Carvalho NS, Carvalho de Oliveira J, Raboni SM, Nogueira MB. A Retrospective Cross-Sectional Analysis of Viral SARI in Pregnant Women in Southern Brazil. Microorganisms 2024; 12:1555. [PMID: 39203399 PMCID: PMC11356489 DOI: 10.3390/microorganisms12081555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 06/28/2024] [Accepted: 07/02/2024] [Indexed: 09/03/2024] Open
Abstract
Pregnant women (PW) are at a higher risk of diseases and hospitalization from viral respiratory infections, particularly influenza and SARS-CoV-2, due to cardiopulmonary and immunological changes. This study assessed the impact of viral respiratory infections on PW hospitalized with severe acute respiratory infection (SARI) prior to the COVID-19 pandemic. It is a cross-sectional study with 42 PW and 85 non-pregnant women (NPW) admitted with SARI to two tertiary hospitals between January 2015 and December 2019. The rates of virus prevalence, SARI hospitalization, length of hospital stay, oxygen supplementation, intensive care unit (ICU) admission, and death were comparable between PW and NPW. A multivariate analysis showed that PW had a higher rate of viral SARI hospitalizations (OR = 2.37; 95% CI = 1.02-5.48) as compared to NPW, with the influenza virus being the most prevalent (aOR = 7.58; 95% CI = 1.53-37.66). The length of hospital stays (aOR = 0.83; 95% CI = 0.73-0.95) and admissions to the ICU (aOR = 0.028; 95% CI = 0.004-0.25) were lower in PW as compared to hospitalized NPW. The influenza virus had a greater impact on the frequency of SARI in the group of PW, and these had a better outcome than NPW due to the earlier antiviral treatment they received.
Collapse
Affiliation(s)
- Sonia Maria Lissa
- Postgraduate Program in Tocogynecology and Women’s Health, Federal University of Parana Universidade Federal do Paraná, Curitiba 80060-900, Brazil; (S.M.L.)
| | - Bruna Amaral Lapinski
- Postgraduate Program in Internal Medicine and Health Science, Federal University of Parana, Curitiba 80060-900, Brazil;
| | - Maria Ester Graf
- Epidemiology Division, Hospital do Trabalhador, Curitiba 81050-000, Brazil;
| | - Somaia Reda
- Gynecology and Obstetrics Division, Hospital do Trabalhador, Curitiba 81050-000, Brazil;
| | - Maria do Carmo Debur
- Public Health Laboratory, São José dos Pinhais 83060-500, Brazil; (M.d.C.D.); (M.P.)
| | - Mayra Presibella
- Public Health Laboratory, São José dos Pinhais 83060-500, Brazil; (M.d.C.D.); (M.P.)
| | - Luciane Aparecida Pereira
- Virology Laboratory, Hospital de Clínicas, Federal University of Parana, Curitiba 80060-900, Brazil;
| | - Newton Sérgio de Carvalho
- Postgraduate Program in Tocogynecology and Women’s Health, Federal University of Parana Universidade Federal do Paraná, Curitiba 80060-900, Brazil; (S.M.L.)
- Department of Tocogynecology, Federal University of Parana, Curitiba 80060-900, Brazil
| | | | - Sonia Mara Raboni
- Infectious Diseases Division, Hospital de Clínicas, Federal University of Parana, Curitiba 80060-900, Brazil;
| | - Meri Bordignon Nogueira
- Postgraduate Program in Tocogynecology and Women’s Health, Federal University of Parana Universidade Federal do Paraná, Curitiba 80060-900, Brazil; (S.M.L.)
- Virology Laboratory, Hospital de Clínicas, Federal University of Parana, Curitiba 80060-900, Brazil;
| |
Collapse
|
8
|
Chen S, Wang W, Yan G, Liu M, Li M, Chen P, Ma Q, Zhang J, Tang Y, Zhou L, You D. Amniotic Fluid Proteomics Analysis and In Vitro Validation to Identify Potential Biomarkers of Preterm Birth. Reprod Sci 2024; 31:2032-2042. [PMID: 38453771 PMCID: PMC11217130 DOI: 10.1007/s43032-024-01457-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 01/08/2024] [Indexed: 03/09/2024]
Abstract
This study aimed to investigate the regulation of amniotic fibroblast (AFC) function by vitamin K-dependent protein Z (PROZ) during preterm birth (PTB) and its potential role in adverse pregnancy outcomes. Proteomic samples were collected from amniotic fluid in the second trimester, and AFC were isolated from the amniotic membrane and cultured in vitro. The expression of extracellular and intracellular PROZ in AFC was modulated, and their biological properties and functions were evaluated. Clinical analysis revealed a significant upregulation of PROZ expression in amniotic fluid from preterm pregnant women. In vitro experiments demonstrated that PROZ stimulated AFC migration, enhanced their proliferative capacity, and reduced collagen secretion. Overexpression of PROZ further enhanced cell migration and proliferation, while knockdown of PROZ had the opposite effect. PROZ plays a crucial role in promoting the proliferation and migration of amniotic membrane fibroblasts. Increased PROZ expression in amniotic fluid is associated with the occurrence of PTB. These findings shed light on the potential involvement of PROZ in adverse pregnancy outcomes and provide a basis for further research on its regulatory mechanisms during PTB.
Collapse
Affiliation(s)
- Siguo Chen
- Yunnan Provincial Key Laboratory of Public Health and Biosafety & School of Public Health, Kunming Medical University, Kunming, Yunnan, 650000, China
| | - Weizhou Wang
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, Yunnan, China
| | - Guanghong Yan
- Yunnan Provincial Key Laboratory of Public Health and Biosafety & School of Public Health, Kunming Medical University, Kunming, Yunnan, 650000, China
| | - Mengmei Liu
- Yunnan Provincial Key Laboratory of Public Health and Biosafety & School of Public Health, Kunming Medical University, Kunming, Yunnan, 650000, China
| | - Min Li
- Yunnan Provincial Key Laboratory of Public Health and Biosafety & School of Public Health, Kunming Medical University, Kunming, Yunnan, 650000, China
| | - Ping Chen
- Yunnan Provincial Key Laboratory of Public Health and Biosafety & School of Public Health, Kunming Medical University, Kunming, Yunnan, 650000, China
| | - Qingyan Ma
- Yunnan Provincial Key Laboratory of Public Health and Biosafety & School of Public Health, Kunming Medical University, Kunming, Yunnan, 650000, China
| | - Jinman Zhang
- Department of Medical Genetics, First People's Hospital of Yunnan Province, Kunming, 650034, Yunnan, China
| | - Ying Tang
- The Electron Microscopy Laboratory, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650000, Yunnan, China
| | - Linglin Zhou
- Department of Obstetrics, Gejiu City People's Hospital, Gejiu, 661000, Yunnan, China
| | - Dingyun You
- Yunnan Provincial Key Laboratory of Public Health and Biosafety & School of Public Health, Kunming Medical University, Kunming, Yunnan, 650000, China.
| |
Collapse
|
9
|
Creisher PS, Klein SL. Pathogenesis of viral infections during pregnancy. Clin Microbiol Rev 2024; 37:e0007323. [PMID: 38421182 PMCID: PMC11237665 DOI: 10.1128/cmr.00073-23] [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] [Indexed: 03/02/2024] Open
Abstract
SUMMARYViral infections during pregnancy are associated with significant adverse perinatal and fetal outcomes. Pregnancy is a unique immunologic and physiologic state, which can influence control of virus replication, severity of disease, and vertical transmission. The placenta is the organ of the maternal-fetal interface and provides defense against microbial infection while supporting the semi-allogeneic fetus via tolerogenic immune responses. Some viruses, such as cytomegalovirus, Zika virus, and rubella virus, can breach these defenses, directly infecting the fetus and having long-lasting consequences. Even without direct placental infection, other viruses, including respiratory viruses like influenza viruses and severe acute respiratory syndrome coronavirus 2, still cause placental damage and inflammation. Concentrations of progesterone and estrogens rise during pregnancy and contribute to immunological adaptations, placentation, and placental development and play a pivotal role in creating a tolerogenic environment at the maternal-fetal interface. Animal models, including mice, nonhuman primates, rabbits, and guinea pigs, are instrumental for mechanistic insights into the pathogenesis of viral infections during pregnancy and identification of targetable treatments to improve health outcomes of pregnant individuals and offspring.
Collapse
Affiliation(s)
- Patrick S Creisher
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Sabra L Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
10
|
Wang J, Liu W, Zhuang Y, Yang J, Zhao Y, Hong A, Du J, Kong H, Wang J, Jiang Y, Wang Y. Influenza A virus infection disrupts the function of syncytiotrophoblast cells and contributes to adverse pregnancy outcomes. J Med Virol 2024; 96:e29687. [PMID: 38783821 DOI: 10.1002/jmv.29687] [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: 02/12/2024] [Revised: 04/23/2024] [Accepted: 05/12/2024] [Indexed: 05/25/2024]
Abstract
Pregnancy heightens susceptibility to influenza A virus (IAV) infection, thereby increasing the risk of severe pneumonia and maternal mortality. It also raises the chances of adverse outcomes in offspring, such as fetal growth restriction, preterm birth, miscarriage, and stillbirth in offsprings. However, the underlying mechanisms behind these effects remain largely unknown. Syncytiotrophoblast cells, crucial in forming the placental barrier, nutrient exchange and hormone secretion, have not been extensively studied for their responses to IAV. In our experiment, we used Forskolin-treated BeWo cells to mimic syncytiotrophoblast cells in vitro, and infected them with H1N1, H5N1 and H7N9 virus stains. Our results showed that syncytiotrophoblast cells, with their higher intensity of sialic acid receptors, strongly support IAV infection and replication. Notably, high-dose viral infection and prolonged exposure resulted in a significant decrease in fusion index, as well as gene and protein expression levels associated with trophoblast differentiation, β-human chorionic gonadotropin secretion, estrogen and progesterone biosynthesis, and nutrient transport. In pregnant BALB/c mice infected with the H1N1 virus, we observed significant decreases in trophoblast differentiation and hormone secretion gene expression levels. IAV infection also resulted in preterm labor, fetal growth restriction, and increased maternal and fetal morbidity and mortality. Our findings indicate that IAV infection in syncytiotrophoblastic cells can result in adverse pregnancy outcomes by altering trophoblast differentiation, suppressing of β-hCG secretion, and disrupting placental barrier function.
Collapse
Affiliation(s)
- Jiao Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenyu Liu
- State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, China
| | - Yichao Zhuang
- State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, China
| | - Jiaxin Yang
- State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, China
| | - Yetian Zhao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Aihui Hong
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jingjing Du
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Huihui Kong
- State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, China
| | - Jingfei Wang
- State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, China
| | - Yongping Jiang
- State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, China
| | - Yan Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
11
|
Özer Aslan İ, Doğan K, Kural A, Baghaki S, Helvaci N, Ekin M, Yaşar L. Is it possible to identify COVID-19 infection-related biomarkers during pregnancy?: A prospective study in Turkish population. Medicine (Baltimore) 2024; 103:e38062. [PMID: 38728492 PMCID: PMC11081580 DOI: 10.1097/md.0000000000038062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 04/08/2024] [Indexed: 05/12/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) has raised concerns about the potential complications it may cause in pregnant women. Therefore, biomarkers that can predict the course of COVID-19 in pregnant women may be of great benefit as they would provide valuable insights into the prognosis and, thus, the management of the disease. In this context, the objective of this study is to identify the biomarkers that can predict COVID-19 progression in pregnant women, focusing on composite hemogram parameters and systemic inflammatory and spike markers. The population of this single-center prospective case-control study consisted of all consecutive pregnant women with single healthy fetuses who tested positive for COVID-19 and who were admitted to Bakirköy Dr Sadi Konuk Training and Research Hospital in Istanbul, Turkey, a COVID-19 referral hospital, between April 2020 and March 2021, with an obstetric indication, during their second or third trimester. The control group consisted of consecutive pregnant women with a single healthy fetus who were admitted to the same hospital within the same date range, had demographic and obstetric characteristics matching the patient group, but tested negative for COVID-19. The patient and control groups were compared in terms of platelet-to-lymphocyte ratio (PLR), platelet-to-neutrophil ratio (PNR), and neutrophil-to-lymphocyte ratio (NLR), and systemic inflammatory and spike markers, including C-reactive protein (CRP), interleukin-6 (IL-6), interleukin-10 (IL-10), cluster of differentiation 26 (CD26), and B7 homolog 4 (B7H4). There were 45 (51.1%) and 43 (48.8%) pregnant women in the patient and control groups, respectively. There was no significant difference between the groups in demographic and obstetric characteristics (P > .05). The PNR, PLR, and CRP values were significantly higher in the patient group than in the control group (P < .05). On the other hand, there was no significant difference between the groups in IL-6, IL-10, CD26, and B7H4 levels (P > .05). The findings of our study showed that specific inflammatory markers, such as CRP, PLR, and PNR, can potentially predict the course of COVID-19 in pregnant women. However, more comprehensive, well-controlled studies are needed to corroborate our study's findings and investigate other potential inflammatory markers.
Collapse
Affiliation(s)
- İlke Özer Aslan
- Department of Obstetrics and Gynecology, Faculty of Medicine, Tekirdag Namik Kemal University, Tekirdag, Türkiye
| | - Keziban Doğan
- Department of Obstetrics and Gynecology, University of Health Sciences, Bakirkoy Dr.Sadi Konuk Training and Research Hospital, Istanbul, Türkiye
| | - Alev Kural
- Department of Biochemistry, University of Health Sciences, Bakirkoy Dr.Sadi Konuk Training and Research Hospital, Istanbul, Türkiye
| | - Sema Baghaki
- Department of Obstetrics and Gynecology, University of Pittsburgh, PA
| | - Nazli Helvaci
- Department of Biochemistry, Hamidiye Faculty of Medicine, University of Health Sciences, Istanbul, Türkiye
| | - Murat Ekin
- Department of Obstetrics and Gynecology, University of Health Sciences, Bakirkoy Dr.Sadi Konuk Training and Research Hospital, Istanbul, Türkiye
| | - Levent Yaşar
- Department of Obstetrics and Gynecology, University of Health Sciences, Bakirkoy Dr.Sadi Konuk Training and Research Hospital, Istanbul, Türkiye
| |
Collapse
|
12
|
Mohapatra L, Patel G, Tripathi AS, Alka, Mishra D, Parida SK, Yasir M, Maurya RK. Swine Flu. RISING CONTAGIOUS DISEASES 2024:50-65. [DOI: 10.1002/9781394188741.ch6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
13
|
Creisher PS, Parish MA, Lei J, Liu J, Perry JL, Campbell AD, Sherer ML, Burd I, Klein SL. Suppression of progesterone by influenza A virus mediates adverse maternal and fetal outcomes in mice. mBio 2024; 15:e0306523. [PMID: 38190129 PMCID: PMC10865978 DOI: 10.1128/mbio.03065-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 12/06/2023] [Indexed: 01/09/2024] Open
Abstract
Influenza A virus infection during pregnancy can cause adverse maternal and fetal outcomes but the mechanism responsible remains elusive. Infection of outbred mice with 2009 H1N1 at embryonic day (E) 10 resulted in significant maternal morbidity, placental tissue damage and inflammation, fetal growth restriction, and developmental delays that lasted through weaning. Restriction of pulmonary virus replication was not inhibited during pregnancy, but infected dams had suppressed circulating and placental progesterone (P4) concentrations that were caused by H1N1-induced upregulation of pulmonary cyclooxygenase (COX)-1-, but not COX-2-, dependent synthesis and secretion of prostaglandin (PG) F2α. Treatment with 17-α-hydroxyprogesterone caproate (17-OHPC), a synthetic progestin that is safe to use in pregnancy, ameliorated the adverse maternal and fetal outcomes from H1N1 infection and prevented placental cell death and inflammation. These findings highlight the therapeutic potential of progestin treatments for influenza during pregnancy.IMPORTANCEPregnant individuals are at risk of severe outcomes from both seasonal and pandemic influenza A viruses. Influenza infection during pregnancy is associated with adverse fetal outcomes at birth and adverse consequences for offspring into adulthood. When outbred dams, with semi-allogenic fetuses, were infected with 2009 H1N1, in addition to pulmonary virus replication, lung damage, and inflammation, the placenta showed evidence of transient cell death and inflammation that was mediated by increased activity along the arachidonic acid pathway leading to suppression of circulating progesterone. Placental damage and suppressed progesterone were associated with detrimental effects on perinatal growth and developmental delays in offspring. Treatment of H1N1-infected pregnant mice with 17-OHPC, a synthetic progestin treatment that is safe to use in pregnancy, prevented placental damage and inflammation and adverse fetal outcomes. This novel therapeutic option for the treatment of influenza during pregnancy should be explored clinically.
Collapse
Affiliation(s)
- Patrick S. Creisher
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Maclaine A. Parish
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Jun Lei
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jin Liu
- Department of Gynecology and Obstetrics, Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jamie L. Perry
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Ariana D. Campbell
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Morgan L. Sherer
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Irina Burd
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Sabra L. Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
14
|
Burucúa MM, Risalde MA, Cheuquepán FA, Quintana S, Pérez SE, Cantón GJ, Moore DP, Odeón AC, Agulló-Ros I, Scioli MV, Barbeito C, Morrell EL, Marín MS. Transplacental infection by bovine alphaherpesvirus type 1 induces protein expression of COX-2, iNOS and inflammatory cytokines in fetal lungs and placentas. Vet Microbiol 2023; 287:109912. [PMID: 37952263 DOI: 10.1016/j.vetmic.2023.109912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 11/01/2023] [Accepted: 11/04/2023] [Indexed: 11/14/2023]
Abstract
Bovine alphaherpesvirus type 1 (BoAHV-1) is associated with respiratory and reproductive syndromes. Until present the immunologic mechanisms involved in BoAHV-1 abortion are partially known. We studied key elements of the innate immune response in the placentas and fetal lungs from cattle experimentally-inoculated with BoAHV-1. These tissues were analyzed by histopathology. Furthermore, virus identification was performed by qPCR and the expression of the inflammatory cytokines such as tumor necrosis factor-alpha, interleukin 1-alpha and inflammatory mediators like inducible nitric oxide synthase and cyclooxeganse-2 was evaluated by immunohistochemistry. The viral transplacental infection was confirmed by the detection of BoAHV-1 by qPCR in the placenta and fetal organs, which revealed mild inflammatory lesions. Inducible nitric oxide synthase immunolabelling was high in the lungs of infected fetuses and placentas, as well as for tumor necrosis factor-alpha in the pulmonary parenchyma and cyclooxeganse-2 in fetal annexes. However, the expression of interleukin 1-alpha was weak in these organs. To our knowledge, this is the first study that provides strong evidence of an early immune response to BoAHV-1 infection in the conceptus. Advances in the knowledge of the complex immunological interactions at the feto-maternal unit during BoAHV-1 infection are needed to clarify the pathogenesis of abortion.
Collapse
Affiliation(s)
- Mercedes M Burucúa
- Instituto de Innovación para la Producción Agropecuaria y el Desarrollo Sostenible (IPADS Balcarce), INTA-CONICET, Balcarce, Buenos Aires, Argentina
| | - María A Risalde
- Departamento de Anatomía y Anatomía Patológica Comparadas y Toxicología, Grupo de Investigación GISAZ, UIC Zoonosis y Enfermedades Emergentes ENZOEM, Universidad de Córdoba, Córdoba, Spain
| | - Felipe A Cheuquepán
- Instituto de Innovación para la Producción Agropecuaria y el Desarrollo Sostenible (IPADS Balcarce), INTA-CONICET, Balcarce, Buenos Aires, Argentina
| | - Silvina Quintana
- Instituto de Investigaciones de Producción, Sanidad y Ambiente (IIPROSAM), FCEyN, UNMDP-CONICET, Mar del Plata, Buenos Aires, Argentina; Instituto de Biología Molecular Aplicada, Mar del Plata, Buenos Aires, Argentina
| | - Sandra E Pérez
- Centro de Investigaciones Veterinarias de Tandil (CIVETAN) - CONICET, Facultad de Ciencias Veterinarias, UNCPBA, Tandil, Buenos Aires, Argentina
| | - Germán J Cantón
- Instituto de Innovación para la Producción Agropecuaria y el Desarrollo Sostenible (IPADS Balcarce), INTA-CONICET, Balcarce, Buenos Aires, Argentina
| | - Dadin P Moore
- Instituto de Innovación para la Producción Agropecuaria y el Desarrollo Sostenible (IPADS Balcarce), INTA-CONICET, Balcarce, Buenos Aires, Argentina; Facultad de Ciencias Agrarias, UNMdP, Balcarce, Buenos Aires, Argentina
| | - Anselmo C Odeón
- Facultad de Ciencias Agrarias, UNMdP, Balcarce, Buenos Aires, Argentina
| | - Irene Agulló-Ros
- Departamento de Anatomía y Anatomía Patológica Comparadas y Toxicología, Grupo de Investigación GISAZ, UIC Zoonosis y Enfermedades Emergentes ENZOEM, Universidad de Córdoba, Córdoba, Spain
| | - María Valeria Scioli
- Instituto de Innovación para la Producción Agropecuaria y el Desarrollo Sostenible (IPADS Balcarce), INTA-CONICET, Balcarce, Buenos Aires, Argentina
| | - Claudio Barbeito
- Laboratorio de Histología y Embriología Descriptiva Experimental y Comparada (LHYEDEC), Facultad de Ciencias Veterinarias, UNLP, CONICET, Buenos Aires, Argentina
| | - Eleonora L Morrell
- Instituto de Innovación para la Producción Agropecuaria y el Desarrollo Sostenible (IPADS Balcarce), INTA-CONICET, Balcarce, Buenos Aires, Argentina.
| | - Maia S Marín
- Instituto de Innovación para la Producción Agropecuaria y el Desarrollo Sostenible (IPADS Balcarce), INTA-CONICET, Balcarce, Buenos Aires, Argentina
| |
Collapse
|
15
|
Creisher PS, Parish MA, Lei J, Liu J, Perry JL, Campbell AD, Sherer ML, Burd I, Klein SL. Suppression of progesterone by influenza A virus mediates adverse maternal and fetal outcomes in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.11.557146. [PMID: 37745453 PMCID: PMC10515826 DOI: 10.1101/2023.09.11.557146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Influenza A virus infection during pregnancy can cause adverse maternal and fetal outcomes, but the mechanism responsible remains elusive. Infection of outbred mice with 2009 H1N1 at embryonic day (E) 10 resulted in significant maternal morbidity, placental tissue damage and inflammation, fetal growth restriction, and developmental delays that lasted through weaning. Restriction of pulmonary virus replication was not inhibited during pregnancy, but infected dams had suppressed circulating and placental progesterone (P4) concentrations that were caused by H1N1-induced upregulation of pulmonary cyclooxygenase (COX)-1, but not COX-2-, dependent synthesis and secretion of prostaglandin (PG) F2α. Treatment with 17-α-hydroxyprogesterone caproate (17-OHPC), a synthetic progestin that is safe to use in pregnancy, ameliorated the adverse maternal and fetal outcomes from H1N1 infection and prevented placental cell death and inflammation. These findings highlight the therapeutic potential of progestin treatments for influenza during pregnancy.
Collapse
|
16
|
Creisher PS, Perry JL, Zhong W, Lei J, Mulka KR, Ryan WH, Zhou R, Akin EH, Liu A, Mitzner W, Burd I, Pekosz A, Klein SL. Adverse outcomes in SARS-CoV-2-infected pregnant mice are gestational age-dependent and resolve with antiviral treatment. J Clin Invest 2023; 133:e170687. [PMID: 37581940 PMCID: PMC10575736 DOI: 10.1172/jci170687] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 08/10/2023] [Indexed: 08/17/2023] Open
Abstract
SARS-CoV-2 infection during pregnancy is associated with severe COVID-19 and adverse fetal outcomes, but the underlying mechanisms remain poorly understood. Moreover, clinical studies assessing therapeutics against SARS-CoV-2 in pregnancy are limited. To address these gaps, we developed a mouse model of SARS-CoV-2 infection during pregnancy. Outbred CD1 mice were infected at E6, E10, or E16 with a mouse-adapted SARS-CoV-2 (maSCV2) virus. Outcomes were gestational age-dependent, with greater morbidity, reduced antiviral immunity, greater viral titers, and impaired fetal growth and neurodevelopment occurring with infection at E16 (third trimester equivalent) than with infection at either E6 (first trimester equivalent) or E10 (second trimester equivalent). To assess the efficacy of ritonavir-boosted nirmatrelvir, which is recommended for individuals who are pregnant with COVID-19, we treated E16-infected dams with mouse-equivalent doses of nirmatrelvir and ritonavir. Treatment reduced pulmonary viral titers, decreased maternal morbidity, and prevented offspring growth restriction and neurodevelopmental impairments. Our results highlight that severe COVID-19 during pregnancy and fetal growth restriction is associated with heightened virus replication in maternal lungs. Ritonavir-boosted nirmatrelvir mitigated maternal morbidity along with fetal growth and neurodevelopment restriction after SARS-CoV-2 infection. These findings prompt the need for further consideration of pregnancy in preclinical and clinical studies of therapeutics against viral infections.
Collapse
Affiliation(s)
- Patrick S. Creisher
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Jamie L. Perry
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Weizhi Zhong
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Jun Lei
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Kathleen R. Mulka
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - W. Hurley Ryan
- Department of Environmental Health and Engineering, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Ruifeng Zhou
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Elgin H. Akin
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Anguo Liu
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Wayne Mitzner
- Department of Environmental Health and Engineering, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Irina Burd
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of Environmental Health and Engineering, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Sabra L. Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
17
|
Creisher PS, Perry JL, Zhong W, Lei J, Mulka KR, Ryan H, Zhou R, Akin EH, Liu A, Mitzner W, Burd I, Pekosz A, Klein SL. Adverse outcomes in SARS-CoV-2 infected pregnant mice are gestational age-dependent and resolve with antiviral treatment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.23.533961. [PMID: 36993658 PMCID: PMC10055386 DOI: 10.1101/2023.03.23.533961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
SARS-CoV-2 infection during pregnancy is associated with severe COVID-19 and adverse fetal outcomes, but the underlying mechanisms remain poorly understood. Moreover, clinical studies assessing therapeutics against SARS-CoV-2 in pregnancy are limited. To address these gaps, we developed a mouse model of SARS-CoV-2 infection during pregnancy. Outbred CD1 mice were infected at embryonic day (E) 6, E10, or E16 with a mouse adapted SARS-CoV-2 (maSCV2) virus. Outcomes were gestational age-dependent, with greater morbidity, reduced anti-viral immunity, greater viral titers, and more adverse fetal outcomes occurring with infection at E16 (3rd trimester-equivalent) than with infection at either E6 (1st trimester-equivalent) or E10 (2nd trimester-equivalent). To assess the efficacy of ritonavir-boosted nirmatrelvir (recommended for pregnant individuals with COVID-19), we treated E16-infected dams with mouse equivalent doses of nirmatrelvir and ritonavir. Treatment reduced pulmonary viral titers, decreased maternal morbidity, and prevented adverse offspring outcomes. Our results highlight that severe COVID-19 during pregnancy and adverse fetal outcomes are associated with heightened virus replication in maternal lungs. Ritonavir-boosted nirmatrelvir mitigated adverse maternal and fetal outcomes of SARS-CoV-2 infection. These findings prompt the need for further consideration of pregnancy in preclinical and clinical studies of therapeutics against viral infections.
Collapse
|
18
|
Drury NL, Mustapha T, Shore RA, Zhao J, Wright GA, Hoffmann AR, Talcott SU, Regan A, Tighe RM, Zhang R, Johnson NM. Maternal exposure to ultrafine particles enhances influenza infection during pregnancy. Part Fibre Toxicol 2023; 20:11. [PMID: 37069680 PMCID: PMC10106898 DOI: 10.1186/s12989-023-00521-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 04/01/2023] [Indexed: 04/19/2023] Open
Abstract
BACKGROUND Interactions between air pollution and infectious agents are increasingly recognized and critical to identify, especially to protect vulnerable populations. Pregnancy represents a vulnerable period for influenza infection and air pollution exposure, yet interactions during pregnancy remain unclear. Maternal exposure to ultrafine particles (UFPs, [Formula: see text] 100 nm diameter), a class of particulate matter ubiquitous in urban environments, elicits unique pulmonary immune responses. We hypothesized that UFP exposure during pregnancy would lead to aberrant immune responses to influenza enhancing infection severity. RESULTS Building from our well-characterized C57Bl/6N mouse model employing daily gestational UFP exposure from gestational day (GD) 0.5-13.5, we carried out a pilot study wherein pregnant dams were subsequently infected with Influenza A/Puerto Rico/8/1934 (PR8) on GD14.5. Findings indicate that PR8 infection caused decreased weight gain in filtered air (FA) and UFP-exposed groups. Co-exposure to UFPs and viral infection led to pronounced elevation in PR8 viral titer and reduced pulmonary inflammation, signifying potential suppression of innate and adaptive immune defenses. Pulmonary expression of the pro-viral factor sphingosine kinase 1 (Sphk1) and pro-inflammatory cytokine interleukin-1β (IL-1 [Formula: see text]) was significantly increased in pregnant mice exposed to UFPs and infected with PR8; expression correlated with higher viral titer. CONCLUSIONS Results from our model provide initial insight into how maternal UFP exposure during pregnancy enhances respiratory viral infection risk. This model is an important first step in establishing future regulatory and clinical strategies for protecting pregnant women exposed to UFPs.
Collapse
Affiliation(s)
- Nicholas L Drury
- Department of Environmental and Occupational Health, Texas A&M University, 212 Adriance Lab Rd, 1266 TAMU, College Station, TX, 77843, USA
- Department of Nutrition, Texas A&M University, College Station, TX, 77843, USA
| | - Toriq Mustapha
- Department of Environmental and Occupational Health, Texas A&M University, 212 Adriance Lab Rd, 1266 TAMU, College Station, TX, 77843, USA
| | - Ross A Shore
- Department of Environmental and Occupational Health, Texas A&M University, 212 Adriance Lab Rd, 1266 TAMU, College Station, TX, 77843, USA
| | - Jiayun Zhao
- Department of Chemistry, Texas A&M University, College Station, TX, 77843, USA
| | - Gus A Wright
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, 77843, USA
| | - Aline Rodrigues Hoffmann
- Department of Comparative, Diagnostic, and Population Medicine, University of Florida, Gainesville, FL, 32653, USA
| | - Susanne U Talcott
- Department of Nutrition, Texas A&M University, College Station, TX, 77843, USA
| | - Annette Regan
- School of Nursing and Health Professions, University of San Francisco, Orange County, CA, 92868, USA
| | - Robert M Tighe
- Department of Medicine, Duke University, Durham, NC, 27710, USA
| | - Renyi Zhang
- Department of Chemistry, Texas A&M University, College Station, TX, 77843, USA
- Department of Atmospheric Sciences, Texas A&M University, College Station, TX, 77843, USA
| | - Natalie M Johnson
- Department of Environmental and Occupational Health, Texas A&M University, 212 Adriance Lab Rd, 1266 TAMU, College Station, TX, 77843, USA.
| |
Collapse
|
19
|
Trinh QD, Pham NTK, Takada K, Ushijima H, Komine-Aizawa S, Hayakawa S. Roles of TGF-β1 in Viral Infection during Pregnancy: Research Update and Perspectives. Int J Mol Sci 2023; 24:ijms24076489. [PMID: 37047462 PMCID: PMC10095195 DOI: 10.3390/ijms24076489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 03/25/2023] [Accepted: 03/28/2023] [Indexed: 04/14/2023] Open
Abstract
Transforming growth factor-beta 1 (TGF-β1) is a pleiotropic growth factor playing various roles in the human body including cell growth and development. More functions of TGF-β1 have been discovered, especially its roles in viral infection. TGF-β1 is abundant at the maternal-fetal interface during pregnancy and plays an important function in immune tolerance, an essential key factor for pregnancy success. It plays some critical roles in viral infection in pregnancy, such as its effects on the infection and replication of human cytomegalovirus in syncytiotrophoblasts. Interestingly, its role in the enhancement of Zika virus (ZIKV) infection and replication in first-trimester trophoblasts has recently been reported. The above up-to-date findings have opened one of the promising approaches to studying the mechanisms of viral infection during pregnancy with links to corresponding congenital syndromes. In this article, we review our current and recent advances in understanding the roles of TGF-β1 in viral infection. Our discussion focuses on viral infection during pregnancy, especially in the first trimester. We highlight the mutual roles of viral infection and TGF-β1 in specific contexts and possible functions of the Smad pathway in viral infection, with a special note on ZIKV infection. In addition, we discuss promising approaches to performing further studies on this topic.
Collapse
Affiliation(s)
- Quang Duy Trinh
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Ngan Thi Kim Pham
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Kazuhide Takada
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Hiroshi Ushijima
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Shihoko Komine-Aizawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Satoshi Hayakawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| |
Collapse
|
20
|
Yuan F, Schieber T, Stein TL, Sestak RM, Olson CJ, Chen C, Huber VC, Lechtenberg K, McGill J, Fang Y. Establish a Pregnant Sow–Neonate Model to Assess Maternal Immunity of a Candidate Influenza Vaccine. Vaccines (Basel) 2023; 11:vaccines11030646. [PMID: 36992230 PMCID: PMC10056052 DOI: 10.3390/vaccines11030646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/01/2023] [Accepted: 03/08/2023] [Indexed: 03/15/2023] Open
Abstract
While it is well appreciated that maternal immunity can provide neonatal protection, the contribution of maternal vaccination toward generating such immunity is not well characterized. In our previous work, we created a candidate influenza vaccine using our chimeric hemagglutinin (HA) construct, HA-129. The HA-129 was expressed as part of a whole-virus vaccine that was built on the A/swine/Texas/4199-2/98-H3N2 backbone to generate the recombinant virus TX98-129. The TX98-129 candidate vaccine has the ability to induce broadly protective immune responses against genetically diversified influenza viruses in both mice and nursery pigs. In the current study, we established a pregnant sow–neonate model to evaluate the maternal immunity induced by this candidate vaccine to protect pregnant sows and their neonatal piglets against influenza virus infection. In pregnant sows, the results consistently show that TX98-129 induced a robust immune response against the TX98-129 virus and the parental viruses that were used to construct HA-129. After challenge with a field strain of influenza A virus, a significant increase in antibody titers was observed in vaccinated sows at both 5 and 22 days post challenge (dpc). The challenge virus was detected at a low level in the nasal swab of only one vaccinated sow at 5 dpc. Evaluation of cytokine responses in blood and lung tissue showed that levels of IFN-α and IL-1β were increased in the lung of vaccinated sows at 5 dpc, when compared to unvaccinated pigs. Further analysis of the T-cell subpopulation in PBMCs showed a higher ratio of IFN-γ-secreting CD4+CD8+ and CD8+ cytotoxic T cells in vaccinated sows at 22 dpc after stimulation with either challenge virus or vaccine virus. Finally, we used a neonatal challenge model to demonstrate that vaccine-induced maternal immunity can be passively transferred to newborn piglets. This was observed in the form of both increased antibody titers and deceased viral loads in neonates born from immunized sows. In summary, this study provides a swine model system to evaluate the impact of vaccination on maternal immunity and fetal/neonatal development.
Collapse
Affiliation(s)
- Fangfeng Yuan
- Department of Pathobiology, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA
- Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, KS 66506, USA
| | | | - Tara L. Stein
- Division of Basic Biomedical Sciences, Sanford School of Medicine, The University of South Dakota, Vermillion, SD 57069, USA
| | - Rachel M. Sestak
- Division of Basic Biomedical Sciences, Sanford School of Medicine, The University of South Dakota, Vermillion, SD 57069, USA
| | - Callie J. Olson
- Division of Basic Biomedical Sciences, Sanford School of Medicine, The University of South Dakota, Vermillion, SD 57069, USA
| | - Chi Chen
- Department of Pathobiology, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA
| | - Victor C. Huber
- Division of Basic Biomedical Sciences, Sanford School of Medicine, The University of South Dakota, Vermillion, SD 57069, USA
| | | | - Jodi McGill
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA 50011, USA
| | - Ying Fang
- Department of Pathobiology, University of Illinois Urbana-Champaign, Urbana, IL 61802, USA
- Department of Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, KS 66506, USA
- Correspondence:
| |
Collapse
|
21
|
Creisher PS, Seddu K, Mueller AL, Klein SL. Biological Sex and Pregnancy Affect Influenza Pathogenesis and Vaccination. Curr Top Microbiol Immunol 2023; 441:111-137. [PMID: 37695427 DOI: 10.1007/978-3-031-35139-6_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Males and females differ in the outcome of influenza A virus (IAV) infections, which depends significantly on age. During seasonal influenza epidemics, young children (< 5 years of age) and aged adults (65+ years of age) are at greatest risk for severe disease, and among these age groups, males tend to suffer a worse outcome from IAV infection than females. Following infection with pandemic strains of IAVs, females of reproductive ages (i.e., 15-49 years of age) experience a worse outcome than their male counterparts. Although females of reproductive ages experience worse outcomes from IAV infection, females typically have greater immune responses to influenza vaccination as compared with males. Among females of reproductive ages, pregnancy is one factor linked to an increased risk of severe outcome of influenza. Small animal models of influenza virus infection and vaccination illustrate that immune responses and repair of damaged tissue following IAV infection also differ between the sexes and impact the outcome of infection. There is growing evidence that sex steroid hormones, including estrogens, progesterone, and testosterone, directly impact immune responses during IAV infection and vaccination. Greater consideration of the combined effects of sex and age as biological variables in epidemiological, clinical, and animal studies of influenza pathogenesis is needed.
Collapse
Affiliation(s)
- Patrick S Creisher
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, 615 North Wolfe Street, Baltimore, MD, United States
| | - Kumba Seddu
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, 615 North Wolfe Street, Baltimore, MD, United States
| | - Alice L Mueller
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, 615 North Wolfe Street, Baltimore, MD, United States
| | - Sabra L Klein
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, 615 North Wolfe Street, Baltimore, MD, United States.
| |
Collapse
|
22
|
Otero AM, Antonson AM. At the crux of maternal immune activation: Viruses, microglia, microbes, and IL-17A. Immunol Rev 2022; 311:205-223. [PMID: 35979731 PMCID: PMC9804202 DOI: 10.1111/imr.13125] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Inflammation during prenatal development can be detrimental to neurodevelopmental processes, increasing the risk of neuropsychiatric disorders. Prenatal exposure to maternal viral infection during pregnancy is a leading environmental risk factor for manifestation of these disorders. Preclinical animal models of maternal immune activation (MIA), established to investigate this link, have revealed common immune and microbial signaling pathways that link mother and fetus and set the tone for prenatal neurodevelopment. In particular, maternal intestinal T helper 17 cells, educated by endogenous microbes, appear to be key drivers of effector IL-17A signals capable of reaching the fetal brain and causing neuropathologies. Fetal microglial cells are particularly sensitive to maternally derived inflammatory and microbial signals, and they shift their functional phenotype in response to MIA. Resulting cortical malformations and miswired interneuron circuits cause aberrant offspring behaviors that recapitulate core symptoms of human neurodevelopmental disorders. Still, the popular use of "sterile" immunostimulants to initiate MIA has limited translation to the clinic, as these stimulants fail to capture biologically relevant innate and adaptive inflammatory sequelae induced by live pathogen infection. Thus, there is a need for more translatable MIA models, with a focus on relevant pathogens like seasonal influenza viruses.
Collapse
Affiliation(s)
- Ashley M. Otero
- Neuroscience ProgramUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| | - Adrienne M. Antonson
- Department of Animal SciencesUniversity of Illinois Urbana‐ChampaignUrbanaIllinoisUSA
| |
Collapse
|
23
|
Li M, Li A, Huang H, Munson J, Obadan A, Fuller DH, Waldorf KMA. Impact of progesterone on innate immunity and cell death after influenza A virus H1N1 2009 infection of lung and placental cells in vitro. FRONTIERS IN VIROLOGY (LAUSANNE, SWITZERLAND) 2022; 2:953208. [PMID: 36713466 PMCID: PMC9879262 DOI: 10.3389/fviro.2022.953208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The influenza A virus (IAV) 2009 H1N1 pandemic was associated with an increased risk of maternal mortality, preterm birth, and stillbirth. The underlying mechanism for severe maternal lung disease and stillbirth is incompletely understood, but IAV infection is known to activate innate immunity triggering the release of cytokines. Elucidating the impact of progesterone (P4), a key hormone elevated in pregnancy, on the innate immune and inflammatory response to IAV infection is a critical step in understanding the pathogenesis of adverse maternal-fetal outcomes. IAV H1N1 pdm/09 was used to infect cell lines Calu-3 (lung adenoma) and ACH-3P (extravillous trophoblast) with or without P4 (100 nM) at multiplicity of infections (MOI) 0, 0.5, and 3. Cells were harvested at 24 and 48 hours post infection (hpi) and analyzed for cytopathic effects (CPE), replicating virus (TCID50), cytotoxicity (Lactate Dehydrogenase (LDH) assay), and NLRP3 inflammasome activation (caspase-1 activity, fluorometric assay). Activation of antiviral innate immunity was quantified (RT-qPCR, Luminex) by measuring biomarker gene and protein expression of innate immune activation (IFIT1, IFNB), inflammation (IL6), interferon signaling (MXA), chemokines (IL-8, IL-10). Both Calu-3 and ACH-3P were highly permissible to IAV infection at each timepoint as demonstrated by CPE and recovery of replicating virus. In Calu-3, progesterone treatment was associated with a significant increase in cytotoxicity, increased gene expression of IL6, and increased protein expression of IFN-β, IL-6, and IL-18. Conversely, in ACH-3P, progesterone treatment was associated with significantly suppressed cytotoxicity, decreased gene expression of IFNB, IL6 and IL1B, and increased protein expression of IFN-β and IL-6. In both cell lines, caspase-1 activity was significantly decreased after progesterone treatment, indicating NLRP3 inflammasome activation was not underlying the higher cell death in Calu-3. In summary, these data provide evidence that progesterone plays a dual role by ameliorating viral infection in the placenta but exacerbating influenza A virus-associated injury in the lung through nongenomic modulation of the innate immune response.
Collapse
Affiliation(s)
- Miranda Li
- Department of Biological Sciences, Columbia University, New York, New York, United States of America
- Department of Obstetrics and Gynecology, University of Washington School of Medicine, Seattle Washington, United States of America
| | - Amanda Li
- Department of Obstetrics and Gynecology, University of Washington School of Medicine, Seattle Washington, United States of America
- Department of Biology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Hazel Huang
- Department of Obstetrics and Gynecology, University of Washington School of Medicine, Seattle Washington, United States of America
| | - Jeff Munson
- Department of Psychiatry, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Adebimpe Obadan
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
| | - Deborah H. Fuller
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
| | - Kristina M. Adams Waldorf
- Department of Obstetrics and Gynecology, University of Washington School of Medicine, Seattle Washington, United States of America
- Department of Global Health, University of Washington School of Medicine, Seattle, Washington, United States of America
| |
Collapse
|
24
|
Germano C, Messina A, Tavella E, Vitale R, Avellis V, Barboni M, Attini R, Revelli A, Zola P, Manzoni P, Masturzo B. Fetal Brain Damage during Maternal COVID-19: Emerging Hypothesis, Mechanism, and Possible Mitigation through Maternal-Targeted Nutritional Supplementation. Nutrients 2022; 14:3303. [PMID: 36014809 PMCID: PMC9414753 DOI: 10.3390/nu14163303] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/03/2022] [Accepted: 08/09/2022] [Indexed: 11/24/2022] Open
Abstract
The recent outbreak of the novel Coronavirus (SARS-CoV-2 or CoV-2) pandemic in 2019 and the risk of CoV-2 infection during pregnancy led the scientific community to investigate the potential negative effects of Coronavirus infection on pregnancy outcomes and fetal development. In particular, as CoV-2 neurotropism has been demonstrated in adults, recent studies suggested a possible risk of fetal brain damage and fetal brain development impairment, with consequent psychiatric manifestations in offspring of mothers affected by COronaVIrus Disease (COVID) during pregnancy. Through the understanding of CoV-2's pathogenesis and the pathways responsible for cell damage, along with the available data about neurotropic virus attitudes, different strategies have been suggested to lower the risk of neurologic disease in newborns. In this regard, the role of nutrition in mitigating fetal damages related to oxidative stress and the inflammatory environment during viral infection has been investigated, and arginine, n3PUFA, vitamins B1 and B9, choline, and flavonoids were found to be promising in and out of pregnancy. The aim of this review is to provide an overview of the current knowledge on the mechanism of fetal brain damage and the impact of nutrition in reducing inflammation related to worse neurological outcomes in the context of CoV-2 infections during pregnancy.
Collapse
Affiliation(s)
- Chiara Germano
- Department of Maternal, Neonatal and Infant Medicine, University Hospital “Degli Infermi”, 13875 Ponderano, Italy
- Sant’Anna Hospital, Department of Surgical Sciences, University of Turin, 10126 Turin, Italy
| | - Alessandro Messina
- Sant’Anna Hospital, Department of Surgical Sciences, University of Turin, 10126 Turin, Italy
| | - Elena Tavella
- Sant’Anna Hospital, Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy
| | - Raffaele Vitale
- Sant’Anna Hospital, Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy
| | - Vincenzo Avellis
- Sant’Anna Hospital, Department of Public Health and Pediatric Sciences, University of Turin, 10126 Turin, Italy
| | - Martina Barboni
- Sant’Anna Hospital, Department of Surgical Sciences, University of Turin, 10126 Turin, Italy
| | - Rossella Attini
- Sant’Anna Hospital, Department of Surgical Sciences, University of Turin, 10126 Turin, Italy
| | - Alberto Revelli
- Sant’Anna Hospital, Department of Surgical Sciences, University of Turin, 10126 Turin, Italy
| | - Paolo Zola
- Sant’Anna Hospital, Department of Surgical Sciences, University of Turin, 10126 Turin, Italy
| | - Paolo Manzoni
- Department of Maternal, Neonatal and Infant Medicine, University Hospital “Degli Infermi”, 13875 Ponderano, Italy
| | - Bianca Masturzo
- Department of Maternal, Neonatal and Infant Medicine, University Hospital “Degli Infermi”, 13875 Ponderano, Italy
| |
Collapse
|
25
|
Cervantes O, Talavera IC, Every E, Coler B, Li M, Li A, Li H, Adams Waldorf K. Role of hormones in the pregnancy and sex-specific outcomes to infections with respiratory viruses. Immunol Rev 2022; 308:123-148. [PMID: 35373371 PMCID: PMC9189035 DOI: 10.1111/imr.13078] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 01/13/2023]
Abstract
Pregnant women infected with pathogenic respiratory viruses, such as influenza A viruses (IAV) and coronaviruses, are at higher risk for mortality, hospitalization, preterm birth, and stillbirth. Several factors are likely to contribute to the susceptibility of pregnant individuals to severe lung disease including changes in pulmonary physiology, immune defenses, and effector functions of some immune cells. Pregnancy is also a physiologic state characterized by higher levels of multiple hormones that may impact the effector functions of immune cells, such as progesterone, estrogen, human chorionic gonadotropin, prolactin, and relaxin. Each of these hormones acts to support a tolerogenic immune state of pregnancy, which helps prevent fetal rejection, but may also contribute to an impaired antiviral response. In this review, we address the unique role of adaptive and innate immune cells in the control of pathogenic respiratory viruses and how pregnancy and specific hormones can impact their effector actions. We highlight viruses with sex-specific differences in infection outcomes and why pregnancy hormones may contribute to fetal protection but aid the virus at the expense of the mother's health.
Collapse
Affiliation(s)
- Orlando Cervantes
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
| | - Irene Cruz Talavera
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Emma Every
- University of Washington School of Medicine, Spokane, Washington, United States of America
| | - Brahm Coler
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
- Elson S. Floyd College of Medicine, Washington State University, Spokane, Washington, United States of America
| | - Miranda Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
- Department of Biological Sciences, Columbia University, New York City, New York, United States of America
| | - Amanda Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
- Case Western Reserve, Cleveland, Ohio, United States of America
| | - Hanning Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
| | - Kristina Adams Waldorf
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
26
|
Froggatt HM, Heaton NS. Nonrespiratory sites of influenza-associated disease: mechanisms and experimental systems for continued study. FEBS J 2022; 289:4038-4060. [PMID: 35060315 PMCID: PMC9300775 DOI: 10.1111/febs.16363] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 10/20/2021] [Accepted: 01/19/2022] [Indexed: 12/15/2022]
Abstract
The productive replication of human influenza viruses is almost exclusively restricted to cells in the respiratory tract. However, a key aspect of the host response to viral infection is the production of inflammatory cytokines and chemokines that are not similarly tissue restricted. As such, circulating inflammatory mediators, as well as the resulting activated immune cells, can induce damage throughout the body, particularly in individuals with underlying conditions. As a result, more holistic experimental approaches are required to fully understand the pathogenesis and scope of influenza virus-induced disease. This review summarizes what is known about some of the most well-appreciated nonrespiratory tract sites of influenza virus-induced disease, including neurological, cardiovascular, gastrointestinal, muscular and fetal developmental phenotypes. In the context of this discussion, we describe the in vivo experimental systems currently being used to study nonrespiratory symptoms. Finally, we highlight important future questions and potential models that can be used for a more complete understanding of influenza virus-induced disease.
Collapse
Affiliation(s)
- Heather M. Froggatt
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Nicholas S. Heaton
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
| |
Collapse
|
27
|
Abdolrazaghnejad A, Miraj S. Can Coronavirus Disease 2019 Effect on Human Reproduction? Adv Biomed Res 2022; 11:55. [PMID: 35982865 PMCID: PMC9379922 DOI: 10.4103/abr.abr_236_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 01/05/2022] [Accepted: 01/19/2022] [Indexed: 11/04/2022] Open
Abstract
Since the main role in the pathogenesis of the coronavirus is attributed to the angiotensin-converting enzyme (ACE) receptor, it could possibly be a hypothesis in the differential sex-based pathogenesis of the coronavirus. The virus inserts its genetic material into the cell through its ACE2 receptors and replicates it by intracellular proteins. ACE2 receptors are highly expressed in cell membranes of various tissues in the body, including cardiovascular, gastrointestinal, renal, macrophage cells, and especially on the surface of type 2 pneumocytes in the lungs, ovaries, uterus, vagina, placenta, and testes. Therefore, cells having a higher expression of the ACE2 may be a specific target for coronavirus binding and infectivity. Due to the increase of infections in males, concerns have been appeared about the potential impact of coronavirus disease 2019 (COVID-19) on their fertility and reproductive organs. Thus, it is necessary to investigate if COVID-19 disturbs female and male fertility, so this review aimed to study the comprehensive evidences on the association of COVID-19 with human reproduction.
Collapse
Affiliation(s)
- Ali Abdolrazaghnejad
- Department of Emergency Medicine, Khatam-Al-Anbia Hospital, Clinical Immunology Research Center at Zahedan University of Medical Sciences, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Sepideh Miraj
- Department of Obstetrics and Gynecology, Nekouei-Hedayati-Forghani Hospital, Qom University of Medical Sciences, Qom, Iran,Address for correspondence: Dr. Sepideh Miraj, Associate Professor of Infertility and IVF, Department of Obstetrics and Gynecology, Nekouei-Hedayati-Forghani Hospital, Qom University of Medical Sciences, Qom, Iran. E-mail:
| |
Collapse
|
28
|
Upregulated influenza A viral entry factors and enhanced interferon-alpha response in the nasal epithelium of pregnant rats. Heliyon 2022; 8:e09407. [PMID: 35592667 PMCID: PMC9111991 DOI: 10.1016/j.heliyon.2022.e09407] [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: 11/20/2021] [Revised: 03/02/2022] [Accepted: 05/06/2022] [Indexed: 11/22/2022] Open
Abstract
Despite the increased severity of influenza A infection in pregnancy, knowledge about the expression of cell entry factors for influenza A virus (IAV) and the innate immune response in the nasal epithelium, the primary portal of viral entry, is limited. Here, we compared the expression of IAV cell entry factors and the status of the innate immune response in the nasal epithelium of pregnant vs. non-pregnant female rats. IAV cell entry factors — sialic acid [SA] α-2,3- and α-2,6-linked glycans for avian and human IAV, respectively — were detected and quantified with lectin-based immunoblotting and flow cytometry. Baseline frequencies of innate immune cell phenotypes in single cell suspensions of the nasal epithelium were studied with flow cytometry. Subsequently, the magnitude of interferon and cytokine responses was studied with ELISA and cytokine arrays after intranasal resiquimod, a Toll-like receptor 7/8 agonist that mimics IAV infection. We noted substantially increased expression of cell entry factors for both avian and human IAV in the nasal epithelium during pregnancy. Assessment of the innate immune state of the nasal epithelium during pregnancy revealed two previously unreported features: (i) increased presence of tissue-resident plasmacytoid dendritic cells, and (ii) markedly enhanced release of interferon-α but not of the other interferons or cytokines 2 h after intranasal resiquimod. Collectively, our findings challenge the conventional notion of pregnancy-induced immunosuppression as a cause for severe influenza A disease and suggest the need for focused studies on viral tropism during pregnancy to better understand the proximate cause for the observed immunopathology.
Collapse
|
29
|
Aguilera J, Han X, Cao S, Balmes J, Lurmann F, Tyner T, Lutzker L, Noth E, Hammond SK, Sampath V, Burt T, Utz PJ, Khatri P, Aghaeepour N, Maecker H, Prunicki M, Nadeau K. Increases in ambient air pollutants during pregnancy are linked to increases in methylation of IL4, IL10, and IFNγ. Clin Epigenetics 2022; 14:40. [PMID: 35287715 PMCID: PMC8919561 DOI: 10.1186/s13148-022-01254-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 02/21/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Ambient air pollutant (AAP) exposure is associated with adverse pregnancy outcomes, such as preeclampsia, preterm labor, and low birth weight. Previous studies have shown methylation of immune genes associate with exposure to air pollutants in pregnant women, but the cell-mediated response in the context of typical pregnancy cell alterations has not been investigated. Pregnancy causes attenuation in cell-mediated immunity with alterations in the Th1/Th2/Th17/Treg environment, contributing to maternal susceptibility. We recruited women (n = 186) who were 20 weeks pregnant from Fresno, CA, an area with chronically elevated AAP levels. Associations of average pollution concentration estimates for 1 week, 1 month, 3 months, and 6 months prior to blood draw were associated with Th cell subset (Th1, Th2, Th17, and Treg) percentages and methylation of CpG sites (IL4, IL10, IFNγ, and FoxP3). Linear regression models were adjusted for weight, age, season, race, and asthma, using a Q value as the false-discovery-rate-adjusted p-value across all genes. RESULTS Short-term and mid-term AAP exposures to fine particulate matter (PM2.5), nitrogen dioxide (NO2) carbon monoxide (CO), and polycyclic aromatic hydrocarbons (PAH456) were associated with percentages of immune cells. A decrease in Th1 cell percentage was negatively associated with PM2.5 (1 mo/3 mo: Q < 0.05), NO2 (1 mo/3 mo/6 mo: Q < 0.05), and PAH456 (1 week/1 mo/3 mo: Q < 0.05). Th2 cell percentages were negatively associated with PM2.5 (1 week/1 mo/3 mo/6 mo: Q < 0.06), and NO2 (1 week/1 mo/3 mo/6 mo: Q < 0.06). Th17 cell percentage was negatively associated with NO2 (3 mo/6 mo: Q < 0.01), CO (1 week/1 mo: Q < 0.1), PM2.5 (3 mo/6 mo: Q < 0.05), and PAH456 (1 mo/3 mo/6 mo: Q < 0.08). Methylation of the IL10 gene was positively associated with CO (1 week/1 mo/3 mo: Q < 0.01), NO2 (1 mo/3 mo/6 mo: Q < 0.08), PAH456 (1 week/1 mo/3 mo: Q < 0.01), and PM2.5 (3 mo: Q = 0.06) while IL4 gene methylation was positively associated with concentrations of CO (1 week/1 mo/3 mo/6 mo: Q < 0.09). Also, IFNγ gene methylation was positively associated with CO (1 week/1 mo/3 mo: Q < 0.05) and PAH456 (1 week/1 mo/3 mo: Q < 0.06). CONCLUSION Exposure to several AAPs was negatively associated with T-helper subsets involved in pro-inflammatory and anti-inflammatory responses during pregnancy. Methylation of IL4, IL10, and IFNγ genes with pollution exposure confirms previous research. These results offer insights into the detrimental effects of air pollution during pregnancy, the demand for more epigenetic studies, and mitigation strategies to decrease pollution exposure during pregnancy.
Collapse
Affiliation(s)
- Juan Aguilera
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University, 240 Pasteur, Stanford, CA, 94305, USA
| | - Xiaorui Han
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University, 240 Pasteur, Stanford, CA, 94305, USA
| | - Shu Cao
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University, 240 Pasteur, Stanford, CA, 94305, USA
| | - John Balmes
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | | | - Tim Tyner
- University of California, San Francisco-Fresno, Fresno, CA, USA
- Central California Asthma Collaborative, Fresno, USA
| | - Liza Lutzker
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA, USA
| | - Elizabeth Noth
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA, USA
| | - S Katharine Hammond
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, CA, USA
| | - Vanitha Sampath
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University, 240 Pasteur, Stanford, CA, 94305, USA
| | - Trevor Burt
- Department of Pediatrics, Division of Neonatology and the Translating Duke Health Children's Health and Discovery Initiative, Duke University School of Medicine, 701 W Main St., Chesterfield Building, Suite 510, Durham, NC, 27701, USA
| | - P J Utz
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford University, 291 Campus Drive, Stanford, CA, 94305, USA
| | - Purvesh Khatri
- Center for Biomedical Informatics, Department of Medicine, Stanford University School of Medicine, Stanford University, 291 Campus Drive, Stanford, CA, 94305, USA
| | - Nima Aghaeepour
- Departments of Biomedical Data Sciences, Stanford University, 291 Campus Drive, Stanford, CA, 94305, USA
| | - Holden Maecker
- Institute for Immunity, Transplantation and Infection, Stanford University, 291 Campus Drive, Stanford, CA, 94305, USA
| | - Mary Prunicki
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University, 240 Pasteur, Stanford, CA, 94305, USA
| | - Kari Nadeau
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University, 240 Pasteur, Stanford, CA, 94305, USA.
| |
Collapse
|
30
|
Akine D, Sasahara T, Koido A, Abe K, Abe K, Oki A, Takeyasu N, Hashimoto I. Case of a pregnant woman with probable prolonged SARS-CoV-2 viral shedding 221 days after diagnosis. J Infect Chemother 2022; 28:998-1000. [PMID: 35367149 PMCID: PMC8958094 DOI: 10.1016/j.jiac.2022.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 03/10/2022] [Accepted: 03/15/2022] [Indexed: 11/02/2022]
Abstract
We describe a case of probable prolonged severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Alpha(B.1.1.7) variant shedding for 221 days from the diagnosis, in a healthy 20-year-old Japanese pregnant woman with a normal delivery. To our knowledge, this is the longest duration of SARS-CoV-2 shedding reported in an immunocompetent individual to date.
Collapse
|
31
|
Ackerman CM, Nguyen JL, Ambati S, Reimbaeva M, Emir B, Cabrera J, Benigno M, Malhotra D, Hammond J, Bahtiyar MO. Clinical and Pregnancy Outcomes of Coronavirus Disease 2019 Among Hospitalized Pregnant Women in the United States. Open Forum Infect Dis 2022; 9:ofab429. [PMID: 35071680 PMCID: PMC8522379 DOI: 10.1093/ofid/ofab429] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 09/01/2021] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Pregnant women with coronavirus disease 2019 (COVID-19) may be at greater risk of poor maternal and pregnancy outcomes. This retrospective analysis reports clinical and pregnancy outcomes among hospitalized pregnant women with COVID-19 in the United States. METHODS The Premier Healthcare Database-Special Release was used to examine the impact of COVID-19 among pregnant women aged 15-44 years who were hospitalized and who delivered compared with pregnant women without COVID-19. Outcomes evaluated were COVID-19 clinical progression, including the use of supplemental oxygen therapy, intensive care unit admission, critical illness, receipt of invasive mechanical ventilation/extracorporeal membrane oxygenation, maternal death, and pregnancy outcomes, including preterm delivery and stillbirth. RESULTS Overall, 473 902 hospitalized pregnant women were included, 8584 (1.8%) of whom had a COVID-19 diagnosis (mean age = 28.4 [standard deviation = 6.1] years; 40% Hispanic). The risk of poor clinical and pregnancy outcomes was greater among pregnant women with COVID-19 compared with pregnant women without a COVID-19 diagnosis in 2020; the risk of poor clinical and pregnancy outcomes increased with increasing age. Hispanic and Black non-Hispanic women were consistently observed to have the highest relative risk of experiencing poor clinical or pregnancy outcomes across all age groups. CONCLUSIONS Overall, COVID-19 had a significant negative impact on maternal health and pregnancy outcomes. These data help inform clinical practice and counseling to pregnant women regarding the risks of COVID-19. Clinical studies evaluating the safety and efficacy of vaccines against severe acute respiratory syndrome coronavirus 2 in pregnant women are urgently needed.
Collapse
Affiliation(s)
- Christina M Ackerman
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, USA
| | | | | | | | | | - Javier Cabrera
- Department of Statistics Rutgers University, New Brunswick, New Jersey, USA
- Cardiovascular Institute, Rutgers University, New Brunswick, New Jersey, USA
| | | | | | | | - Mert Ozan Bahtiyar
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
32
|
Rashidi BH, Bandarian F, Bandarian M. Maternal and neonatal outcomes of pregnancies of infertile women during the COVID-19 pandemic: a real world evidence. JBRA Assist Reprod 2022; 26:594-598. [PMID: 35403419 PMCID: PMC9635598 DOI: 10.5935/1518-0557.20210119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE The COVID-19 pandemic began in Dec. 2019 and its effects on pregnancy outcomes are still unknown. This study aimed to evaluate the pregnancy outcomes of infertile women who conceived during the COVID-19 pandemic. METHODS This cross-sectional study included infertile women who conceived during the COVID-19 pandemic. Infertile women referred to the infertility center at the Vali-e-Asr hospital who conceived spontaneously or with the aid of ART (IUI, ICSI) were included and followed until delivery or pregnancy termination. RESULTS A total of 38 pregnant women (34 conceiving after ART and four spontaneously) were included. Seventeen (44.74%) of the 38 pregnant women developed COVID-19 symptoms. No significant difference was detected in maternal and neonatal outcomes, including miscarriage, PROM, low birth weight, or premature birth between pregnancies with and without COVID-19 symptoms. A significant difference was found between the two groups in delivery route. CONCLUSIONS No associations were found with maternal and neonatal morbidity in women conceiving during the COVID-19 pandemic and in pregnant women with and without COVID-19 symptoms.
Collapse
Affiliation(s)
- Batool Hossein Rashidi
- Health Reproductive Research Center, Imam Khomeini complex
Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Bandarian
- Metabolomics and Genomics Research Center, Endocrinology and
Metabolism Molecular- Cellular Sciences Institute, Tehran University of Medical
Sciences, Tehran, Iran , Endocrinology and Metabolism Research Center, Endocrinology and
Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences,
Tehran, Iran
| | - Mahin Bandarian
- Dep. of Obstetrics and Gynecology, Ziaeian Hospital, Tehran
University of Medical Sciences, Tehran, Iran
| |
Collapse
|
33
|
Chopra S, Syal A, Arya Y. Acute kidney injury in COVID-19: Considerations in pregnancy. Tzu Chi Med J 2022; 34:29-34. [PMID: 35233353 PMCID: PMC8830551 DOI: 10.4103/tcmj.tcmj_290_20] [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: 12/06/2020] [Revised: 01/19/2021] [Accepted: 03/26/2021] [Indexed: 01/08/2023] Open
Abstract
The clinical manifestations of COVID-19 are diverse with the involvement of different organ systems. Renal involvement is particularly noteworthy with acute kidney injury (AKI) being an important disease manifestation, particularly in pregnancy. Pregnancy itself serves as a high-risk condition for COVID-19 disease and a risk factor for deterioration, developing a more severe illness than nonpregnant women, and subsequent higher intensive care unit admission, oxygen therapy, and ventilatory support. There are reports in the literature highlighting the involvement of vital organs in pregnancy; however, data pertaining to AKI in pregnancy during COVID-19 are lacking in terms of risk factors, disease management, and outcomes. The entire spectrum of hormonal changes and adaptive mechanisms in pregnancy can be adversely affected by this viral infection. A literature search regarding AKI in COVID-19 in pregnancy was performed on PubMed, Scopus, Google Scholar, and ScienceDirect, and the relevant articles were selected. Our review highlights key issues pertaining to AKI in COVID-19 in pregnancy in an attempt to overcome, albeit partly, the scarcity of corroborative literature regarding the same.
Collapse
|
34
|
Dauby N, Flamand V. From maternal breath to infant's cells: Impact of maternal respiratory infections on infants 'immune responses. Front Pediatr 2022; 10:1046100. [PMID: 36419921 PMCID: PMC9676445 DOI: 10.3389/fped.2022.1046100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 10/19/2022] [Indexed: 11/09/2022] Open
Abstract
In utero exposure to maternally-derived antigens following chronic infection is associated with modulation of infants 'immune response, differential susceptibility to post-natal infections and immune response toward vaccines. The maternal environment, both internal (microbiota) and external (exposure to environmental microbes) also modulates infant's immune response but also the clinical phenotype after birth. Vertical transmission of ubiquitous respiratory pathogens such as influenza and COVID-19 is uncommon. Evidence suggest that in utero exposure to maternal influenza and SARS-CoV-2 infections may have a significant impact on the developing immune system with activation of both innate and adaptive responses, possibly related to placental inflammation. Here in, we review how maternal respiratory infections, associated with airway, systemic and placental inflammation but also changes in maternal microbiota might impact infant's immune responses after birth. The clinical impact of immune modifications observed following maternal respiratory infections remains unexplored. Given the high frequencies of respiratory infections during pregnancy (COVID-19, influenza but also RSV and HMPV), the impact on global child health could be important.
Collapse
Affiliation(s)
- Nicolas Dauby
- Institute for Medical Immunology, ULB Center for Research in Immunology, Université Libre de Bruxelles (ULB), Brussels, Belgium.,Department of Infectious Diseases, CHU Saint-Pierre, Brussels, Belgium.,School of Public Health, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Véronique Flamand
- Institute for Medical Immunology, ULB Center for Research in Immunology, Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
35
|
Januszewski M, Ziuzia-Januszewska L, Jakimiuk AA, Wierzba W, Głuszko A, Żytyńska-Daniluk J, Jakimiuk AJ. Is the Course of COVID-19 Different during Pregnancy? A Retrospective Comparative Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:12011. [PMID: 34831766 PMCID: PMC8620897 DOI: 10.3390/ijerph182212011] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/12/2021] [Accepted: 11/14/2021] [Indexed: 01/08/2023]
Abstract
The COVID-19 pandemic has challenged health systems around the world. Maternal-foetal medicine, which has been particularly affected, must consider scientific data on the physiological processes occurring in the pregnant woman's body to develop relevant standards of care. Our study retrospectively compared the clinical and laboratory characteristics of 52 COVID-19 pregnant patients with 53 controls. Most of the pregnant patients required medical attention during the third trimester and therefore we propose that vaccination is needed prior to the 30th week of pregnancy. We found no differences between the 2 groups in the course of illness classification system, days of hospital stay, need for oxygen supplementation, need for mechanical ventilation, and ICU admission. Moreover, clinical manifestations and imaging findings were comparable. Pregnant patients needed a greater oxygen flow rate and required high flow oxygen therapy more frequently. Considering pregnancy-related physiological adaptations, we found that COVID-19 infection in pregnant patients is associated with higher levels of inflammatory markers, apart from serum ferritin, than in non-pregnant women, and concluded that biomarkers of cardiac and muscle injury, as well as kidney function, may not be good predictors of COVID-19 clinical course in pregnant patients at the time of admission, but more research needs to be conducted on this topic.
Collapse
Affiliation(s)
- Marcin Januszewski
- Department of Obstetrics and Gynecology, Central Clinical Hospital of the Ministry of Interior and Administration, 02-507 Warsaw, Poland; (M.J.); (W.W.)
| | - Laura Ziuzia-Januszewska
- Department of Otolaryngology, Central Clinical Hospital of the Ministry of Interior and Administration, 02-507 Warsaw, Poland;
| | - Alicja A. Jakimiuk
- Department of Plastic Surgery, Central Clinical Hospital of the Ministry of Interior and Administration, 02-507 Warsaw, Poland;
| | - Waldemar Wierzba
- Department of Obstetrics and Gynecology, Central Clinical Hospital of the Ministry of Interior and Administration, 02-507 Warsaw, Poland; (M.J.); (W.W.)
- Satellite Campus in Warsaw, University of Humanities and Economics, 01-513 Warsaw, Poland
| | - Anna Głuszko
- Department of Neonatology, Central Clinical Hospital of the Ministry of Interior and Administration, 02-507 Warsaw, Poland; (A.G.); (J.Ż.-D.)
| | - Joanna Żytyńska-Daniluk
- Department of Neonatology, Central Clinical Hospital of the Ministry of Interior and Administration, 02-507 Warsaw, Poland; (A.G.); (J.Ż.-D.)
| | - Artur J. Jakimiuk
- Department of Obstetrics and Gynecology, Central Clinical Hospital of the Ministry of Interior and Administration, 02-507 Warsaw, Poland; (M.J.); (W.W.)
- Center for Reproductive Health, Institute of Mother and Child, 01-211 Warsaw, Poland
| |
Collapse
|
36
|
Menne Z, Pliasas VC, Compans RW, Glover S, Kyriakis CS, Skountzou I. Bivalent vaccination with NA1 and NA2 neuraminidase virus-like particles is protective against challenge with H1N1 and H3N2 influenza A viruses in a murine model. Virology 2021; 562:197-208. [PMID: 34375782 PMCID: PMC8479372 DOI: 10.1016/j.virol.2021.08.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/25/2021] [Accepted: 08/04/2021] [Indexed: 11/29/2022]
Abstract
Neuraminidase (NA) is the second most abundant glycoprotein on the surface of influenza A viruses (IAV). Neuraminidase type 1 (NA1) based virus-like particles (VLPs) have previously been shown to protect against challenge with H1N1 and H3N2 IAV. In this study, we produced neuraminidase type 2 (NA2) VLPs derived from the sequence of the seasonal IAV A/Perth/16/2009. Intramuscular vaccination of mice with NA2 VLPs induced high anti-NA serum IgG levels capable of inhibiting NA activity. NA2 VLP vaccination protected against mortality in a lethal A/Hong Kong/1/1968 (H3N2) virus challenge model, but not against lethal challenge with A/California/04/2009 (H1N1) virus. However, bivalent vaccination with NA1 and NA2 VLPs demonstrated no antigenic competition in anti-NA IgG responses and protected against lethal challenge with H1N1 and H3N2 viruses. Here we demonstrate that vaccination with NA VLPs is protective against influenza challenge and supports focusing on anti-NA responses in the development of future vaccination strategies.
Collapse
MESH Headings
- Animals
- Disease Models, Animal
- Dose-Response Relationship, Immunologic
- Immunity, Heterologous
- Immunoglobulin G/immunology
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H3N2 Subtype/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/immunology
- Injections, Intramuscular
- Mice
- Neuraminidase/immunology
- Orthomyxoviridae Infections/prevention & control
- Vaccination/methods
- Vaccines, Combined/administration & dosage
- Vaccines, Combined/immunology
- Vaccines, Virus-Like Particle/administration & dosage
- Vaccines, Virus-Like Particle/immunology
- Viral Proteins/immunology
Collapse
Affiliation(s)
- Zach Menne
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, 30322, USA; Centers for Excellence in Influenza Research and Surveillance, Emory-UGA Center, Atlanta, GA, USA
| | - Vasilis C Pliasas
- Centers for Excellence in Influenza Research and Surveillance, Emory-UGA Center, Atlanta, GA, USA; Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, 36849, USA
| | - Richard W Compans
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, 30322, USA; Centers for Excellence in Influenza Research and Surveillance, Emory-UGA Center, Atlanta, GA, USA
| | - Sheniqua Glover
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, 36849, USA
| | - Constantinos S Kyriakis
- Centers for Excellence in Influenza Research and Surveillance, Emory-UGA Center, Atlanta, GA, USA; Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, 36849, USA
| | - Ioanna Skountzou
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, 30322, USA; Centers for Excellence in Influenza Research and Surveillance, Emory-UGA Center, Atlanta, GA, USA.
| |
Collapse
|
37
|
Ponce-Alonso M, Fernández-Félix BM, Halperin A, Rodríguez-Domínguez M, Sánchez-Díaz AM, Cantón R, Muriel A, Zamora J, Del Campo R. Propensity-Score Analysis Reveals that Sex is Not a Prognostic Factor for Mortality in Intensive Care Unit-Admitted Patients with Septic Bacteremia. Int J Infect Dis 2021; 110:36-44. [PMID: 34274507 DOI: 10.1016/j.ijid.2021.07.034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/12/2021] [Accepted: 07/13/2021] [Indexed: 01/12/2023] Open
Abstract
OBJECTIVE Men have been considered to have a higher incidence of infectious diseases, with controversy over the possibility that sex could influence the prognosis of the infection. This study aimed to explore this assumption in patients admitted to the intensive care unit (ICU) with septic bacteremia. METHODS A retrospective analysis (2006-2017) of septic patients with microbiologically confirmed bacteremia (n=440) was performed. Risk of ICU and in-hospital mortality in males versus females was compared by univariate analysis and a propensity score analysis integrating their clinical characteristics. RESULTS Sepsis more frequently occurred in males (80.2% vs 76.1%) as well as in-hospital (48.0% vs 41.3%) and ICU (39.9% vs 36.5%) mortality. Univariate analyses showed that males had a higher Charlson comorbidity index and worse McCabe prognostic score. However, the propensity score in 296 matched patients demonstrated that females had higher risk of both ICU (OR 1.39; 95% CI 0.89-2.19) and in-hospital mortality (OR 1.18; 95% CI 0.77-1.83), but without statistical significance. CONCLUSION Males with sepsis had worse clinical characteristics when admitted to the ICU, but sex had no influence on mortality. These data contribute to helping reduce the sex-dependent gap present in healthcare provision.
Collapse
Affiliation(s)
- Manuel Ponce-Alonso
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal, Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain; Red Española de Investigación en Patología Infecciosa (REIPI), Madrid, Spain
| | - Borja M Fernández-Félix
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain; Unidad de Bioestadística Clínica, Hospital Ramón y Cajal, Madrid, Spain; CIBER Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Ana Halperin
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal, Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain; Red Española de Investigación en Patología Infecciosa (REIPI), Madrid, Spain
| | - Mario Rodríguez-Domínguez
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal, Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain; CIBER Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Ana M Sánchez-Díaz
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal, Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain; Red Española de Investigación en Patología Infecciosa (REIPI), Madrid, Spain
| | - Rafael Cantón
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal, Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain; Red Española de Investigación en Patología Infecciosa (REIPI), Madrid, Spain
| | - Alfonso Muriel
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain; Unidad de Bioestadística Clínica, Hospital Ramón y Cajal, Madrid, Spain; CIBER Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Javier Zamora
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain; Unidad de Bioestadística Clínica, Hospital Ramón y Cajal, Madrid, Spain; CIBER Epidemiology and Public Health (CIBERESP), Madrid, Spain; Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom.
| | - Rosa Del Campo
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal, Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain; Red Española de Investigación en Patología Infecciosa (REIPI), Madrid, Spain; University Alfonso X El Sabio, Villanueva de la Cañada, Madrid, Spain.
| |
Collapse
|
38
|
Zgutka K, Prasanth K, Pinero-Bernardo S, Lew LQ, Cervellione K, Rhythm R, Rahman L, Dolmaian G, Cohen L. Infant outcomes and maternal COVID-19 status at delivery. J Perinat Med 2021; 49:691-696. [PMID: 33713593 DOI: 10.1515/jpm-2020-0481] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 02/24/2021] [Indexed: 12/18/2022]
Abstract
OBJECTIVES To compare clinical characteristics and outcomes of infants born to COVID-19 to non COVID-19 mothers at delivery in a community hospital in Queens, New York. METHODS Case-control study conducted March 15 to June 15, 2020. Cases were infants born to mothers with laboratory-confirmed COVID-19 infection at delivery. The infant of non COVID-19 mother born before and after each case were selected as controls. RESULTS Of 695 deliveries, 62 (8.9%) infants were born to COVID-19 mothers; 124 controls were selected. Among cases, 18.3% were preterm compared to 8.1% in controls (p=0.04). In preterm cases, birth weight was not significantly different between groups. However, there was a significantly higher proportion of neonatal intensive care unit (NICU) admissions, need for respiratory support, suspected sepsis, hyperbilirubinemia, feeding intolerance and longer length of stay (LOS) in preterm cases. Among term cases, birth weight and adverse outcomes were not significantly different between cases and controls except for more feeding intolerance in cases. All infants born to COVID-19 mothers were COVID-19 negative at 24 and 48 h of life. No infants expired during birth hospitalization. CONCLUSIONS Significantly, more infants of COVID-19 mothers were premature compared to controls. Preterm cases were more likely to have adverse outcomes despite having similar birth weight and gestational age. These differences were not seen among full term infants. Health care providers should anticipate the need for NICU care when a COVID-19 mother presents in labor.
Collapse
Affiliation(s)
- Kinga Zgutka
- Department of Pediatrics, Flushing Hospital Medical Center, Flushing, NY, USA
| | - Kaninghat Prasanth
- Division of Neonatology, Department of Pediatrics, Flushing Hospital Medical Center, Flushing, NY, USA
| | | | - Lily Q Lew
- Department of Pediatrics, Flushing Hospital Medical Center, Flushing, NY, USA
| | - Kelly Cervellione
- Department of Clinical Research, Medisys Health Network, Flushing, NY, USA
| | - Rhythm Rhythm
- Department of Pediatrics, Flushing Hospital Medical Center, Flushing, NY, USA
| | - Lubna Rahman
- Department of Pediatrics, Flushing Hospital Medical Center, Flushing, NY, USA
| | - Gigliola Dolmaian
- Division of Neonatology, Department of Pediatrics, Flushing Hospital Medical Center, Flushing, NY, USA
| | - Lourdes Cohen
- Division of Neonatology, Department of Pediatrics, Flushing Hospital Medical Center, Flushing, NY, USA
| |
Collapse
|
39
|
Animal Models Utilized for the Development of Influenza Virus Vaccines. Vaccines (Basel) 2021; 9:vaccines9070787. [PMID: 34358203 PMCID: PMC8310120 DOI: 10.3390/vaccines9070787] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/08/2021] [Accepted: 07/10/2021] [Indexed: 12/25/2022] Open
Abstract
Animal models have been an important tool for the development of influenza virus vaccines since the 1940s. Over the past 80 years, influenza virus vaccines have evolved into more complex formulations, including trivalent and quadrivalent inactivated vaccines, live-attenuated vaccines, and subunit vaccines. However, annual effectiveness data shows that current vaccines have varying levels of protection that range between 40–60% and must be reformulated every few years to combat antigenic drift. To address these issues, novel influenza virus vaccines are currently in development. These vaccines rely heavily on animal models to determine efficacy and immunogenicity. In this review, we describe seasonal and novel influenza virus vaccines and highlight important animal models used to develop them.
Collapse
|
40
|
Figueiro-Filho EA, Hobson SR, Farine D, Yudin MH. Highly expressed ACE-2 receptors during pregnancy: A protective factor for SARS-COV-2 infection? Med Hypotheses 2021; 153:110641. [PMID: 34256245 PMCID: PMC8262774 DOI: 10.1016/j.mehy.2021.110641] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 05/25/2021] [Accepted: 07/04/2021] [Indexed: 01/10/2023]
Abstract
While previous viral pandemics showed that pregnancy was a risk factor for susceptibility and adverse outcomes, current evidence is conflicting whether SARS-CoV-2 infection during pregnancy is more severe than in the general population, with relatively low maternal and fetal/neonatal mortality rates. SARS-CoV-2 is known to enter host cells via the ACE-2 receptors, competitively occupying their binding sites. In theory, viral invasion can lead to a reduction in available ACE-2 receptors and consequently an unbalanced regulation between the ACE-AngII-AT1 axis and the ACE-2-Ang-(1-7)-MAS axis, thus enhancing pathological vasoconstriction, fibrosis, inflammation and thrombotic processes. We hypothesize that the normal pregnant state of highly expressed ACE-2 receptors leads to higher Ang-(1-7) levels and consequently more vasodilation and anti-inflammatory response to SARS-COV-2 infection. We suggest that this up-regulation of ACE-2 receptors in human gestation may actually be clinically protective and propose a potential research line to investigate this hypothesis, which may lead to future novel therapeutics.
Collapse
Affiliation(s)
- Ernesto Antonio Figueiro-Filho
- Mount Sinai Hospital, Maternal Fetal Medicine Division, Department of Obstetrics and Gynecology, University of Toronto, Toronto, Canada.
| | - Sebastian R Hobson
- Mount Sinai Hospital, Maternal Fetal Medicine Division, Department of Obstetrics and Gynecology, University of Toronto, Toronto, Canada
| | - Dan Farine
- Mount Sinai Hospital, Maternal Fetal Medicine Division, Department of Obstetrics and Gynecology, University of Toronto, Toronto, Canada
| | - Mark H Yudin
- Saint Michael's Hospital, Maternal Fetal Medicine Division, Department of Obstetrics and Gynecology, University of Toronto, Toronto, Canada
| |
Collapse
|
41
|
Flerlage T, Boyd DF, Meliopoulos V, Thomas PG, Schultz-Cherry S. Influenza virus and SARS-CoV-2: pathogenesis and host responses in the respiratory tract. Nat Rev Microbiol 2021; 19:425-441. [PMID: 33824495 PMCID: PMC8023351 DOI: 10.1038/s41579-021-00542-7] [Citation(s) in RCA: 231] [Impact Index Per Article: 57.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2021] [Indexed: 01/31/2023]
Abstract
Influenza viruses cause annual epidemics and occasional pandemics of respiratory tract infections that produce a wide spectrum of clinical disease severity in humans. The novel betacoronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged in December 2019 and has since caused a pandemic. Both viral and host factors determine the extent and severity of virus-induced lung damage. The host's response to viral infection is necessary for viral clearance but may be deleterious and contribute to severe disease phenotypes. Similarly, tissue repair mechanisms are required for recovery from infection across the spectrum of disease severity; however, dysregulated repair responses may lead to chronic lung dysfunction. Understanding of the mechanisms of immunopathology and tissue repair following viral lower respiratory tract infection may broaden treatment options. In this Review, we discuss the pathogenesis, the contribution of the host response to severe clinical phenotypes and highlight early and late epithelial repair mechanisms following influenza virus infection, each of which has been well characterized. Although we are still learning about SARS-CoV-2 and its disease manifestations in humans, throughout the Review we discuss what is known about SARS-CoV-2 in the context of this broad knowledge of influenza virus, highlighting the similarities and differences between the respiratory viruses.
Collapse
Affiliation(s)
- Tim Flerlage
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - David F Boyd
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Victoria Meliopoulos
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Stacey Schultz-Cherry
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
42
|
Abstract
Progesterone is crucial for the maintenance of pregnancy. During pregnancy hepatitis E virus (HEV) infection is associated with increased fulminant hepatic failure and mortality rates. In this study, we determined whether progesterone modulates HEV replication and HEV-induced innate cytokine response in Huh7-S10-3 human liver cells. We first demonstrated that Huh7-S10-3 liver cells expressed SH3-domain-containing progesterone receptor membrane component (PGRMC)1/2 receptors involved in the progesterone nonclassical signaling pathway, while the classical progesterone receptor isoforms progesterone receptor-A and -B protein levels were undetectable. We showed that the genotype 3 HEV (strain P6) induced mRNA expression of type III interferon (IFN-λ1), but not other innate cytokines in Huh7-S10-3 cells. Pretreatment with progesterone at concentrations of 80 nM, 160 nM, or 480 nM, which are the physiological concentrations typically seen in the first- to third-trimester during pregnancy, significantly increased HEV replication in Huh7-S10-3 cells. However, pretreatment of cells with progesterone (80 nM) did not affect the level of HEV-induced IFN-λ1 mRNA expression. We further showed that loss of PGRMC1/2 receptors by small interfering RNA (siRNA) knockdown leads to an increase in HEV-induced IFN-λ1 expression levels at early time points via the extracellular signal-regulated kinase pathway and thus resulted in a reduced level of HEV replication. Collectively, the results indicated that progesterone-mediated modulation of HEV replication in human liver cells is plausibly through SH3-domain containing proteins such as PGRMC1/2, but not likely through immunomodulation of HEV-induced interferon response in liver cells. The results have important implications in understanding the underlying mechanisms of high mortality and fulminant hepatitis in HEV-infected pregnant women.
Collapse
|
43
|
Bukowska-Ośko I, Popiel M, Kowalczyk P. The Immunological Role of the Placenta in SARS-CoV-2 Infection-Viral Transmission, Immune Regulation, and Lactoferrin Activity. Int J Mol Sci 2021; 22:5799. [PMID: 34071527 PMCID: PMC8198160 DOI: 10.3390/ijms22115799] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/25/2021] [Accepted: 05/27/2021] [Indexed: 02/07/2023] Open
Abstract
A pandemic of acute respiratory infections, due to a new type of coronavirus, can cause Severe Acute Respiratory Syndrome 2 (SARS-CoV-2) and has created the need for a better understanding of the clinical, epidemiological, and pathological features of COVID-19, especially in high-risk groups, such as pregnant women. Viral infections in pregnant women may have a much more severe course, and result in an increase in the rate of complications, including spontaneous abortion, stillbirth, and premature birth-which may cause long-term consequences in the offspring. In this review, we focus on the mother-fetal-placenta interface and its role in the potential transmission of SARS-CoV-2, including expression of viral receptors and proteases, placental pathology, and the presence of the virus in neonatal tissues and fluids. This review summarizes the current knowledge on the anti-viral activity of lactoferrin during viral infection in pregnant women, analyzes its role in the pathogenicity of pandemic virus particles, and describes the potential evidence for placental blocking/limiting of the transmission of the virus.
Collapse
Affiliation(s)
- Iwona Bukowska-Ośko
- Department of Immunopathology of Infectious and Parasitic Diseases, Medical University of Warsaw, 02-091Warsaw, Poland;
| | - Marta Popiel
- Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3, 05-110 Jabłonna, Poland;
| | - Paweł Kowalczyk
- Department of Animal Nutrition, The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, Instytucka 3, 05-110 Jabłonna, Poland;
| |
Collapse
|
44
|
Alaya F, Worrall AP, O'Toole F, Doyle J, Duffy RM, Geary MP. Health-related quality of life and quality of care in pregnant and postnatal women during the coronavirus disease 2019 pandemic: A cohort study. Int J Gynaecol Obstet 2021; 154:100-105. [PMID: 33864252 PMCID: PMC9087707 DOI: 10.1002/ijgo.13711] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/05/2021] [Accepted: 04/12/2021] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Health-related quality of life (HRQoL) and the delivery of high-quality care are ongoing concerns when caring for pregnant women during the coronavirus disease 2019 (COVID-19) pandemic. We compared self-reported HRQoL and hospital quality of care among perinatal women with and without COVID-19. METHODS This is a prospective cohort study of perinatal women attending a tertiary maternity unit during the pandemic. Eighteen women who tested positive for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and 20 SARS-CoV-2-negative women were recruited. Participants completed the Short Form Health Survey (SF-12), Clinical Outcomes in Routine Evaluation-Outcome Measure, and Quality from the Patient's Perspective questionnaires. Mean scores were compared. RESULTS Of the Non-COVID-19 cohort, 95% (n = 19) were Caucasian, whereas 67% (n = 12) of the COVID-19 cohort were not Caucasian (χ2 = 16.01, P < 0.001). The mean SF-12 for physical health in the COVID-19 cohort had significantly lower scores (P < 0.002). There was no difference in mental health and well-being between cohorts. The quality of care experienced was notably similar and very positive. CONCLUSION There was a significantly greater burden on physical health among pregnant women with COVID-19. Mental health and psychological status were similar in both groups. High quality of care during a pandemic is possible to deliver in a maternity setting, irrespective of COVID-19 status.
Collapse
Affiliation(s)
- Fátimah Alaya
- Rotunda Hospital, Dublin, Ireland.,Royal College of Surgeons Ireland, Dublin, Ireland
| | | | | | | | | | - Michael P Geary
- Rotunda Hospital, Dublin, Ireland.,Royal College of Surgeons Ireland, Dublin, Ireland
| |
Collapse
|
45
|
Clinical findings, treatments and obstetric results of pregnant women diagnosed with coronavirus disease 2019. JOURNAL OF SURGERY AND MEDICINE 2021. [DOI: 10.28982/josam.907597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
46
|
Granja MG, Oliveira ACDR, de Figueiredo CS, Gomes AP, Ferreira EC, Giestal-de-Araujo E, de Castro-Faria-Neto HC. SARS-CoV-2 Infection in Pregnant Women: Neuroimmune-Endocrine Changes at the Maternal-Fetal Interface. Neuroimmunomodulation 2021; 28:1-21. [PMID: 33910207 PMCID: PMC8247841 DOI: 10.1159/000515556] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 02/23/2021] [Indexed: 12/15/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) has devastating effects on the population worldwide. Given this scenario, the extent of the impact of the disease on more vulnerable individuals, such as pregnant women, is of great concern. Although pregnancy may be a risk factor in respiratory virus infections, there are no considerable differences regarding COVID-19 severity observed between pregnant and nonpregnant women. In these circumstances, an emergent concern is the possibility of neurodevelopmental and neuropsychiatric harm for the offspring of infected mothers. Currently, there is no stronger evidence indicating vertical transmission of SARS-CoV-2; however, the exacerbated inflammatory response observed in the disease could lead to several impairments in the offspring's brain. Furthermore, in the face of historical knowledge on possible long-term consequences for the progeny's brain after infection by viruses, we must consider that this might be another deleterious facet of COVID-19. In light of neuroimmune interactions at the maternal-fetal interface, we review here the possible harmful outcomes to the offspring brains of mothers infected by SARS-CoV-2.
Collapse
Affiliation(s)
- Marcelo Gomes Granja
- Molecular and Cellular Biology Program, Federal University of State of Rio de Janeiro − UNIRIO, Rio de Janeiro, Rajasthan, Brazil
- Immunopharmacology Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation − Fiocruz, Rio de Janeiro, Rajasthan, Brazil
| | | | | | - Alex Portes Gomes
- Medical Science Program, Neurology and Neuroscience, Fluminense Federal University − UFF, Niterói, Rajasthan, Brazil
| | - Erica Camila Ferreira
- Immunopharmacology Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation − Fiocruz, Rio de Janeiro, Rajasthan, Brazil
| | - Elizabeth Giestal-de-Araujo
- Neuroscience Program, Fluminense Federal University − UFF, Niterói, Rajasthan, Brazil
- National Institute of Technology-Neuroimmunomodulation − INCT-NIM, Rio de Janeiro, Rajasthan, Brazil
| | - Hugo Caire de Castro-Faria-Neto
- Immunopharmacology Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation − Fiocruz, Rio de Janeiro, Rajasthan, Brazil
- National Institute of Technology-Neuroimmunomodulation − INCT-NIM, Rio de Janeiro, Rajasthan, Brazil
| |
Collapse
|
47
|
Regan AK, Munoz FM. Efficacy and safety of influenza vaccination during pregnancy: realizing the potential of maternal influenza immunization. Expert Rev Vaccines 2021; 20:649-660. [PMID: 33832397 DOI: 10.1080/14760584.2021.1915138] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Introduction: Pregnant women are at higher risk of severe complications following influenza infection compared to the general population. Influenza vaccination during pregnancy can offer direct protection to pregnant women and passive immunity to infants up to 6 months of age via maternal antibodies. Pregnant women are a high priority group for influenza immunization.Areas covered: This review provides an overview of the basis for recommending influenza vaccine to pregnant women, current immunization policies, the evidence supporting the safety and effectiveness of maternal vaccination, and future research needed. We conducted a search of PubMed for articles describing the safety or efficacy of influenza vaccines administered during pregnancy. Published articles from inception to 17 November 2020 were reviewed.Expert opinion: Experimental and observational evidence support the efficacy, effectiveness and safety of influenza immunization during pregnancy. These data support the continued provision of inactivated influenza vaccine to pregnant women, as recommended by global immunization policies. To achieve success with maternal influenza immunization programs, further work is needed to inform policy development in low- and middle-income settings and implementation and promotion in high-income settings.
Collapse
Affiliation(s)
- Annette K Regan
- School of Nursing and Health Professions, University of San Francisco, San Francisco, CA, United States.,Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, United States.,Wesfarmers Centre for Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
| | - Flor M Munoz
- Department of Pediatrics, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX, United States.,Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
48
|
Lu QC, Zhang TY, Bundhun PK, Chen C. One "misunderstood" health issue: demonstrating and communicating the safety of influenza a vaccination in pregnancy: a systematic review and meta-analysis. BMC Public Health 2021; 21:703. [PMID: 33836695 PMCID: PMC8034177 DOI: 10.1186/s12889-021-10740-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 03/30/2021] [Indexed: 03/19/2023] Open
Abstract
Background The American College of Obstetricians and Gynecologists (ACOG) makes certain recommendations including the annual influenza vaccination of pregnant and pre-pregnant women during influenza (flu) season with an inactivated influenza vaccine as soon as it becomes available. The Centers for Disease Control and Prevention’s (CDC) Advisory Committee on Immunization Practices in association with ACOG state that the vaccine is safe to be given any trimester during pregnancy. However, due to a lack of communication, the public is unaware of the effects of influenza A vaccination in pregnancy. Since this is a vital public health concern, we aimed to communicate with evidence, the safety of influenza A vaccination in pregnancy in order to improve the rate of influenza A vaccines in pregnant women. Methods This health communication issue was based on the impact of influenza vaccine on fetal outcomes. Therefore, a search was carried out through medical-based online databases including: Cochrane Central, EMBASE, Web of Science, MEDLINE, http://www.ClinicalTrials.gov, and Google scholar for relevant English-based publications. Adverse fetal outcomes were considered as the endpoints of this analysis. The most specific RevMan 5.3 (latest version) software was used to carry out this analysis. Risk ratios (RR) with 95% confidence intervals (CI) were involved in data and results representation and interpretation. Results A total number of 679, 992 pregnant women participated in this analysis. Based on this current analysis, premature/preterm birth (< 37 weeks) was significantly reduced in pregnant women who were vaccinated for influenza A (RR: 0.80, 95% CI: 0.69–0.92; P = 0.002) as compared to those women who were not vaccinated. Similarly, influenza A vaccination decreased the risk for very preterm birth (< 32 weeks) (RR: 0.70, 95% CI: 0.58–0.84; P = 0.0001). The risks for infants with low birth weight (RR: 0.71, 95% CI: 0.49–1.04; P = 0.08), very low birth weight (RR: 0.69, 95% CI: 0.23–2.11; P = 0.52) and infants small for gestational age (RR: 0.93, 95% CI: 0.83–1.05; P = 0.26) were not increased with the vaccine. Influenza A vaccination was not associated with increased risks of stillbirth (RR: 0.63, 95% CI: 0.38–1.03; P = 0.07), birth defects (RR: 0.67, 95% CI: 0.26–1.72; P = 0.41), admission to neonatal intensive care unit or Apgar score < 7 in 5 min. Conclusion Influenza vaccine is completely safe in pregnancy. It significantly lowers premature birth and is not associated with any serious adverse neonatal outcome. Hence, this important piece of information should be communicated and conveyed to all pregnant women, for a safer and healthier pregnancy. At last, this public health issue should further be addressed to the population through media and other communication means in order to improve the rate of influenza A vaccines in pregnant women for a healthier and more productive population.
Collapse
Affiliation(s)
- Qing Chun Lu
- Department of Obstetrics and Gynaecology, National Hospital of Guangxi Zhuang Autonomous Region, Affiliated to Guangxi Medical University, Nanning, Guangxi, 530000, People's Republic of China
| | - Tie Yun Zhang
- Department of Communication, School of Journalism and New Media, Xi'An JiaoTong University, Xi'An, Shanxi, 710000, People's Republic of China
| | | | - Cheng Chen
- Department of Broadcasting and Television, School of Journalism and Information Communication, Huazhong University of Science and Technology, Wuhan, 430000, People's Republic of China.
| |
Collapse
|
49
|
Wang R, Yan W, Du M, Tao L, Liu J. The effect of influenza virus infection on pregnancy outcomes: A systematic review and meta-analysis of cohort studies. Int J Infect Dis 2021; 105:567-578. [PMID: 33647509 DOI: 10.1016/j.ijid.2021.02.095] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/20/2021] [Accepted: 02/24/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Adverse pregnancy outcomes are risk factors for neonatal mortality and morbidity. While some studies have demonstrated notable associations between influenza and adverse pregnancy outcomes, the findings have contrasted with other studies. This meta-analysis was conducted to assess the effect of influenza infection on pregnancy outcomes. METHODS We searched PubMed, Embase, Cochrane Library and Web of Science from inception to 4 November 2020. Relative risks (RRs) with 95% confidence intervals (CIs) were pooled using random-effects or fixed-effects models. RESULTS A total of 17 studies involving 2,351,204 participants were included. Influenza infection increased the risk of stillbirth (RR = 3.62, 95% CI: 1.60-8.20), with no significant effect on preterm birth (RR = 1.17, 95%CI: 0.95-1.45), fetal death (RR = 0.93, 95%CI: 0.73-1.18), small for gestational age (SGA) (RR = 1.10, 95%CI: 0.98-1.24) and low birth weight (LBW) (RR = 1.88, 95%CI: 0.46-7.66). In a subgroup analysis of LBW, the association was evident in studies conducted during the 2009 H1N1 pandemic (RR = 2.28, 95%CI: 1.81-2.87), with no evidence of an association in pre-pandemic or post-pandemic studies. CONCLUSIONS Influenza virus infection was associated with an increased risk of stillbirth, but its effect on preterm birth, fetal death, SGA and LBW is still uncertain.
Collapse
Affiliation(s)
- Ruitong Wang
- School of Public Health, Peking University, No.38 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Wenxin Yan
- School of Public Health, Peking University, No.38 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Min Du
- School of Public Health, Peking University, No.38 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Liyuan Tao
- Research Center of Clinical Epidemiology, Peking University Third Hospital, No.49 Huayuan North Road, Haidian District, Beijing, 100083, China
| | - Jue Liu
- School of Public Health, Peking University, No.38 Xueyuan Road, Haidian District, Beijing, 100191, China.
| |
Collapse
|
50
|
Chow EJ, Beigi RH, Riley LE, Uyeki TM. Clinical Effectiveness and Safety of Antivirals for Influenza in Pregnancy. Open Forum Infect Dis 2021; 8:ofab138. [PMID: 34189160 DOI: 10.1093/ofid/ofab138] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 03/19/2021] [Indexed: 11/14/2022] Open
Abstract
Seasonal influenza epidemics result in substantial health care burden annually. Early initiation of antiviral treatment of influenza has been shown to reduce the risk of complications and duration of illness. Pregnant and postpartum women may be at increased risk for influenza-associated complications; however, pregnant women have been generally excluded from clinical trials of antiviral treatment of influenza. In this review, we summarize the available evidence on the clinical effectiveness and safety of antiviral treatment of pregnant women with influenza. Observational data show a reduction of severe outcomes when pregnant and postpartum women are treated with oseltamivir and other neuraminidase inhibitors without increased risk of adverse maternal, fetal, or neonatal outcomes. Due to lack of safety and efficacy data for baloxavir treatment of pregnant and postpartum women, baloxavir is currently not recommended for use in these populations.
Collapse
Affiliation(s)
- Eric J Chow
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Richard H Beigi
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,University of Pittsburgh Magee-Womens Hospital, Pittsburgh, Pennsylvania, USA
| | - Laura E Riley
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York Presbyterian Hospital, New York, New York, USA
| | - Timothy M Uyeki
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|