1
|
Simoni MK, Negatu SG, Park JY, Mani S, Arreguin MC, Amses KR, Huh DD, Mainigi M, Jurado KA. Type I interferon exposure of an implantation-on-a-chip device alters invasive extravillous trophoblast function. Cell Rep Med 2025; 6:101991. [PMID: 40054459 PMCID: PMC11970386 DOI: 10.1016/j.xcrm.2025.101991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 12/06/2024] [Accepted: 02/05/2025] [Indexed: 03/12/2025]
Abstract
Inappropriate type I interferon (IFN) signaling during embryo implantation and placentation is linked to poor pregnancy outcomes. Here, we evaluate the consequence of elevated type I IFN exposure on implantation using a human implantation in an organ-on-a-chip device. We reveal that type I IFN reduces extravillous trophoblast (EVT) invasion capacity. Analyzing single-cell transcriptomes, we uncover that IFN truncates invasive EVT emergence in the implantation-on-a-chip device by stunting EVT epithelial-to-mesenchymal transition. Disruptions to the epithelial-to-mesenchymal transition are associated with the pathogenesis of preeclampsia, a life-threatening disorder of pregnancy. Strikingly, IFN stimulation induces genes associated with increased preeclampsia risk in EVTs. These dysregulated EVT phenotypes ultimately reduce EVT-mediated endothelial cell vascular remodeling in the implantation-on-a-chip device. Overall, our work implicates unwarranted type I IFN as a maternal disturbance that can result in abnormal EVT function that could trigger preeclampsia.
Collapse
Affiliation(s)
- Michael K Simoni
- Department of Obstetrics and Gynecology, Hospital at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Seble G Negatu
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Ju Young Park
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sneha Mani
- Department of Obstetrics and Gynecology, Hospital at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Montserrat C Arreguin
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Kevin R Amses
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Dan Dongeun Huh
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Monica Mainigi
- Department of Obstetrics and Gynecology, Hospital at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Kellie A Jurado
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
2
|
Balduit A, Agostinis C, Mangogna A, Zito G, Stampalija T, Ricci G, Bulla R. Systematic review of the complement components as potential biomarkers of pre-eclampsia: pitfalls and opportunities. Front Immunol 2024; 15:1419540. [PMID: 38983853 PMCID: PMC11232388 DOI: 10.3389/fimmu.2024.1419540] [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: 04/18/2024] [Accepted: 06/07/2024] [Indexed: 07/11/2024] Open
Abstract
The complement system (C) is a crucial component of the innate immune system. An increasing body of research has progressively shed light on the pivotal role of C in immunological tolerance at the feto-maternal interface. Excessive C activation or impaired C regulation may determine the onset of pregnancy-related pathological conditions, including pre-eclampsia (PE). Thus, several studies have investigated the presence of C components or split products in blood matrixes (i.e., plasma, serum), urine, and amniotic fluid in PE. In the current study, we systematically reviewed the currently available scientific literature reporting measurements of C components as circulating biomarkers in PE, based on a literature search using Pubmed, Scopus, and Embase databases. A total of 41 out of 456 studies were selected after full-text analysis. Fourteen studies (34.1%) were identified as measuring the blood concentrations of the classical pathway, 5 (12.1%) for the lectin pathway, 28 (68.3%) for the alternative pathway, 17 (41.5%) for the terminal pathway components, and 16 (39%) for C regulators. Retrieved results consistently reported C4, C3, and factor H reduction, and increased circulating levels of C4d, Bb, factor D, C3a, C5a, and C5b-9 in PE compared to normal pregnancies, depicting an overall scenario of excessive C activation and aberrant C regulation. With evidence of C activation and dysregulation, C-targeted therapy is an intriguing perspective in PE management. Moreover, we also discussed emerging pitfalls in C analysis, mainly due to a lack of experimental uniformity and biased cohort selection among different studies and laboratories, aiming to raise a more comprehensive awareness for future standardization. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42024503070.
Collapse
Affiliation(s)
- Andrea Balduit
- Institute for Maternal and Child Health - IRCCS “Burlo Garofolo”, Trieste, Italy
| | - Chiara Agostinis
- Institute for Maternal and Child Health - IRCCS “Burlo Garofolo”, Trieste, Italy
| | - Alessandro Mangogna
- Institute for Maternal and Child Health - IRCCS “Burlo Garofolo”, Trieste, Italy
| | - Gabriella Zito
- Institute for Maternal and Child Health - IRCCS “Burlo Garofolo”, Trieste, Italy
| | - Tamara Stampalija
- Institute for Maternal and Child Health - IRCCS “Burlo Garofolo”, Trieste, Italy
- Department of Medical, Surgical and Health Science, University of Trieste, Trieste, Italy
| | - Giuseppe Ricci
- Institute for Maternal and Child Health - IRCCS “Burlo Garofolo”, Trieste, Italy
- Department of Medical, Surgical and Health Science, University of Trieste, Trieste, Italy
| | - Roberta Bulla
- Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
3
|
Govender S, David M, Naicker T. Is the Complement System Dysregulated in Preeclampsia Comorbid with HIV Infection? Int J Mol Sci 2024; 25:6232. [PMID: 38892429 PMCID: PMC11172754 DOI: 10.3390/ijms25116232] [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: 05/02/2024] [Revised: 05/27/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
South Africa is the epicentre of the global HIV pandemic, with 13.9% of its population infected. Preeclampsia (PE), a hypertensive disorder of pregnancy, is often comorbid with HIV infection, leading to multi-organ dysfunction and convulsions. The exact pathophysiology of preeclampsia is triggered by an altered maternal immune response or defective development of maternal tolerance to the semi-allogenic foetus via the complement system. The complement system plays a vital role in the innate immune system, generating inflammation, mediating the clearance of microbes and injured tissue materials, and a mediator of adaptive immunity. Moreover, the complement system has a dual effect, of protecting the host against HIV infection and enhancing HIV infectivity. An upregulation of regulatory proteins has been implicated as an adaptive phenomenon in response to elevated complement-mediated cell lysis in HIV infection, further aggravated by preeclamptic complement activation. In light of the high prevalence of HIV infection and preeclampsia in South Africa, this review discusses the association of complement proteins and their role in the synergy of HIV infection and preeclampsia in South Africa. It aims to identify women at elevated risk, leading to early diagnosis and better management with targeted drug therapy, thereby improving the understanding of immunological dysregulation.
Collapse
Affiliation(s)
| | | | - Thajasvarie Naicker
- Optics and Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa; (S.G.); (M.D.)
| |
Collapse
|
4
|
Simoni MK, Negatu SG, Park JY, Mani S, Arreguin MC, Amses K, Huh DD, Mainigi M, Jurado KA. Type I interferon alters invasive extravillous trophoblast function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.11.584521. [PMID: 38559122 PMCID: PMC10979977 DOI: 10.1101/2024.03.11.584521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Inappropriate type I interferon (IFN) signaling during embryo implantation and placentation is linked to poor pregnancy outcomes. Here, we evaluated the consequence of elevated type I IFN exposure on implantation using a biomimetic model of human implantation in an organ-on-a-chip device. We found that type I IFN reduced extravillous trophoblast (EVT) invasion capacity. Analyzing single-cell transcriptomes, we uncovered that IFN truncated endovascular EVT emergence in the implantation-on-a-chip device by stunting EVT epithelial-to-mesenchymal transition. Disruptions to the epithelial-to-mesenchymal transition is associated with the pathogenesis of preeclampsia, a life-threatening hypertensive disorder of pregnancy. Strikingly, unwarranted IFN stimulation induced genes associated with increased preeclampsia risk and a preeclamptic gene-like signature in EVTs. These dysregulated EVT phenotypes ultimately reduced EVT-mediated endothelial cell vascular remodeling in the implantation-on-a-chip device. Overall, our work indicates IFN signaling can alter EVT epithelial-to-mesenchymal transition progression which results in diminished EVT-mediated spiral artery remodeling and a preeclampsia gene signature upon sustained stimulation. Our work implicates unwarranted type I IFN as a maternal disturbance that can result in abnormal EVT function that could trigger preeclampsia.
Collapse
|
5
|
Yasmin H, Agostinis C, Toffoli M, Roy T, Pegoraro S, Balduit A, Zito G, Di Simone N, Ricci G, Madan T, Kishore U, Bulla R. Protective role of complement factor H against the development of preeclampsia. Front Immunol 2024; 15:1351898. [PMID: 38464530 PMCID: PMC10920295 DOI: 10.3389/fimmu.2024.1351898] [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: 12/07/2023] [Accepted: 01/30/2024] [Indexed: 03/12/2024] Open
Abstract
Pregnancy is an immunologically regulated, complex process. A tightly controlled complement system plays a crucial role in the successful establishment of pregnancy and parturition. Complement inhibitors at the feto-maternal interface are likely to prevent inappropriate complement activation to protect the fetus. In the present study, we aimed to understand the role of Factor H (FH), a negative regulator of complement activation, in normal pregnancy and in a model of pathological pregnancy, i.e. preeclampsia (PE). The distribution and expression of FH was investigated in placental tissues, various placental cells, and in the sera of healthy (CTRL) or PE pregnant women via immunohistochemistry, RT-qPCR, ELISA, and Western blot. Our results showed a differential expression of FH among the placental cell types, decidual stromal cells (DSCs), decidual endothelial cells (DECs), and extravillous trophoblasts (EVTs). Interestingly, FH was found to be considerably less expressed in the placental tissues of PE patients compared to normal placental tissue both at mRNA and protein levels. Similar results were obtained by measuring circulating FH levels in the sera of third trimester CTRL and PE mothers. Syncytiotrophoblast microvesicles, isolated from the placental tissues of PE and CTRL women, downregulated FH expression by DECs. The present study appears to suggest that FH is ubiquitously present in the normal placenta and plays a homeostatic role during pregnancy.
Collapse
Affiliation(s)
- Hadida Yasmin
- Immunology and Cell Biology Laboratory, Department of Zoology, Cooch Behar Panchanan Barma University, Cooch Behar, West Bengal, India
| | - Chiara Agostinis
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Miriam Toffoli
- Department of Medical, Surgical and Health Science, University of Trieste, Trieste, Italy
| | - Tamali Roy
- Immunology and Cell Biology Laboratory, Department of Zoology, Cooch Behar Panchanan Barma University, Cooch Behar, West Bengal, India
| | - Silvia Pegoraro
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Andrea Balduit
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Gabriella Zito
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Nicoletta Di Simone
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Milan, Italy
| | - Giuseppe Ricci
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
- Department of Medical, Surgical and Health Science, University of Trieste, Trieste, Italy
| | - Taruna Madan
- Department of Innate Immunity, ICMR-National Institute for Research in Reproductive and Child Health (NIRRCH), Mumbai, India
| | - Uday Kishore
- Department of Veterinary Medicine, U.A.E. University, Al Ain, United Arab Emirates
- Zayed Centre for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Roberta Bulla
- Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
6
|
Gajić M, Schröder-Heurich B, Mayer-Pickel K. Deciphering the immunological interactions: targeting preeclampsia with Hydroxychloroquine's biological mechanisms. Front Pharmacol 2024; 15:1298928. [PMID: 38375029 PMCID: PMC10875033 DOI: 10.3389/fphar.2024.1298928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 01/23/2024] [Indexed: 02/21/2024] Open
Abstract
Preeclampsia (PE) is a complex pregnancy-related disorder characterized by hypertension, followed by organ dysfunction and uteroplacental abnormalities. It remains a major cause of maternal and neonatal morbidity and mortality worldwide. Although the pathophysiology of PE has not been fully elucidated, a two-stage model has been proposed. In this model, a poorly perfused placenta releases various factors into the maternal circulation during the first stage, including pro-inflammatory cytokines, anti-angiogenic factors, and damage-associated molecular patterns into the maternal circulation. In the second stage, these factors lead to a systemic vascular dysfunction with consecutive clinical maternal and/or fetal manifestations. Despite advances in feto-maternal management, effective prophylactic and therapeutic options for PE are still lacking. Since termination of pregnancy is the only curative therapy, regardless of gestational age, new treatment/prophylactic options are urgently needed. Hydroxychloroquine (HCQ) is mainly used to treat malaria as well as certain autoimmune conditions such as systemic lupus and rheumatoid arthritis. The exact mechanism of action of HCQ is not fully understood, but several mechanisms of action have been proposed based on its pharmacological properties. Interestingly, many of them might counteract the proposed processes involved in the development of PE. Therefore, based on a literature review, we aimed to investigate the interrelated biological processes of HCQ and PE and to identify potential molecular targets in these processes.
Collapse
Affiliation(s)
- Maja Gajić
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | | | | |
Collapse
|
7
|
David M, Singh S, Naicker T. Is the central complement component C3 altered in the synergy of HIV infection and preeclampsia? Eur J Obstet Gynecol Reprod Biol X 2023; 20:100257. [PMID: 37885814 PMCID: PMC10598405 DOI: 10.1016/j.eurox.2023.100257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/26/2023] [Accepted: 10/17/2023] [Indexed: 10/28/2023] Open
Abstract
Objective In light of complement activation in preeclampsia and HIV infection, this study evaluates the concentration of complement component 3 (C3) in HIV-associated preeclampsia. Method The study population (n = 76) was equally stratified by pregnancy type (normotensive pregnant and preeclampsia) and by HIV status (HIV positive and HIV negative). The plasma concentration of C3 was determined using a Bioplex immunoassay procedure. Results We report a significant increase in C3 concentration in the HIV-negative versus the HIV-positive groups (p < 0.05), regardless of pregnancy type. However, based on pregnancy type and irrespective of HIV status, C3 concentration was similar between normotensive versus preeclampsia. Concentration of C3 was significantly increased in the HIV-positive preeclamptic compared HIV-negative preeclamptic groups (p = 0.04). The correlation of C3 with all study groups was non-significant. Conclusion This study demonstrates that C3 was upregulated in HIV-associated PE compared to HIV- associated normotensive pregnancies. The dysregulation of C3 expression by HIV infection may be attributed to antiretroviral therapy.
Collapse
Affiliation(s)
- Mikyle David
- Optics and Imaging Centre, Doris Duke Medical Research Institute, Nelson R. Mandela School of Medicine, College of Health Sciences, University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa
| | - Shoohana Singh
- Optics and Imaging Centre, Doris Duke Medical Research Institute, Nelson R. Mandela School of Medicine, College of Health Sciences, University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa
| | - Thajasvarie Naicker
- Optics and Imaging Centre, Doris Duke Medical Research Institute, Nelson R. Mandela School of Medicine, College of Health Sciences, University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa
| |
Collapse
|
8
|
Deer E, Herrock O, Campbell N, Cornelius D, Fitzgerald S, Amaral LM, LaMarca B. The role of immune cells and mediators in preeclampsia. Nat Rev Nephrol 2023; 19:257-270. [PMID: 36635411 PMCID: PMC10038936 DOI: 10.1038/s41581-022-00670-0] [Citation(s) in RCA: 95] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2022] [Indexed: 01/14/2023]
Abstract
Preeclampsia is a hypertensive disorder of major concern in pregnancy than can lead to intrauterine growth restriction, placental abruption and stillbirth. The pathophysiology of preeclampsia is multifactorial, including not only kidney dysfunction but also endothelial dysfunction, as the maternal endothelium becomes exposed to placental factors that are released into the circulation and increase systemic levels of vasoconstrictors, oxidative stress, anti-angiogenic factors and inflammatory mediators. Importantly, inflammation can lead to insufficient placental perfusion and low birthweight in offspring. Various innate and adaptive immune cells and mediators have been implicated in the development of preeclampsia, in which oxidative stress is associated with activation of the maternal inflammatory response. Immune cells such as regulatory T cells, macrophages, natural killer cells, and neutrophils are known to have major causative roles in the pathology of preeclampsia, but the contributions of additional immune cells such as B cells, inflammatory cytokines and anti-angiotensin II type 1 receptor autoantibodies are also now recognized. Immunological interventions, therefore, have therapeutic potential in this disease. Here, we provide an overview of the immune responses that are involved in the pathogenesis of preeclampsia, including the role of innate and adaptive immune cells and mediators.
Collapse
Affiliation(s)
- Evangeline Deer
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Owen Herrock
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Nathan Campbell
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Denise Cornelius
- Emergency Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Sarah Fitzgerald
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Lorena M Amaral
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Babbette LaMarca
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS, USA.
- Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, MS, USA.
| |
Collapse
|
9
|
Zhang X, Chen Y, Sun D, Zhu X, Ying X, Yao Y, Fei W, Zheng C. Emerging pharmacologic interventions for pre-eclampsia treatment. Expert Opin Ther Targets 2022; 26:739-759. [PMID: 36223503 DOI: 10.1080/14728222.2022.2134779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Pre-eclampsia is a serious pregnancy complication and a major global concern for the mortality of both mother and fetus. Existing symptomatic treatments do not delay disease progression; thus, timely delivery of the baby is the most effective measure. However, the risk of various maternal and fetal injuries remains. AREAS COVERED In this review, we summarize the potential strategies for pharmacologic interventions in pre-eclamptic therapy. Specifically, we discuss the pathophysiological process of various effective candidate therapeutics that act on potential pathways and molecular targets to inhibit key stages of the disease. We refer to this pathogenesis-focused drug discovery model as a pathogenesis-target-drug (P-T-D) strategy. Finally, we discuss the introduction of nanotechnologies to improve the safety and efficacy of therapeutics via their specific placental targeting ability and placental retention effects. EXPERT OPINION Despite the active development of novel pharmacological treatments based on our current knowledge of pre-eclamptic pathogenesis, investigations are still in the early phase. Thus, further exploration of the pathological mechanisms, integrated with the P-T-D strategy and novel nanosystems, could encourage the development of more effective and safer strategies. Such advances could lead to a shift from expectant management to mechanistic-based therapy for pre-eclampsia.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Yue Chen
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Dongli Sun
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Xiaojun Zhu
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Xia Ying
- Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Yao Yao
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Weidong Fei
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Caihong Zheng
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| |
Collapse
|
10
|
Livson S, Virtanen S, Lokki AI, Holster T, Rahkonen L, Kalliala I, Nieminen P, Salonen A, Meri S. Cervicovaginal Complement Activation and Microbiota During Pregnancy and in Parturition. Front Immunol 2022; 13:925630. [PMID: 35958597 PMCID: PMC9358961 DOI: 10.3389/fimmu.2022.925630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/23/2022] [Indexed: 11/24/2022] Open
Abstract
Background Vaginal microbiome and the local innate immune defense, including the complement system, contribute to anti- and proinflammatory homeostasis during pregnancy and parturition. The relationship between commensal vaginal bacteria and complement activation during pregnancy and delivery is not known. Objective To study the association of the cervicovaginal microbiota composition to activation and regulation of the complement system during pregnancy and labor. Study design We recruited women during late pregnancy (weeks 41 + 5 to 42 + 0, n=48) and women in active labor (weeks 38 + 4 to 42 + 2, n=25). Mucosal swabs were taken from the external cervix and lateral fornix of the vagina. From the same sampling site, microbiota was analyzed with 16S RNA gene amplicon sequencing. A Western blot technique was used to detect complement C3, C4 and factor B activation and presence of complement inhibitors. For semiquantitative analysis, the bands of the electrophoresed proteins in gels were digitized on a flatbed photo scanner and staining intensities were analyzed using ImageJ/Fiji win-64 software. Patient data was collected from medical records and questionnaires. Results The vaginal microbiota was Lactobacillus-dominant in most of the samples (n=60), L. iners and L. crispatus being the dominant species. L. gasseri and L. jensenii were found to be more abundant during pregnancy than active labor. L. jensenii abundance correlated with C4 activation during pregnancy but not in labor. Gardnerella vaginalis was associated with C4 activation both during pregnancy and labor. The amount of L. gasseri correlated with factor B activation during pregnancy but not during labor. Atopobium vaginae was more abundant during pregnancy than labor and correlated with C4 activation during labor and with factor B activation during pregnancy. Activation of the alternative pathway factor B was significantly stronger during pregnancy compared to labor. During labor complement activation may be inhibited by the abundant presence of factor H and FHL1. Conclusions These results indicate that bacterial composition of the vaginal microbiota could have a role in the local activation and regulation of complement-mediated inflammation during pregnancy. At the time of parturition complement activation appears to be more strictly regulated than during pregnancy.
Collapse
Affiliation(s)
- Sivan Livson
- Department of Obstetrics and Gynecology, Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland
- Department of Bacteriology and Immunology and Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Seppo Virtanen
- Department of Obstetrics and Gynecology, Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - A. Inkeri Lokki
- Department of Obstetrics and Gynecology, Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland
- Department of Bacteriology and Immunology and Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Tiina Holster
- Department of Obstetrics and Gynecology, Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland
| | - Leena Rahkonen
- Department of Obstetrics and Gynecology, Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland
| | - Ilkka Kalliala
- Department of Obstetrics and Gynecology, Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Pekka Nieminen
- Department of Obstetrics and Gynecology, Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Anne Salonen
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Seppo Meri
- Department of Bacteriology and Immunology and Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- Hospital District of Helsinki and Uusimaa (HUS) Diagnostic Center, Hospital District of Helsinki and Uusimaa laboratorio (HUSLAB), Helsinki University Hospital Laboratory, Helsinki, Finland
| |
Collapse
|
11
|
Kerboua KE, Saadia L. The evaluation of some serum immunochemical markers in early onset pre-eclamptic women from Algeria. Am J Reprod Immunol 2022; 88:e13587. [PMID: 35770379 DOI: 10.1111/aji.13587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 06/03/2022] [Accepted: 06/14/2022] [Indexed: 11/30/2022] Open
Abstract
PROBLEM Progress in understanding the underlying mechanism responsible for the syndrome pre-eclampsia should reconfigure antenatal clinical care and minimize human and financial costs, yet at present there is no accurate theory that permits development of reliable predictive tests and prophylactic intervention to mitigate disease. To contribute to this ongoing effort, we aimed to assess various circulating markers pertaining to different theories. METHOD OF STUDY Serum samples from thirty-four women with established early onset preeclampsia (ePE) were assessed in terms of oxidative stress (malondialdehyde- MDA), angiogenic status (PlGF & sFLT-1), complement system (The alternative pathway -AP50 & complement factor H- CFH) and circulating inflammatory markers (Interleukin 6- IL-6 & Procalcitonin- PCT). Control groups of gestational age matched patients included 20 gestational hypertensive (GH) and 6 normotensive pregnant women (NPW). RESULTS Our work shows that PlGF is the only serum marker who does exhibit a continued decrease from NPW to GH to ePE (rpearson = -0.428, p = .002). The ePE group had a profound impairment in circulating PlGF (66.93 ± 20.62 pg/ml) compared to GH (142.67 ± 39.79 pg/ml; p = .069) and NPW (636.83 ± 392.66 pg/ml; p = .002). Then, PlGF >71.29 pg/ml pg/ml is the cut-off that has the highest negative predictive value enabling exclusion of ePE (Sp 78%, Se 70%, p = .000). No such interesting results could be obtained with the other markers. CONCLUSION Our data confirm that the angiogenic factor PlGF may be highly relevant in biological mechanisms underlying the development of ePE. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Kheir Eddine Kerboua
- Laboratory of Desert Medicine, Faculty of Medicine, University of Kasdi Merbah, Ouargla, Algeria.,Laboratory of Immunology, Regional Military University Hospital of Oran, Oran, Algeria
| | - Lasla Saadia
- Laboratory of Biochemistry, Regional Military University Hospital of Oran, Oran, Algeria
| |
Collapse
|
12
|
Molecular Advances in Preeclampsia and HELLP Syndrome. Int J Mol Sci 2022; 23:ijms23073851. [PMID: 35409211 PMCID: PMC8999044 DOI: 10.3390/ijms23073851] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/28/2022] [Accepted: 03/28/2022] [Indexed: 01/27/2023] Open
Abstract
Preeclampsia (PE) constitutes one of the principal reasons for maternal and perinatal morbidity and mortality worldwide. The circumstance typically implicates formerly healthful normotensive women, after 20 weeks of gestation, typically withinside the third trimester, without regarded threat elements or past deliveries. PE can be further complicated with hemolysis and thrombocytopenia, leading to the emergence of HELLP syndrome (Hemolysis, Elevated Liver enzymes, Low platelets). Both conditions are classified as hypertensive diseases of pregnancy (HDP), and their pathogenesis has been linked to an excessive maternal inflammatory response, accompanied by enhanced endothelial activation. Several studies have found that in pregnancies affected by PE/HELLP, von Willebrand factor (vWF) antigen levels (vWF:Ag) are significantly elevated, while its cleaving protease (ADAMTS-13, A Disintegrin-like and Metalloprotease with Thrombospondin type 1 motif, member 13) activity is normal to decreased. Furthermore, the higher urine excretion of the terminal complement complex C5b-9, as well as its greater deposition in the placental surface in preeclamptic women, imply that the utero-placental unit’s distinctive deficits are intimately tied to disproportionate complement activation. The goal of this updated evaluation is to provide the most up-to-date molecular advances in the pathophysiology of PE/HELLP syndromes. Recent medical data on vWF:Ag levels in patients with PE, ADAMTS-13, and dysregulation of the complement system, are highlighted and evaluated. Furthermore, we discuss the relationship between those entities and the progression of the disease, as well as their significance in the diagnostic process. Finally, considering the difficulties in analyzing and controlling those symptoms in pregnant women, we can provide a current diagnostic and therapeutic algorithm.
Collapse
|
13
|
Dijkstra DJ, Lokki AI, Gierman LM, Borggreven NV, van der Keur C, Eikmans M, Gelderman KA, Laivuori H, Iversen AC, van der Hoorn MLP, Trouw LA. Circulating Levels of Anti-C1q and Anti-Factor H Autoantibodies and Their Targets in Normal Pregnancy and Preeclampsia. Front Immunol 2022; 13:842451. [PMID: 35432365 PMCID: PMC9009242 DOI: 10.3389/fimmu.2022.842451] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 03/07/2022] [Indexed: 11/10/2022] Open
Abstract
Preeclampsia (PE) generally manifests in the second half of pregnancy with hypertension and proteinuria. The understanding of the origin and mechanism behind PE is incomplete, although there is clearly an immune component to this disorder. The placenta constitutes a complicated immune interface between fetal and maternal cells, where regulation and tolerance are key. Stress factors from placental dysfunction in PE are released to the maternal circulation evoking the maternal response. Several complement factors play a role within this intricate landscape, including C1q in vascular remodeling and Factor H (FH) as the key regulator of alternative pathway complement activation. We hypothesize that decreased levels of C1q or FH, or disturbance of their function by autoantibodies, may be associated with PE. Autoantibodies against C1q and FH and the concentrations of C1q and FH were measured by ELISA in maternal sera from women with preeclamptic and normal pregnancies. Samples originated from cohorts collected in the Netherlands (n=63 PE; n=174 control pregnancies, n=51 nonpregnant), Finland (n=181 PE; n=63 control pregnancies) and Norway (n=59 PE; n=27 control pregnancies). Serum C1q and FH concentrations were higher in control pregnancy than in nonpregnant women. No significant differences were observed for serum C1q between preeclamptic and control pregnancy in any of the three cohorts. Serum levels of FH were lower in preeclamptic pregnancies compared to control pregnancies in two of the cohorts, this effect was driven by the early onset PE cases. Neither anti-C1q autoantibodies nor anti-FH autoantibodies levels differed between women with PE and normal pregnancies. In conclusion, levels of anti-C1q and anti-FH autoantibodies are not increased in PE. C1q and FH are increased in pregnancy, but importantly, a decrease in FH concentration is associated with PE.
Collapse
Affiliation(s)
- Douwe Jan Dijkstra
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands,*Correspondence: Douwe Jan Dijkstra, ; Leendert Adrianus Trouw,
| | - A. Inkeri Lokki
- Department of Bacteriology and Immunology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland,Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Lobke Marijn Gierman
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | | | - Carin van der Keur
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Michael Eikmans
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Kyra Andrea Gelderman
- Department of Immunopathology and Haemostasis, Sanquin Diagnostic Services, Amsterdam, Netherlands
| | - Hannele Laivuori
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland,Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland,Department of Obstetrics and Gynecology, Tampere University Hospital and Tampere University, Faculty of Medicine and Health Technology, Tampere Center for Child, Adolescent, and Maternal Health Research, Tampere, Finland
| | | | - Ann-Charlotte Iversen
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | | | - Leendert Adrianus Trouw
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands,*Correspondence: Douwe Jan Dijkstra, ; Leendert Adrianus Trouw,
| |
Collapse
|
14
|
Complement activation and regulation in preeclampsia and hemolysis, elevated liver enzymes, and low platelet count syndrome. Am J Obstet Gynecol 2022; 226:S1059-S1070. [PMID: 32986992 DOI: 10.1016/j.ajog.2020.09.038] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/19/2020] [Accepted: 09/23/2020] [Indexed: 12/20/2022]
Abstract
The complement system is critical to human health owing to its central role in host defense and innate immunity. During pregnancy, the complement system must be appropriately regulated to allow for immunologic tolerance to the developing fetus and placenta. Although some degree of complement activation can be seen in normal pregnancy, the fetus seems to be protected in part through the placental expression of complement regulatory proteins, which inhibit complement activation at different steps along the complement activation cascade. In women who develop preeclampsia and hemolysis, elevated liver enzymes, and low platelet count syndrome, there is a shift toward increased complement activation and decreased complement regulation. There is an increase in placental deposition of C5b-9, which is the terminal effector of classical, lectin, and alternative complement pathways. C5b-9 deposition stimulates trophoblasts to secrete soluble fms-like tyrosine kinase-1, which sequesters vascular endothelial growth factor and placental growth factor. Pathogenic mutations or deletions in complement regulatory genes, which predispose to increased complement activation, have been detected in women with preeclampsia and hemolysis, elevated liver enzymes, and low platelet count syndrome. Before the disease, biomarkers of alternative complement pathway activation are increased; during active disease, biomarkers of terminal complement pathway activation are increased. Urinary excretion of C5b-9 is associated with preeclampsia with severe features and distinguishes it from other hypertensive disorders of pregnancy. Taken together, existing data link preeclampsia and hemolysis, elevated liver enzymes, and low platelet count syndrome with increased activation of the terminal complement pathway that, in some cases, may be influenced by genetic alterations in complement regulators. These findings suggest that the inhibition of the terminal complement pathway, possibly through C5 blockade, may be an effective strategy to treat preeclampsia and hemolysis, elevated liver enzymes, and low platelet count syndrome, but this strategy warrants further evaluation in clinical trials.
Collapse
|
15
|
Liu M, Luo X, Xu Q, Yu H, Gao L, Zhou R, Wang T. Adipsin of the Alternative Complement Pathway Is a Potential Predictor for Preeclampsia in Early Pregnancy. Front Immunol 2021; 12:702385. [PMID: 34671343 PMCID: PMC8521101 DOI: 10.3389/fimmu.2021.702385] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 09/15/2021] [Indexed: 01/05/2023] Open
Abstract
Objective The concentrations of complement proteins (adipsin, C3a, and C5a) and soluble endoglin (sENG) in the plasma were measured in this study, and their value as early-pregnancy predictors and potential diagnostic marker of preeclampsia was assessed, respectively. Experimental Design Plasma samples were obtained from healthy and preeclampsia pregnant women before delivery for a cross-sectional study. Plasma samples were collected from healthy and preeclampsia pregnant women throughout pregnancy and postpartum for a follow-up study. Enzyme-linked immunosorbent assays were used to detect plasma levels of several complement proteins (adipsin, C3a, and C5a) and sENG. Results The plasma levels of adipsin, C5a, and sENG were significantly increased before delivery in pregnant women with preeclampsia. During pregnancy, the plasma adipsin, C5a, and sENG levels were increased from the third trimester in healthy pregnant women; plasma adipsin levels remained stable after delivery, while C3a levels increased in the second trimester and remained stable afterward. Furthermore, levels of adipsin, C5a, and sENG were higher in preeclampsia patients at different stages of pregnancy; the C3a level presents a similar change and no difference was found in the third trimester. In the first trimester, receiver-operating curve (ROC) curve analysis showed that adipsin (AUC, 0.83 ± 0.06, P=0.001) and sENG (AUC, 0.74 ± 0.09, P=0.021) presented high value as predictors of early pregnancy. Conclusions Adipsin is likely a novel plasma biomarker to monitor the increased risk of preeclampsia in early pregnancy. Moreover, the increased plasma levels of adipsin, C5a, and sENG before delivery may be associated with preeclampsia.
Collapse
Affiliation(s)
- Min Liu
- Department of Obstetrics and Gynecology, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xiaolei Luo
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Qin Xu
- Department of Obstetrics and Gynecology, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Hongbiao Yu
- Department of Obstetrics and Gynecology, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Linbo Gao
- Department of Obstetrics and Gynecology, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Rong Zhou
- Department of Obstetrics and Gynecology, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Tao Wang
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
16
|
The function of adipsin and C9 protein in the complement system in HIV-associated preeclampsia. Arch Gynecol Obstet 2021; 304:1467-1473. [PMID: 33881585 DOI: 10.1007/s00404-021-06069-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 04/09/2021] [Indexed: 12/27/2022]
Abstract
OBJECTIVE In preeclampsia, there are excessive complement components expressed due to increased complement activation; therefore, this study investigated the concentration of adipsin and C9 in HIV-associated preeclampsia. METHOD The study population (n = 76) was stratified by pregnancy type (normotensive pregnant and preeclampsia) and by HIV status. Serum was assayed for the concentration of adipsin and C9 using a Bioplex immunoassay procedure. RESULTS Maternal weight did not differ (p = 0.1196) across the study groups. The concentration of adipsin was statistically different between the PE vs normotensive pregnant groups, irrespective of HIV status (p = 0.0439). There was no significant difference in adipsin concentration between HIV-negative vs HIV-positive groups, irrespective of pregnancy type (p = 0.6290). Additionally, there was a significant difference in adipsin concentration between HIV-negative normotensive vs HIV-negative preeclampsia (p < 0.05), as well as a difference between HIV-negative preeclampsia vs HIV-positive preeclampsia (p < 0.05). C9 protein expression was not statistically different between the normotensive and PE groups, regardless of HIV status (p = 0.5365). No statistical significance in C9 expression was found between HIV-positive vs HIV-negative groups, regardless of pregnancy type (p = 0.3166). Similarly, no statistical significance was noted across all study groups (p = 0.0774). CONCLUSION This study demonstrates that there is a strong correlation between the up-regulation of adipsin and PE and that adipsin is a promising biomarker to use as a diagnostic tool for PE.
Collapse
|
17
|
Matsuyama T, Tomimatsu T, Mimura K, Yagi K, Kawanishi Y, Kakigano A, Nakamura H, Endo M, Kimura T. Complement activation by an angiogenic imbalance leads to systemic vascular endothelial dysfunction: A new proposal for the pathophysiology of preeclampsia. J Reprod Immunol 2021; 145:103322. [PMID: 33887508 DOI: 10.1016/j.jri.2021.103322] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/18/2021] [Accepted: 04/13/2021] [Indexed: 10/21/2022]
Abstract
The underlying mechanism of preeclampsia by which an angiogenic imbalance results in systemic vascular endothelial dysfunction remains unclear. Complement activation directly induces endothelial dysfunction and is known to be involved in preeclampsia; nevertheless, the association between complement activation and angiogenic imbalance has not been established. This study aimed to evaluate whether angiogenic imbalance affects the expression and secretion of inhibitory complement factor H (CFH) in endothelial cells, resulting in complement activation and systemic vascular endothelial dysfunction. Viability of human umbilical vein endothelial cells (HUVECs) was assessed upon CFH knockdown by targeted-siRNA, and were incubated with complement factors. HUVECs were also treated with placental growth factor (PlGF) and/or soluble fms-like tyrosine kinase 1 (sFlt1), and CFH expression and secretion were measured. These cells were evaluated by cell viability assay and cell surface complement activation was quantified by immunocytochemical assessment of C5b-9 deposition. HUVECs transfected with CFH-siRNA had significantly lower viability than that of control cells. Moreover, the expression and secretion of CFH were significantly increased upon PlGF treatment compared with PlGF + sFlt1 combo. HUVECs treated with PlGF had less C5b-9 deposition and higher viability than HUVECs treated with PlGF + sFlt1. In summary, CFH was found to be essential for endothelial cell survival by inhibiting complement activation. An angiogenic imbalance, including decreased PlGF and increased sFlt1, suppresses CFH expression and secretion, resulting in complement activation on the surface of endothelial cells and systemic vascular endothelial dysfunction.
Collapse
Affiliation(s)
- Tatsuya Matsuyama
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Takuji Tomimatsu
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.
| | - Kazuya Mimura
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kazunobu Yagi
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoko Kawanishi
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Aiko Kakigano
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hitomi Nakamura
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Masayuki Endo
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Tadashi Kimura
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
18
|
Girardi G, Lingo JJ, Fleming SD, Regal JF. Essential Role of Complement in Pregnancy: From Implantation to Parturition and Beyond. Front Immunol 2020; 11:1681. [PMID: 32849586 PMCID: PMC7411130 DOI: 10.3389/fimmu.2020.01681] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 06/23/2020] [Indexed: 12/12/2022] Open
Abstract
The complement cascade was identified over 100 years ago, yet investigation of its role in pregnancy remains an area of intense research. Complement inhibitors at the maternal-fetal interface prevent inappropriate complement activation to protect the fetus. However, this versatile proteolytic cascade also favorably influences numerous stages of pregnancy, including implantation, fetal development, and labor. Inappropriate complement activation in pregnancy can have adverse lifelong sequelae for both mother and child. This review summarizes the current understanding of complement activation during all stages of pregnancy. In addition, consequences of complement dysregulation during adverse pregnancy outcomes from miscarriage, preeclampsia, and pre-term birth are examined. Finally, future research directions into complement activation during pregnancy are considered.
Collapse
Affiliation(s)
- Guillermina Girardi
- Department of Basic Medical Sciences, College of Medicine, Member of QU Health, Qatar University, Doha, Qatar
| | - Joshua J Lingo
- Division of Biology, Kansas State University, Manhattan, KS, United States
| | - Sherry D Fleming
- Division of Biology, Kansas State University, Manhattan, KS, United States
| | - Jean F Regal
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, United States
| |
Collapse
|