1
|
Smit ER, Romijn M, Langerhorst P, van der Zwaan C, van der Staaij H, Rotteveel J, van Kaam AH, Fustolo-Gunnink SF, Hoogendijk AJ, Onland W, Finken MJJ, van den Biggelaar M. Distinct protein patterns related to postnatal development in small for gestational age preterm infants. Pediatr Res 2024:10.1038/s41390-024-03481-0. [PMID: 39152333 DOI: 10.1038/s41390-024-03481-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 07/18/2024] [Accepted: 07/30/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND Preterm infants, especially those born small for gestational age (SGA), are at risk of short-term and long-term health complications. Characterization of changes in circulating proteins postnatally in preterm infants may provide valuable fundamental insights into this population. Here, we investigated postnatal developmental patterns in preterm infants and explored protein signatures that deviate between SGA infants and appropriate for gestational age (AGA) infants using a mass spectrometry (MS)-based proteomics workflow. METHODS Longitudinal serum samples obtained at postnatal days 0, 3, 7, 14, and 28 from 67 preterm infants were analyzed using unbiased MS-based proteomics. RESULTS 314 out of 833 quantified serum proteins change postnatally, including previously described age-related changes in immunoglobulins, hemoglobin subunits, and new developmental patterns, e.g. apolipoproteins (APOA4) and terminal complement cascade (C9) proteins. Limited differences between SGA and AGA infants were found at birth while longitudinal monitoring revealed 69 deviating proteins, including insulin-sensitizing hormone adiponectin, platelet proteins, and 24 proteins with an annotated function in the immune response. CONCLUSIONS This study shows the potential of MS-based serum profiling in defining circulating protein trajectories in the preterm infant population and its ability to identify longitudinal alterations in protein levels associated with SGA. IMPACT Postnatal changes of circulating proteins in preterm infants have not fully been elucidated but may contribute to development of health complications. Mass spectrometry-based analysis is an attractive approach to study circulating proteins in preterm infants with limited material. Longitudinal plasma profiling reveals postnatal developmental-related patterns in preterm infants (314/833 proteins) including previously described changes, but also previously unreported proteins. Longitudinal monitoring revealed an immune response signature between SGA and AGA infants. This study highlights the importance of taking postnatal changes into account for translational studies in preterm infants.
Collapse
Affiliation(s)
- Eva R Smit
- Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands
| | - Michelle Romijn
- Department of Neonatology, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, the Netherlands
- Department of Pediatric Endocrinology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Pieter Langerhorst
- Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands
| | - Carmen van der Zwaan
- Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands
| | - Hilde van der Staaij
- Sanquin Research & Lab Services, Sanquin Blood Supply Foundation, Amsterdam, the Netherlands
- Department of Pediatrics, Division of Neonatology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, the Netherlands
- Department of Pediatric Hematology, Emma Children's Hospital, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Joost Rotteveel
- Amsterdam Reproduction and Development Research Institute, Amsterdam, the Netherlands
- Department of Pediatric Endocrinology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Anton H van Kaam
- Department of Neonatology, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, the Netherlands
| | - Suzanne F Fustolo-Gunnink
- Sanquin Research & Lab Services, Sanquin Blood Supply Foundation, Amsterdam, the Netherlands
- Department of Pediatrics, Division of Neonatology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, the Netherlands
- Department of Pediatric Hematology, Emma Children's Hospital, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Arie J Hoogendijk
- Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands
| | - Wes Onland
- Department of Neonatology, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, the Netherlands
| | - Martijn J J Finken
- Amsterdam Reproduction and Development Research Institute, Amsterdam, the Netherlands
- Department of Pediatric Endocrinology, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | | |
Collapse
|
2
|
Guo L, Guo A, Lan X, Tian S, Sun F, Su Y, Chen ZJ, Cao Y, Li Y. Oligoasthenospermia is correlated with increased preeclampsia incidence in subfertile couples undergoing in vitro fertilization and embryo transfer: a secondary analysis of a randomized clinical trial. F&S SCIENCE 2024:S2666-335X(24)00055-7. [PMID: 39153572 DOI: 10.1016/j.xfss.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 08/12/2024] [Accepted: 08/12/2024] [Indexed: 08/19/2024]
Abstract
OBJECTIVE To evaluate whether intergroup differences in the risk of maternal pregnancy complications after in vitro fertilization (IVF) vary with male factor. DESIGN A post hoc exploratory secondary analysis of data from a multicenter, randomized, controlled noninferiority trial (NCT03118141). SETTING Academic fertility centers. PATIENT(S) A total of 1,131 subfertile women with complete recording of their male partner's semen parameters during the trial were enrolled. All participants underwent intracytoplasmic sperm injection followed by frozen embryo transfer (ET) as part of their assisted reproductive technology treatment protocol. INTERVENTION(S) Women were divided into the oligoasthenospermia (n = 405) and normospermia (n = 726) groups according to the quality of male sperm. MAIN OUTCOME MEASURE(S) Pregnancy complications, principally including the incidence of preeclampsia. RESULT(S) Notably, we found that the risk of maternal preeclampsia was significantly higher in the oligoasthenospermia group than in the normospermia group. After adjustments for confounding factors by multivariate logistic regression analysis, the incidence of preeclampsia in the oligoasthenospermia group was still significantly higher than that in the normospermia group (6.55% vs. 3.60%; odds ratio, 0.529; 95% confidence interval, 0.282-0.992). However, there were no significant differences in terms of embryo quality, cumulative live birth rate, other pregnancy complications, or neonatal outcomes between the 2 groups. CONCLUSION(S) Oligoasthenospermia was associated with a higher risk of maternal preeclampsia in subfertile couples undergoing IVF-ET treatment. In clinical practice, it is essential to thoroughly evaluate the sperm quality and quantity of male partners before IVF-ET. Further research is needed to establish the causal relationships between semen quality and adverse pregnancy complications, particularly preeclampsia, and explore potential interventions.
Collapse
Affiliation(s)
- Ling Guo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong, People's Republic of China; Medical Integration and Practice Center, Shandong University, Jinan, Shandong, People's Republic of China
| | - Anliang Guo
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong, People's Republic of China
| | - Xiangxin Lan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong, People's Republic of China; Medical Integration and Practice Center, Shandong University, Jinan, Shandong, People's Republic of China
| | - Siqi Tian
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong, People's Republic of China; Medical Integration and Practice Center, Shandong University, Jinan, Shandong, People's Republic of China
| | - Fengxuan Sun
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong, People's Republic of China; Medical Integration and Practice Center, Shandong University, Jinan, Shandong, People's Republic of China
| | - Yaxin Su
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong, People's Republic of China
| | - Zi-Jiang Chen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong, People's Republic of China
| | - Yongzhi Cao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong, People's Republic of China; Medical Integration and Practice Center, Shandong University, Jinan, Shandong, People's Republic of China
| | - Yan Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong, People's Republic of China; Medical Integration and Practice Center, Shandong University, Jinan, Shandong, People's Republic of China.
| |
Collapse
|
3
|
Javandoust Gharehbagh F, Soltani-Zangbar MS, Yousefzadeh Y. Immunological mechanisms in preeclampsia: A narrative review. J Reprod Immunol 2024; 164:104282. [PMID: 38901108 DOI: 10.1016/j.jri.2024.104282] [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: 04/29/2024] [Revised: 06/03/2024] [Accepted: 06/15/2024] [Indexed: 06/22/2024]
Abstract
Maternal immunologic mechanisms for tolerance are essential for a successful pregnancy because they prevent maladaptive immune responses to the placenta and semi-allogeneic fetus and promote fetal growth. Preeclampsia is a major global cause of fetal mortality and morbidity. It is characterized by new-onset hypertension and proteinuria that occurs at twenty weeks of pregnancy or later. Preeclampsia is defined by a rise in cytokines that are pro-inflammatory and antiangiogenic components in the fetoplacental unit and the vascular endothelium of pregnant women, as well as an excessive and increasing stimulation of the immune system. Crucially, inflammation can result in low birth weight and inadequate placental perfusion in neonates. Preeclampsia, which is ultimately connected to inflammatory responses, can be impacted by several immunological mechanisms. Our goal in this work was to compile the most recent research on the pathoimmunology of preeclampsia, including studies on angiogenic variables and, in particular, immunological components.
Collapse
Affiliation(s)
| | - Mohammad Sadegh Soltani-Zangbar
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
4
|
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
|
5
|
Liao W, Zeng H, Jiang X, Deng X, Tu S, Lan H, Tang L, Dong W, Ding C. CircPAPPA2 plays a role in preeclampsia pathogenesis via regulation of the miR-942/miR-5006-3p. BMC Pregnancy Childbirth 2024; 24:414. [PMID: 38849756 PMCID: PMC11157718 DOI: 10.1186/s12884-024-06560-6] [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: 01/19/2024] [Accepted: 05/02/2024] [Indexed: 06/09/2024] Open
Abstract
CircRNAs are a class of endogenous non-coding RNAs implicated in the pathogenesis of many pregnancy related diseases, one of which is pre-eclampsia (PE). This study aims to investigate the role of CircPAPPA2 (circbase ID: hsa_circ_0015382) in regulating the migration and invasion of trophoblast cells. RNA sequencing was used to identify the differentially expressed circRNAs in placenta of PE and normal pregnant women. Quantitative polymerase chain reaction (qRT-PCR) was used to verify the expression of circPAPPA2 and two miRNAs (miR-942-5p, 5006-3p) in placenta of PE and normal pregnant women. CCK8 and transwell experiments were performed to assess the function of circPAPPA2 in PE development.The interaction between circPAPPA2 and miR-942-5p/miR-5006-3p was verified by dual-luciferase reporter assay. Finally, bioinformatics analyzed with gene ontology, Kyoto Encyclopedia of the target genes. The results showed that the expression of circPAPPA2 was increased in placenta of PE pregnant women. Also, circPAPPA2 impedes trophoblasts cell proliferation and invasion. Moreover, the expression of circPAPPA2 was positively correlated with systolic blood pressure and urine protein. In addition, circPAPPA2 serves as a sponge of miR-942-5p and miR-5006-3p. In conclusion, CircPAPPA2 regulates trophoblasts cell proliferation and invasion by mediating the miR-942/miR-5006-3p.
Collapse
Affiliation(s)
- Wenyan Liao
- Department of Gynaecology and Obstetrics, Hengyang Medical School, The First Affiliated Hospital, University of South China. NO.69, Chuanshan Road, Hengyang, 421001, Hunan, China
| | - Huan Zeng
- Department of Gynaecology and Obstetrics, Hengyang Medical School, The First Affiliated Hospital, University of South China. NO.69, Chuanshan Road, Hengyang, 421001, Hunan, China
| | - Xinmiao Jiang
- Department of Hepatopancreatobiliary Surgery, Hengyang Medical School, The First Affiliated Hospital, University of South China. NO.69, Chuanshan Road, Hengyang, 421001, Hunan, China
| | - Xin Deng
- Department of Hepatopancreatobiliary Surgery, Hengyang Medical School, The First Affiliated Hospital, University of South China. NO.69, Chuanshan Road, Hengyang, 421001, Hunan, China
| | - Shun Tu
- Department of Hepatopancreatobiliary Surgery, Hengyang Medical School, The First Affiliated Hospital, University of South China. NO.69, Chuanshan Road, Hengyang, 421001, Hunan, China
| | - Hui Lan
- Department of Gynaecology and Obstetrics, Hengyang Medical School, The First Affiliated Hospital, University of South China. NO.69, Chuanshan Road, Hengyang, 421001, Hunan, China
| | - Lingling Tang
- Department of Gynaecology and Obstetrics, Hengyang Medical School, The First Affiliated Hospital, University of South China. NO.69, Chuanshan Road, Hengyang, 421001, Hunan, China
| | - Weilei Dong
- Department of Gynaecology and Obstetrics, Hengyang Medical School, The First Affiliated Hospital, University of South China. NO.69, Chuanshan Road, Hengyang, 421001, Hunan, China.
| | - Chengming Ding
- Department of Hepatopancreatobiliary Surgery, Hengyang Medical School, The First Affiliated Hospital, University of South China. NO.69, Chuanshan Road, Hengyang, 421001, Hunan, China.
| |
Collapse
|
6
|
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
|
7
|
Wei X, Yang X. The novel role of activating receptor KIR2DS5 in preeclampsia. Int Immunopharmacol 2023; 125:111087. [PMID: 37864908 DOI: 10.1016/j.intimp.2023.111087] [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: 08/14/2023] [Revised: 10/02/2023] [Accepted: 10/15/2023] [Indexed: 10/23/2023]
Abstract
Preeclampsia (PE) is a serious complication of pregnancy. Decidual natural killer (dNK) cells were reported to participate in the remodeling of spiral arteries through producing a group of cytokines, including granulocyte-macrophage colony stimulating factor (GM-CSF). KIR2DS5 is an activating receptor of NK cells that specifically recognizes HLA-C2 on trophoblasts. Currently, there are no reports regarding the precise mechanism of KIR2DS5 in PE. This study included 30 PE patients and 30 healthy pregnant women. We found that the expressions of KIR2DS5 were significantly lower in PE deciduae compared to those of healthy pregnancies. By transfecting knockdown and overexpression lentivirus vectors of KIR2DS5 into dNK cells isolated from deciduae of early pregnancy, we altered the KIR2DS5 expression level in dNK cells. Then, these dNK cells and trophoblast cell lines were co-cultured as trophoblast-dNK cells. In the trophoblast-dNK cells, we examined the influence of KIR2DS5 on the biological manifestations of trophoblasts. As anticipated, overexpression of KIR2DS5 could facilitate cell proliferation, migration, and invasion. Furthermore, increased expression of KIR2DS5 inhibited cell apoptosis and enhanced the progression of cells from theG1 to theS stage. Further mechanistic study demonstrated a positive relationship between KIR2DS5 and GM-CSF in trophoblast-dNK cells. Accordingly, our observations indicated that a decrease in KIR2DS5 could reduce the expression of GM-CSF via the JAK2/STAT5 pathway, resulting in the failure of the activated signal to be transmitted to dNK cells and ultimately leading to the occurrence of PE. KIR2DS5 may be a new contributor for the prediction and diagnosis of PE.
Collapse
Affiliation(s)
- Xiaoqi Wei
- Department of Obstetrics, The First Hospital of China Medical University, Shenyang 110000, China
| | - Xiuhua Yang
- Department of Obstetrics, The First Hospital of China Medical University, Shenyang 110000, China.
| |
Collapse
|
8
|
Guan S, Bai X, Ding J, Zhuang R. Circulating inflammatory cytokines and hypertensive disorders of pregnancy: a two-sample Mendelian randomization study. Front Immunol 2023; 14:1297929. [PMID: 38035087 PMCID: PMC10687474 DOI: 10.3389/fimmu.2023.1297929] [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: 09/20/2023] [Accepted: 11/06/2023] [Indexed: 12/02/2023] Open
Abstract
Background Hypertensive disorders of pregnancy (HDP) pose a significant risk to maternal and fetal well-being; however, the etiology and pathogenesis of HDP remain ambiguous. It is now widely acknowledged that inflammatory response and the immune system are closely related to HDP. Previous research has identified several inflammatory cytokines are associated with HDP. This study applied Mendelian randomization (MR) analysis to further assess causality. Methods Patients with HDP who participated in the MR analysis presented with four types of HDP: pre-eclampsia or eclampsia (PE); gestational hypertension (GH); pre-existing hypertension complicating pregnancy, childbirth and the puerperium (EH); and pre-eclampsia or poor fetal growth (PF). A two-sample MR analysis was used to analyze the data in the study. The causal relationship between exposure and outcome was analyzed with inverse variance weighting (IVW), MR Egger, weighted median, weighted mode, and simple mode methods, where IVW was the primary method employed. Results Our MR analysis demonstrated a reliable causative effect of Interleukin-9 (IL-9) and macrophage migration inhibitory factor (MIF) on reducing HDP risk, while macrophage inflammatory protein 1-beta (MIP1b), Interleukin-13 (IL-13), and Interleukin-16 (IL-16) were associated with promoting HDP risk. Conclusions This study demonstrated that IL-9, MIF, MIP1b, IL-13, and IL-16 may be cytokines associated with the etiology of HDP, and that a number of inflammatory cytokines are probably involved in the progression of HDP. Additionally, our study revealed that these inflammatory cytokines have causal associations with HDP and may likely be potential therapeutic targets for HDP.
Collapse
Affiliation(s)
| | | | | | - Rujin Zhuang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
9
|
Gumilar KE, Priangga B, Lu CH, Dachlan EG, Tan M. Iron metabolism and ferroptosis: A pathway for understanding preeclampsia. Biomed Pharmacother 2023; 167:115565. [PMID: 37751641 DOI: 10.1016/j.biopha.2023.115565] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 09/28/2023] Open
Abstract
Preeclampsia (PE) is a serious medical condition that poses a significant health risk to women and children worldwide, particularly in the middle- and low-income countries. It is a complex syndrome that occurs as a result of abnormal pregnancy. Hypertension is the most common symptom of PE, with proteinuria and specific organ systems as detrimental targets. PE's pathogenesis is diverse, and its symptoms can overlap with other diseases. In early pregnancy, when the placenta takes over control, oxidative stress may be closely associated with ferroptosis, a type of cell death caused by intracellular iron accumulation. Ferroptosis in the placenta is defined by redox-active iron availability, loss of antioxidant capacity and phospholipids containing polyunsaturated fatty acids (PUFA) oxidation. Recent studies suggest a compelling potential link between ferroptosis and PE. In this article, we comprehensively review the current understanding of PE and discuss one of its emerging underlying mechanisms, the ferroptosis pathway. We also provide perspective and analysis on the implications of this process in the diagnosis, prevention, and treatment of preeclampsia. We aim to bridge the gap between clinicians and basic scientists in understanding this harmful disease and challenge the research community to put more effort into this exciting new area.
Collapse
Affiliation(s)
- Khanisyah Erza Gumilar
- Graduate Institute of Biomedical Science, China Medical University, Taichung 406040, Taiwan, ROC; Department of Obstetrics and Gynecology, Faculty of Medicine, Universitas Airlangga, Surabaya 60115, Indonesia
| | - Bayu Priangga
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universitas Airlangga, Surabaya 60115, Indonesia
| | - Chien-Hsing Lu
- Department of Obstetrics and Gynecology, Taichung Veterans General Hospital, Taichung 40705, Taiwan, ROC
| | - Erry Gumilar Dachlan
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universitas Airlangga, Surabaya 60115, Indonesia
| | - Ming Tan
- Graduate Institute of Biomedical Science, China Medical University, Taichung 406040, Taiwan, ROC; Institute of Biochemistry & Molecular Biology, and Research Center for Cancer Biology, China Medical University, Taichung 406040, Taiwan, ROC.
| |
Collapse
|
10
|
Rduch T, Arn N, Kinkel J, Fischer T, Binet I, Hornung R, Herrmann IK. Magnetic blood purification-based soluble fms-like tyrosine kinase-1 removal in comparison with dextran sulfate apheresis and therapeutic plasma exchange. Artif Organs 2023; 47:1309-1318. [PMID: 36995348 DOI: 10.1111/aor.14531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 03/17/2023] [Accepted: 03/23/2023] [Indexed: 03/31/2023]
Abstract
BACKGROUND Preeclampsia remains one of the most serious complications of pregnancy. Effective therapies are yet to be developed. Recent research has identified an imbalance of angiogenic and antiangiogenic factors as a root cause of preeclampsia. In particular, soluble fms-like tyrosine kinase-1 (sFlt-1) has been shown to bind the angiogenic factors vascular endothelial growth factor (VEGF) and placental growth factor (PlGF), reducing blood vessel growth. Increasing preclinical and clinical evidence suggests that removal of the sFlt-1 protein may benefit patients with early onset preeclampsia. sFlt-1 may be removed by conventional blood purification techniques, such as therapeutic plasma exchange (TPE) and dextran sulfate apheresis (DSA), or emerging technologies, including extracorporeal magnetic blood purification (MBP). METHODS We compare the performance and selectivity of TPE, DSA, and MBP for the therapeutic removal of sFlt-1. For MPB, we employ magnetic nanoparticles functionalized with either sFlt-1 antibodies or the sFlt-1-binding partner, vascular endothelial growth factor (VEGF). RESULTS We demonstrate that sFlt-1 removal by MBP is feasible and significantly more selective than TPE and DSA at comparable sFlt-1 removal efficiencies (MBP 96%, TPE 92%, DSA 78%). During both TPE and DSA, complement factors (incl. C3c and C4) are depleted to a considerable extent (-90% for TPE, -55% for DSA), while in MBP, complement factor concentrations remain unaltered. We further demonstrate that the removal efficacy of sFlt-1 in the MBP approach is strongly dependent on the nanoparticle type and dose and can be optimized to reach clinically feasible throughputs. CONCLUSIONS Taken together, the highly selective removal of sFlt-1 and potential other disease-causing factors by extracorporeal magnetic blood purification may offer new prospects for preeclamptic patients.
Collapse
Affiliation(s)
- Thomas Rduch
- Department of Gynaecology, Cantonal Hospital St. Gallen, Rorschacherstrasse 95, St. Gallen, Switzerland
- Laboratory for Particles-Biology Interactions, Department of Materials Meet Life, Swiss Federal Laboratories for Materials Science and Technology (Empa), Lerchenfeldstrasse 5, St. Gallen, Switzerland
| | - Norbert Arn
- Clinic for Nephrology and Transplant Medicine, Cantonal Hospital St.Gallen, Rorschacherstrasse 95, St.Gallen, Switzerland
| | - Janis Kinkel
- Department of Gynaecology, Cantonal Hospital St. Gallen, Rorschacherstrasse 95, St. Gallen, Switzerland
| | - Tina Fischer
- Department of Gynaecology, Cantonal Hospital St. Gallen, Rorschacherstrasse 95, St. Gallen, Switzerland
| | - Isabelle Binet
- Clinic for Nephrology and Transplant Medicine, Cantonal Hospital St.Gallen, Rorschacherstrasse 95, St.Gallen, Switzerland
| | - René Hornung
- Department of Gynaecology, Cantonal Hospital St. Gallen, Rorschacherstrasse 95, St. Gallen, Switzerland
| | - Inge K Herrmann
- Laboratory for Particles-Biology Interactions, Department of Materials Meet Life, Swiss Federal Laboratories for Materials Science and Technology (Empa), Lerchenfeldstrasse 5, St. Gallen, Switzerland
- Department of Mechanical and Process Engineering, ETH Zurich, Nanoparticle Systems Engineering Laboratory, Institute of Energy and Process Engineering, Sonneggstrasse 3, Zurich, Switzerland
| |
Collapse
|
11
|
Sakowicz A, Bralewska M, Rybak-Krzyszkowska M, Grzesiak M, Pietrucha T. New Ideas for the Prevention and Treatment of Preeclampsia and Their Molecular Inspirations. Int J Mol Sci 2023; 24:12100. [PMID: 37569476 PMCID: PMC10418829 DOI: 10.3390/ijms241512100] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Preeclampsia (PE) is a pregnancy-specific disorder affecting 4-10% of all expectant women. It greatly increases the risk of maternal and foetal death. Although the main symptoms generally appear after week 20 of gestation, scientific studies indicate that the mechanism underpinning PE is initiated at the beginning of gestation. It is known that the pathomechanism of preeclampsia is strongly related to inflammation and oxidative stress, which influence placentation and provoke endothelial dysfunction in the mother. However, as of yet, no "key players" regulating all these processes have been discovered. This might be why current therapeutic strategies intended for prevention or treatment are not fully effective, and the only effective method to stop the disease is the premature induction of delivery, mostly by caesarean section. Therefore, there is a need for further research into new pharmacological strategies for the treatment and prevention of preeclampsia. This review presents new preventive methods and therapies for PE not yet recommended by obstetrical and gynaecological societies. As many of these therapies are in preclinical studies or under evaluation in clinical trials, this paper reports the molecular targets of the tested agents or methods.
Collapse
Affiliation(s)
- Agata Sakowicz
- Department of Medical Biotechnology, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland; (M.B.); (T.P.)
| | - Michalina Bralewska
- Department of Medical Biotechnology, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland; (M.B.); (T.P.)
| | - Magda Rybak-Krzyszkowska
- Department of Obstetrics and Perinatology, University Hospital in Krakow, 31-501 Krakow, Poland;
| | - Mariusz Grzesiak
- Department of Perinatology, Obstetrics and Gynecology, Polish Mother’s Memorial Hospital-Research Institute in Lodz, 93-338 Lodz, Poland;
- Department of Gynecology and Obstetrics, Medical University of Lodz, 93-338 Lodz, Poland
| | - Tadeusz Pietrucha
- Department of Medical Biotechnology, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland; (M.B.); (T.P.)
| |
Collapse
|
12
|
Blakey H, Sun R, Xie L, Russell R, Sarween N, Hodson J, Hargitai B, Marton T, A H Neil D, Wong E, Sheerin NS, Bramham K, Harris CL, Knox E, Drayson M, Lipkin G. Pre-eclampsia is associated with complement pathway activation in the maternal and fetal circulation, and placental tissue. Pregnancy Hypertens 2023; 32:43-49. [PMID: 37088032 DOI: 10.1016/j.preghy.2023.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 02/21/2023] [Accepted: 04/11/2023] [Indexed: 04/25/2023]
Abstract
OBJECTIVES Pre-eclampsia (PE) is a leading cause of obstetric morbidity, with no definitive therapy other than delivery. We aimed to compare complement markers in maternal and fetal circulation, and placental tissue, between women with PE and healthy pregnant controls. STUDY DESIGN Maternal and umbilical cord blood was tested for iC3b, C3, C4, properdin, Ba and C5b-9, and placental tissue for C3d, C4d, C9 and C1q, from women with PE (n = 34) and healthy pregnant controls (n = 33). Maternal properdin and Ba tests were repeated in a separate validation cohort (PE n = 35; healthy pregnant controls n = 35). MAIN OUTCOME MEASURES Complement concentrations in maternal and umbilical cord blood, and placental immunohistochemical complement deposition. RESULTS Women with PE had significantly lower concentrations of properdin (mean: 4828 vs 6877 ng/ml, p < 0.001) and C4 (mean: 0.20 vs 0.31 g/l, p < 0.001), and higher Ba (median: 150 vs 113 ng/ml, p = 0.012), compared to controls. After controlling for gestational age at blood draw, average properdin concentration was 1945 ng/ml lower in PE vs controls (95 % CI: 1487-2402, p < 0.001). Of the cord blood markers assessed, only Ba differed significantly between PE and controls (median: 337 vs 233 ng/ml, p = 0.004). C4d staining of the syncytiotrophoblast membrane was increased in PE vs controls (median immunoreactivity score 3 vs 0, p < 0.001). Maternal properdin and C4 were significantly negatively correlated with placental C4d staining. CONCLUSIONS Our data confirm excessive placental complement deposition associated with significant concurrent changes in maternal and fetal circulating complement biomarkers in PE. Inhibition of complement activation is a potential therapeutic target.
Collapse
Affiliation(s)
- Hannah Blakey
- Renal Medicine Department, Queen Elizabeth Hospital Birmingham, Birmingham, UK; Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK.
| | - Ruyue Sun
- Translational and Clinical Research Institute, Newcastle University, Newcastle, UK
| | - Long Xie
- Translational and Clinical Research Institute, Newcastle University, Newcastle, UK
| | - Rebecca Russell
- Translational and Clinical Research Institute, Newcastle University, Newcastle, UK
| | - Nadia Sarween
- Renal Medicine Department, Queen Elizabeth Hospital Birmingham, Birmingham, UK
| | - James Hodson
- Research Development and Innovation, Institute of Translational Medicine, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Beata Hargitai
- Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - Tamas Marton
- Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - Desley A H Neil
- Renal Medicine Department, Queen Elizabeth Hospital Birmingham, Birmingham, UK
| | - Edwin Wong
- National Renal Complement Therapeutics Centre, Newcastle, UK
| | - Neil S Sheerin
- National Renal Complement Therapeutics Centre, Newcastle, UK
| | - Kate Bramham
- Department of Women and Children's Health, King's College London, London, UK
| | - Claire L Harris
- Translational and Clinical Research Institute, Newcastle University, Newcastle, UK
| | - Ellen Knox
- Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - Mark Drayson
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Graham Lipkin
- Renal Medicine Department, Queen Elizabeth Hospital Birmingham, Birmingham, UK
| |
Collapse
|
13
|
Herrock O, Deer E, Amaral LM, Campbell N, Whitney D, Ingram N, Cornelius DC, Turner T, Hardy-Hardin J, Booz GW, Ibrahim T, LaMarca B. Inhibiting B cell activating factor attenuates preeclamptic symptoms in placental ischemic rats. Am J Reprod Immunol 2023; 89:e13693. [PMID: 36794639 PMCID: PMC10009902 DOI: 10.1111/aji.13693] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/31/2023] [Accepted: 02/08/2023] [Indexed: 02/17/2023] Open
Abstract
PROBLEM Preeclampsia (PE), new-onset hypertension during pregnancy, is associated with a pro-inflammatory state with activated T cells, cytolytic natural killer (NK) cells, dysregulated complement proteins, and B cells secreting agonistic autoantibodies to the angiotensin II type-1 receptor (AT1-AA). The reduced uterine perfusion pressure (RUPP) model of placental ischemia recapitulates these features of PE. Blocking CD40L-CD40 communication between T and B cells or B cell depletion with Rituximab prevents hypertension and AT1-AA production in RUPP rats. This suggests that T cell-dependent B cell activation contributes to the hypertension and AT1-AA associated with PE. B2 cells maturing into antibody producing plasma cells are the product of T cell-dependent B cell-interactions and B cell Activating Factor (BAFF) is an integral cytokine in the development of B2 cells specifically. Thus, we hypothesize that BAFF blockade will selectively deplete B2 cells, therefore reducing blood pressure, AT1-AA, activated NK Cells, and complement in the RUPP rat model of PE. METHOD OF STUDY Gestational Day (GD) 14 pregnant rats underwent the RUPP procedure, and a subset were treated with 1 mg/kg Anti-BAFF antibodies via jugular catheters. On GD19, blood pressure was measured, B cells and NK cells were measured by flow cytometry, AT1-AA was measured by cardiomyocyte bioassay, and complement activation was measured by ELISA. RESULTS Anti-BAFF therapy attenuated hypertension, AT1-AA, NK cell activation, and APRIL levels in RUPP rats without negatively impacting fetal outcomes. CONCLUSIONS This study demonstrates that B2 cells contribute to hypertension, AT1-AA, and NK cell activation in response to placental ischemia during pregnancy.
Collapse
Affiliation(s)
- Owen Herrock
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Evangeline Deer
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Lorena M. Amaral
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Nathan Campbell
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Darby Whitney
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Nicole Ingram
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | | | - Ty Turner
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Ja’Nasa Hardy-Hardin
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - George W. Booz
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Tarek Ibrahim
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
| | - Babbette LaMarca
- Department of Pharmacology & Toxicology, University of Mississippi Medical Center, Jackson, MS
- Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, MS
| |
Collapse
|
14
|
Liu J, Yao L, Wang Y. Resveratrol alleviates preeclampsia-like symptoms in rats through a mechanism involving the miR-363-3p/PEDF/VEGF axis. Microvasc Res 2023; 146:104451. [PMID: 36368448 DOI: 10.1016/j.mvr.2022.104451] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 10/09/2022] [Accepted: 11/02/2022] [Indexed: 11/11/2022]
Abstract
Preeclampsia (PE) is a hypertension-associated disease, and resveratrol (RES) is a polyphenol recognized to present beneficial effects in cardiovascular disease including hypertension. Recently, attentions have come to the therapeutic effect of RES in PE, but the underlying molecular mechanisms remain largely unknown. This study sought to delineate the mechanistic basis regarding bioinformatically identified miR-363-3p/PEDF/VEGF axis for RES treatment in PE. PE-like symptoms were induced in vivo in Sprague-Dawley rats by intraperitoneal injection with Ng-nitro-L-arginine methyl ester (L-NAME), and hypoxia was induced in vitro in trophoblasts by CoCl2. Accordingly, RES was found to enhance viability, migration, angiogenesis, and to repress the apoptosis of hypoxic trophoblasts in vitro. Furthermore, in vivo experiments noted that RES alleviated placental injury and promoted angiogenesis in rats with PE-like symptoms in vivo by increasing VEGF via promoting miR-363-3p-mediated PEDF suppression. Collectively, RES ameliorates PE by upregulating VEGF through miR-363-3p-mediated PEDF downregulation, the mechanism of which may be of promising significance to augment RES efficacy in PE treatment.
Collapse
Affiliation(s)
- Jun Liu
- Department of Obstetrics and Gynecology, Pingxiang Maternity and Child Care Hospital, Pingxiang 337000, PR China.
| | - Lifeng Yao
- Department of Obstetrics and Gynecology, Pingxiang Maternity and Child Care Hospital, Pingxiang 337000, PR China
| | - Yu Wang
- Department of Obstetrics and Gynecology, Pingxiang Maternity and Child Care Hospital, Pingxiang 337000, PR China
| |
Collapse
|
15
|
Agbani EO, Skeith L, Lee A. Preeclampsia: Platelet procoagulant membrane dynamics and critical biomarkers. Res Pract Thromb Haemost 2023; 7:100075. [PMID: 36923708 PMCID: PMC10009545 DOI: 10.1016/j.rpth.2023.100075] [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: 11/02/2022] [Revised: 12/20/2022] [Accepted: 12/29/2022] [Indexed: 02/11/2023] Open
Abstract
A state-of-the-art lecture titled "Preeclampsia and Platelet Procoagulant Membrane Dynamics" was presented at the International Society on Thrombosis and Haemostasis (ISTH) Congress in 2022. Platelet activation is involved in the pathophysiology of preeclampsia and contributes to the prothrombotic state of the disorder. Still, it remains unclear what mechanisms initiate and sustain platelet activation in preeclampsia and how platelets drive the thrombo-hemorrhagic abnormalities in preeclampsia. Here, we highlight our findings that platelets in preeclampsia are preactivated possibly by plasma procoagulant agonist(s) and overexpress facilitative glucose transporter-3 (GLUT3) in addition to GLUT1. Preeclampsia platelets are also partially degranulated, procoagulant, and proaggregatory and can circulate as microaggregates/microthrombi. However, in response to exposed subendothelial collagen, such as in injured vessels during cesarean sections, preeclampsia platelets are unable to mount a full procoagulant response, contributing to blood loss perioperatively. The overexpression of GLUT3 or GLUT1 may be monitored alone or in combination (GLUT1/GLUT3 ratio) as a biomarker for preeclampsia onset, phenotype, and progression. Studies to further understand the mediators of the platelet activation and procoagulant membrane dynamics in preeclampsia can reveal novel drug targets and suitable alternatives to aspirin for the management of prothrombotic tendencies in preeclampsia. Finally, we summarize relevant new data on this topic presented during the 2022 ISTH Congress.
Collapse
Affiliation(s)
- Ejaife O. Agbani
- Department of Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Alberta, Canada
- Libin Cardiovascular Institute, Calgary, Alberta, Canada
- Correspondence Dr Ejaife O. Agbani, Department of Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, T2N 4N1 Alberta, Canada. @EjaifeAgbani
| | - Leslie Skeith
- Libin Cardiovascular Institute, Calgary, Alberta, Canada
- Division of Hematology and Hematological Malignancies, Department of Medicine, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Adrienne Lee
- Division of Hematology and Hematological Malignancies, Department of Medicine, Cumming School of Medicine, University of Calgary, Alberta, Canada
- Division of Hematology, Department of Medicine/Medical Oncology, University of British Columbia, Island Health, Victoria, Canada
| |
Collapse
|
16
|
Severe COVID-19 in pregnancy has a distinct serum profile, including greater complement activation and dysregulation of serum lipids. PLoS One 2022; 17:e0276766. [DOI: 10.1371/journal.pone.0276766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 10/12/2022] [Indexed: 11/17/2022] Open
Abstract
Background
Pregnancies complicated by Coronavirus Disease 2019 (COVID-19) are at an increased risk of severe morbidity due to physiologic changes in immunologic, cardiovascular, and respiratory function. There is little is known about how severity of COVID-19 changes protein and metabolite expression in pregnancy.
Objective
This study aims to investigate the pathophysiology behind various clinical trajectories in pregnant patients diagnosed with COVID-19 using multi-omics profiling.
Study design
This is a prospective cohort study of 30 pregnant patients at a single tertiary care center. Participants were categorized by severity of COVID-19 disease (control, asymptomatic, mild/moderate, or severe). Maternal serum samples underwent LC-MS-based multiomics analysis for profiling of proteins, lipids, electrolytes, and metabolites. Linear regression models were used to assess how disease severity related to analyte levels. Reactome pathway enrichment analysis was conducted on differential analytes.
Results
Of 30 participants, 25 had confirmed diagnosis of COVID-19 (6 asymptomatic (one post-infection), 13 mild/moderate (all post-infection), 6 severe), and 5 participants were controls. Severe COVID-19 was associated with distinct profiles demonstrating significant proteomic and lipidomic signatures which were enriched for annotations related to complement and antibody activity. (FDR < 0.05). Downregulated analytes were not significantly enriched but consisted of annotation terms related to lipoprotein activity (FDR > 0.2). Post-infection mild/moderate COVID-19 did not have significantly altered serum protein, metabolite, or lipid metabolite levels compared to controls.
Conclusions
Pregnancies with severe COVID-19 demonstrate greater inflammation and complement activation and dysregulation of serum lipids. This altered multiomic expression provides insight into the pathophysiology of severe COVID-19 in pregnancy and may serve as potential indicators for adverse pregnancy outcomes.
Collapse
|
17
|
Belmonte B, Mangogna A, Gulino A, Cancila V, Morello G, Agostinis C, Bulla R, Ricci G, Fraggetta F, Botto M, Garred P, Tedesco F. Distinct Roles of Classical and Lectin Pathways of Complement in Preeclamptic Placentae. Front Immunol 2022; 13:882298. [PMID: 35711467 PMCID: PMC9197446 DOI: 10.3389/fimmu.2022.882298] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/28/2022] [Indexed: 12/20/2022] Open
Abstract
Pre-eclampsia is a pregnancy complication characterized by defective vascular remodeling in maternal decidua responsible for reduced blood flow leading to functional and structural alterations in the placenta. We have investigated the contribution of the complement system to decidual vascular changes and showed that trophoblasts surrounding unremodeled vessels prevalent in preeclamptic decidua fail to express C1q that are clearly detected in cells around remodeled vessels predominant in control placenta. The critical role of C1q is supported by the finding that decidual trophoblasts of female C1qa-/- pregnant mice mated to C1qa+/+ male mice surrounding remodeled vessels express C1q of paternal origin. Unlike C1qa-/- pregnant mice, heterozygous C1qa+/- and wild type pregnant mice share a high percentage of remodeled vessels. C1q was also found in decidual vessels and stroma of normal placentae and the staining was stronger in preeclamptic placentae. Failure to detect placental deposition of C1r and C1s associated with C1q rules out complement activation through the classical pathway. Conversely, the intense staining of decidual endothelial cells and villous trophoblast for ficolin-3, MASP-1 and MASP-2 supports the activation of the lectin pathway that proceeds with the cleavage of C4 and C3 and the assembly of the terminal complex. These data extend to humans our previous findings of complement activation through the lectin pathway in an animal model of pre-eclampsia and provide evidence for an important contribution of C1q in decidual vascular remodeling.
Collapse
Affiliation(s)
- Beatrice Belmonte
- Tumor Immunology Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G. D’Alessandro”, University of Palermo, Palermo, Italy
- Pathology Unit, Azienda Sanitaria Provinciale (ASP) Catania, “Gravina” Hospital, Caltagirone, Italy
| | - Alessandro Mangogna
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Alessandro Gulino
- Tumor Immunology Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G. D’Alessandro”, University of Palermo, Palermo, Italy
| | - Valeria Cancila
- Tumor Immunology Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G. D’Alessandro”, University of Palermo, Palermo, Italy
| | - Gaia Morello
- Tumor Immunology Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties “G. D’Alessandro”, University of Palermo, Palermo, Italy
| | - Chiara Agostinis
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Roberta Bulla
- Department of Life Sciences, 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
| | - Filippo Fraggetta
- Pathology Unit, Azienda Sanitaria Provinciale (ASP) Catania, “Gravina” Hospital, Caltagirone, Italy
| | - Marina Botto
- Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
- Imperial Lupus Centre, Imperial College Healthcare National Health Service (NHS) Trust, London, United Kingdom
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Francesco Tedesco
- Istituto Auxologico Italiano, Laboratory of Immuno-Rheumatology, IRCCS, Milan, Italy
- *Correspondence: Francesco Tedesco,
| |
Collapse
|
18
|
Leimi L, Jahnukainen K, Olkinuora H, Meri S, Vettenranta K. Early vascular toxicity after pediatric allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant 2022; 57:705-711. [PMID: 35177827 PMCID: PMC9090633 DOI: 10.1038/s41409-022-01607-8] [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: 04/06/2021] [Revised: 01/27/2022] [Accepted: 02/01/2022] [Indexed: 11/08/2022]
Abstract
Treatment-related mortality and morbidity remain a challenge in hematopoietic stem cell transplantation (HSCT). In this retrospective, single-center study, we analyzed endothelial damage as a potential, common denominator and mechanism for the adverse effects. We evaluated the prevalence of key vascular complications and graft-versus-host disease among 122 pediatric patients with an allogeneic HSCT between 2001 and 2013. The spectrum and frequency of acute adverse events emerging ≤100 days post transplant were graded according to the CTCAE 4.03 and analyzed. We identified a total of 19/122 (15.6%) patients with vascular complications, fulfilling the criteria of capillary leak syndrome, veno-occlusive disease/sinusoidal obstruction syndrome or thrombotic microangiopathy. The patients had a poorer overall survival (77% versus 26%, p < 0.001). Nearly one half (56/122, 45.9%) had at least one, severe (grade 3 or 4) adverse event. Patients with vascular complications had more often edema/effusions (p = 0.023), thrombocytopenia (p = 0.001), gastrointestinal bleeding (p < 0.001), acute kidney injury (p < 0.001), ascites (p < 0.001) or bilirubin increase (p = 0.027). These endotheliopathy-related adverse events appeared early post HSCT, varied in their clinical phenotype and predicted a poor outcome. An unrelated donor but not previous exposure to leukemia or irradiation-based conditioning was identified as a risk factor for vascular complications and endotheliopathy.
Collapse
Affiliation(s)
- Lilli Leimi
- University of Helsinki, Helsinki University Hospital, Children's Hospital, and Pediatric Research Center, Helsinki, Finland.
| | - Kirsi Jahnukainen
- University of Helsinki, Helsinki University Hospital, Children's Hospital, and Pediatric Research Center, Helsinki, Finland
- NORDFERTIL Research Lab Stockholm, Department of Women's and Children's Health, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Helena Olkinuora
- University of Helsinki, Helsinki University Hospital, Children's Hospital, and Pediatric Research Center, Helsinki, Finland
| | - Seppo Meri
- Department of Bacteriology and Immunology and Translational Immunology Research Program, University of Helsinki, and Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Kim Vettenranta
- University of Helsinki, Helsinki University Hospital, Children's Hospital, and Pediatric Research Center, Helsinki, Finland
| |
Collapse
|
19
|
Active Management of Labor Process under Smart Medical Model Improves Vaginal Delivery Outcomes of Pregnant Women with Preeclampsia. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:8926335. [PMID: 35432840 PMCID: PMC9010162 DOI: 10.1155/2022/8926335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/09/2022] [Accepted: 03/26/2022] [Indexed: 11/18/2022]
Abstract
Background In a global environment of increasing cesarean delivery rate, promoting vaginal delivery, reducing the rate of first cesarean section, and the incidence of vaginal delivery complications are the objectives of obstetric medical quality and safety in China. As a common obstetric complication, preeclampsia affects the safety of many pregnant women. It is the obstetrician's great responsibility to promote vaginal delivery and improve delivery outcomes in preeclampsia. To this end, we explored the roles of active labor management under the smart medical model in improving the outcomes of vaginal delivery for pregnant women with preeclampsia. Methods The clinical data of 219 cases of preeclampsia pregnant women who delivered vaginally in our hospital from January 2017 to December 2020 were retrospectively analyzed. According to different labor process management, they were divided into study group (active labor process management group) and control group (normal labor process management group). Active labor process management methods included intrapartum ultrasound, central fetal heart rate monitoring, Doula delivery, labor analgesia, and quality of life care. The differences in delivery process, delivery outcome, bleeding causes, and hemostatic measures were compared between the two groups. Results (1) The incidence of preeclampsia in our hospital showed an increasing trend in recent four years; (2) in smart hospitals, the active management of labor process reduced the probability of transferring to the cesarean section in preeclampsia pregnant women with vaginal trial failure; and (3) active labor process management reduced the rate of lateral episiotomy, decreased the postpartum hemorrhage volume within two hours, and improved the vaginal delivery outcome of preeclampsia pregnant women. Conclusions In the era of the rapid development of the Internet, vigorously promoting the construction of smart hospitals and actively managing the delivery process can reduce the failure rate of vaginal trial delivery and improve the outcomes of vaginal delivery in preeclampsia women.
Collapse
|
20
|
Beer LA, Senapati S, Sammel MD, Barnhart KT, Schreiber CA, Speicher DW. Proteome-defined changes in cellular pathways for decidua and trophoblast tissues associated with location and viability of early-stage pregnancy. Reprod Biol Endocrinol 2022; 20:36. [PMID: 35189928 PMCID: PMC8862331 DOI: 10.1186/s12958-022-00908-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/01/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND In early pregnancy, differentiating between a normal intrauterine pregnancy (IUP) and abnormal gestations including early pregnancy loss (EPL) or ectopic pregnancy (EP) is a major clinical challenge when ultrasound is not yet diagnostic. Clinical treatments for these outcomes are drastically different making early, accurate diagnosis imperative. Hence, a greater understanding of the biological mechanisms involved in these early pregnancy complications could lead to new molecular diagnostics. METHODS Trophoblast and endometrial tissue was collected from consenting women having an IUP (n = 4), EPL (n = 4), or EP (n = 2). Samples were analyzed by LC-MS/MS followed by a label-free proteomics analysis in an exploratory study. For each tissue type, pairwise comparisons of different pregnancy outcomes (EPL vs. IUP and EP vs. IUP) were performed, and protein changes having a fold change ≥ 3 and a Student's t-test p-value ≤ 0.05 were defined as significant. Pathway and network classification tools were used to group significantly changing proteins based on their functional similarities. RESULTS A total of 4792 and 4757 proteins were identified in decidua and trophoblast proteomes. For decidua, 125 protein levels (2.6% of the proteome) were significantly different between EP and IUP, whereas EPL and IUP decidua were more similar with only 68 (1.4%) differences. For trophoblasts, there were 66 (1.4%) differences between EPL and IUP. However, the largest group of 344 differences (7.2%) was observed between EP and IUP trophoblasts. In both tissues, proteins associated with ECM remodeling, cell adhesion and metabolic pathways showed decreases in EP specimens compared with IUP and EPL. In trophoblasts, EP showed elevation of inflammatory and immune response pathways. CONCLUSIONS Overall, differences between an EP and IUP are greater than the changes observed when comparing ongoing IUP and nonviable intrauterine pregnancies (EPL) in both decidua and trophoblast proteomes. Furthermore, differences between EP and IUP were much higher in the trophoblast than in the decidua. This observation is true for the total number of protein changes as well as the extent of changes in upstream regulators and related pathways. This suggests that biomarkers and mechanisms of trophoblast function may be the best predictors of early pregnancy location and viability.
Collapse
Affiliation(s)
- Lynn A Beer
- Center for Systems & Computational Biology, The Wistar Institute, Philadelphia, PA, USA
| | - Suneeta Senapati
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA, USA
| | - Mary D Sammel
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO, USA
| | - Kurt T Barnhart
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Courtney A Schreiber
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA, USA
| | - David W Speicher
- Center for Systems & Computational Biology, The Wistar Institute, Philadelphia, PA, USA.
| |
Collapse
|
21
|
Man AWC, Zhou Y, Lam UDP, Reifenberg G, Werner A, Habermeier A, Closs EI, Daiber A, Münzel T, Xia N, Li H. L-citrulline ameliorates pathophysiology in a rat model of superimposed preeclampsia. Br J Pharmacol 2021; 179:3007-3023. [PMID: 34935131 DOI: 10.1111/bph.15783] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 12/08/2021] [Accepted: 12/15/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Preeclampsia, characterized by hypertension, proteinuria, and fetal growth restriction, is one of the leading causes of maternal and perinatal mortality. By far, there is no effective pharmacological therapy for preeclampsia. The present study was conducted to investigate the effects of L-citrulline supplementation in Dahl salt-sensitive rat, a model of superimposed preeclampsia. EXPERIMENTAL APPROACH Parental DSSR were treated with L-citrulline (2.5 g/L in drinking water) from the day of mating to the end of lactation period. Blood pressure of the rats was monitored throughout pregnancy and markers of preeclampsia were assessed. Endothelial function of the pregnant DSSR was assessed by wire myograph. KEY RESULTS L-citrulline supplementation significantly reduced maternal blood pressure, proteinuria, and levels of circulating soluble fms-like tyrosine kinase 1 in DSSR. L-citrulline improved maternal endothelial function by augmenting the production of nitric oxide in the aorta and improving endothelium-derived hyperpolarizing factor-mediated vasorelaxation in resistance arteries. L-citrulline supplementation improved placental insufficiency and fetal growth, which were associated with an enhancement of angiogenesis and reduction of fibrosis and senescence in the placentas. In addition, L-citrulline downregulated genes involved in the toll-like receptor 4 and nuclear factor-κB signaling pathway. CONCLUSION AND IMPLICATIONS This study shows that L-citrulline supplementation reduces gestational hypertension, improves placentation and fetal growth in a rat model of superimposed preeclampsia. L-citrulline supplementation may represent an effective and safe therapeutic strategy for preeclampsia that benefit both the mother and the fetus.
Collapse
Affiliation(s)
- Andy W C Man
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Yawen Zhou
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Uyen D P Lam
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany.,Biomedical Research Center, Pham Ngoc Thach University of Medicine, Ho Chi Minh City, Vietnam
| | - Gisela Reifenberg
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Anke Werner
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Alice Habermeier
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Ellen I Closs
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Andreas Daiber
- Department of Cardiology, Cardiology I, Johannes Gutenberg University Medical Center, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Thomas Münzel
- Department of Cardiology, Cardiology I, Johannes Gutenberg University Medical Center, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Ning Xia
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Huige Li
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| |
Collapse
|
22
|
Agostinis C, Mangogna A, Balduit A, Aghamajidi A, Ricci G, Kishore U, Bulla R. COVID-19, Pre-Eclampsia, and Complement System. Front Immunol 2021; 12:775168. [PMID: 34868042 PMCID: PMC8635918 DOI: 10.3389/fimmu.2021.775168] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/25/2021] [Indexed: 12/15/2022] Open
Abstract
COVID-19 is characterized by virus-induced injury leading to multi-organ failure, together with inflammatory reaction, endothelial cell (EC) injury, and prothrombotic coagulopathy with thrombotic events. Complement system (C) via its cross-talk with the contact and coagulation systems contributes significantly to the severity and pathological consequences due to SARS-CoV-2 infection. These immunopathological mechanisms overlap in COVID-19 and pre-eclampsia (PE). Thus, mothers contracting SARS-CoV-2 infection during pregnancy are more vulnerable to developing PE. SARS-CoV-2 infection of ECs, via its receptor ACE2 and co-receptor TMPRSS2, can provoke endothelial dysfunction and disruption of vascular integrity, causing hyperinflammation and hypercoagulability. This is aggravated by bradykinin increase due to inhibition of ACE2 activity by the virus. C is important for the progression of normal pregnancy, and its dysregulation can impact in the form of PE-like syndrome as a consequence of SARS-CoV-2 infection. Thus, there is also an overlap between treatment regimens of COVID-19 and PE. C inhibitors, especially those targeting C3 or MASP-2, are exciting options for treating COVID-19 and consequent PE. In this review, we examine the role of C, contact and coagulation systems as well as endothelial hyperactivation with respect to SARS-CoV-2 infection during pregnancy and likely development of PE.
Collapse
Affiliation(s)
- Chiara Agostinis
- Institute for Maternal and Child Health, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Burlo Garofolo, Trieste, Italy
| | - Alessandro Mangogna
- Institute for Maternal and Child Health, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Burlo Garofolo, Trieste, Italy
| | - Andrea Balduit
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Azin Aghamajidi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Giuseppe Ricci
- Institute for Maternal and Child Health, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Burlo Garofolo, Trieste, Italy.,Department of Medical, Surgical and Health Science, University of Trieste, Trieste, Italy
| | - Uday Kishore
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Roberta Bulla
- Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
23
|
Ancuța E, Zamfir R, Martinescu G, Crauciuc DV, Ancuța C. The Complement System, T Cell Response, and Cytokine Shift in Normotensive versus Pre-Eclamptic and Lupus Pregnancy. J Clin Med 2021; 10:jcm10245722. [PMID: 34945017 PMCID: PMC8705505 DOI: 10.3390/jcm10245722] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/02/2021] [Accepted: 12/05/2021] [Indexed: 12/14/2022] Open
Abstract
Successful pregnancy requires an immunological shift with T helper CD4+ bias based on disbalance Th1/Th17 versus Th2/T regulatory (Tregs) required to induce tolerance against the semi-allogeneic fetus and placenta and to support fetal growth. Considered a pregnancy-specific hypertensive disorder, pre-eclampsia is characterized by multifaceted organ involvement related to impaired maternal immune tolerance to paternal antigens triggered by hypoxic placental injury as well as excessive local and systemic anti-angiogenic and inflammatory factor synthesis. Both systemic and local Th1/Th2 shift further expands to Th17 cells and their cytokines (IL-17) complemented by suppressive Treg and Th2 cytokines (IL-10, IL-4); alterations in Th17 and Tregs cause hypertension during pregnancy throughout vasoactive factors and endothelial dysfunction, providing an explanatory link between immunological and vascular events in the pathobiology of pre-eclamptic pregnancy. Apart from immunological changes representative of normotensive pregnancy, lupus pregnancy is generally defined by higher serum pro-inflammatory cytokines, lower Th2 polarization, defective and lower number of Tregs, potential blockade of complement inhibitors by anti-phospholipid antibodies, and similar immune alterations to those seen in pre-eclampsia. The current review underpins the immune mechanisms of pre-eclampsia focusing on local (placental) and systemic (maternal) aberrant adaptive and innate immune response versus normotensive pregnancy and pregnancy in systemic autoimmune conditions, particularly lupus.
Collapse
Affiliation(s)
- Eugen Ancuța
- Research Department, “Elena Doamna” Obstetrics and Gynecology Clinical Hospital, 700398 Iași, Romania; (E.A.); (G.M.)
| | - Radu Zamfir
- Fundeni Clinical Institute, 022328 București, Romania;
| | - Gabriel Martinescu
- Research Department, “Elena Doamna” Obstetrics and Gynecology Clinical Hospital, 700398 Iași, Romania; (E.A.); (G.M.)
| | - Dragoș Valentin Crauciuc
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
- Correspondence: (D.V.C.); or (C.A.); Tel.: +40-740036387 (C.A.)
| | - Codrina Ancuța
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
- 2nd Rheumatology Department, Clinical Rehabilitation Hospital, 700661 Iași, Romania
- Correspondence: (D.V.C.); or (C.A.); Tel.: +40-740036387 (C.A.)
| |
Collapse
|
24
|
Mesenchymal stem cell therapy attenuates complement C3 deposition and improves the delicate equilibrium between angiogenic and anti-angiogenic factors in abortion-prone mice. Mol Immunol 2021; 141:246-256. [PMID: 34875452 DOI: 10.1016/j.molimm.2021.11.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 10/15/2021] [Accepted: 11/10/2021] [Indexed: 12/12/2022]
Abstract
Immunological disorders are one of the main causes of recurrent spontaneous abortions (RSA). A rapidly expanding body of evidence indicates that excessive activation of the complement system is critically involved in the development of miscarriages. In the CBA/J × DBA/2 murine model of recurrent miscarriage, exaggerated and unrestrained complement activation is reported to be the underlying cause of angiogenic factor imbalance and persistent inflammation. We have previously shown that mesenchymal stem cell (MSC) therapy can significantly reduce the abortion rate in abortion-prone mice through regulating the feto-maternal immune response. In the present study, we hypothesized that MSCs might improve the balance of angiogenic factors at the feto-maternal unit of CBA/J × DBA/2 mice by restraining complement activation and deposition. To explore this hypothesis, autologous adipose tissue-derived mesenchymal stem cells (AD-MSCs) were administered intra-peritoneally to abortion-prone mice on the 4.5th day of gestation. Control mice received PBS as vehicle. On day 13.5 of pregnancy, deposition of the complement component C3 and expression levels of Crry, CFD (adipsin), VEGF, PlGF and FLT-1 were measured at the feto-maternal interface by immunohistochemistry and real-time PCR, respectively. Decidual cells were also cultured in RPMI 1640 medium for 48 h and VEGF and sFLT-1 protein levels were quantified in supernatants using enzyme-linked immunosorbent assay (ELISA). Our results indicated that MSC therapy significantly reduced C3 deposition and adipsin transcription in the fetal-maternal interface of abortion-prone mice. Furthermore, administration of MSCs robustly upregulated the mRNA expression levels of Crry, VEGF, PlGF and FLT-1 in the placenta and decidua of CBA/J × DBA/2 mice. Consistently, the in vitro results demonstrated that decidual cells obtained from MSC-treated dams produced increased concentrations of VEGF in culture supernatants, with concomitant decreased levels of sFLT-1 protein. Here, we show for the first time that adoptive transfer of MSCs rectifies the disturbed balance of angiogenic factors observed at the feto-maternal unit of CBA/J × DBA/2 mice, in part at least, through inhibiting excessive complement activation and promoting the production of angiogenic factors. Collectively, these alterations seem to play a pivotal role in reducing the abortion rate and improving the intrauterine condition for the benefit of the fetus.
Collapse
|
25
|
Yang L, Wang L, Wu J, Wang H, Yang G, Zhang L. Changes in Expression of Complement Components in the Ovine Spleen during Early Pregnancy. Animals (Basel) 2021; 11:ani11113183. [PMID: 34827915 PMCID: PMC8614503 DOI: 10.3390/ani11113183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/02/2021] [Accepted: 11/04/2021] [Indexed: 02/06/2023] Open
Abstract
During early gestation in humans, complement regulation is essential for normal fetal growth. It is supposed that a complement pathway participates in maternal splenic immune regulation at the early stage of gestation in ewes. The aim of this study was to analyze the effects of early pregnancy on the expression of complement components in the maternal spleen of ewes. In this study, ovine spleens were sampled on day 16 of nonpregnancy, and days 13, 16 and 25 of gestation. RT-qPCR, Western blot and immunohistochemical analysis were used to detect the changes in expression of complement components in the ovine maternal spleens. Our results reveal that C1q was upregulated during early gestation, C1r, C1s, C2, C3 and C5b increased at day 25 of gestation and C4a and C9 peaked at days 13 and 16 of gestation. In addition, C3 protein was located in the capsule, trabeculae and splenic cords. In conclusion, our results show for the first time that there was modification in the expression of complement components in the ovine spleen at the early stage of gestation, and complement pathways may participate in modulating splenic immune responses at the early stage of gestation.
Collapse
|
26
|
Shangguan Y, Wang Y, Shi W, Guo R, Zeng Z, Hu W, Cai W, Yan Q, Xu Y, Tang D, Dai Y. Systematic proteomics analysis of lysine acetylation reveals critical features of placental proteins in pregnant women with preeclampsia. J Cell Mol Med 2021; 25:10614-10626. [PMID: 34697885 PMCID: PMC8581308 DOI: 10.1111/jcmm.16997] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 09/01/2021] [Accepted: 09/30/2021] [Indexed: 12/14/2022] Open
Abstract
Preeclampsia (PE) is a dangerous hypertensive disorder that occurs during pregnancy. The specific aetiology and pathogenesis of PE have yet to be clarified. To better reveal the specific pathogenesis of PE, we characterized the proteome and acetyl proteome (acetylome) profile of placental tissue from PE and normal-term pregnancy by label-free quantification proteomics technology and PRM analysis. In this research, 373 differentially expressed proteins (DEPs) were identified by proteome analysis. Functional enrichment analysis revealed significant enrichment of DEPs related to angiogenesis and the immune system. COL12A1, C4BPA and F13A1 may be potential biomarkers for PE diagnosis and new therapeutic targets. Additionally, 700 Kac sites were identified on 585 differentially acetylated proteins (DAPs) by acetylome analyses. These DAPs may participate in the occurrence and development of PE by affecting the complement and coagulation cascades pathway, which may have important implications for better understand the pathogenesis of PE. In conclusion, this study systematically analysed the reveals critical features of placental proteins in pregnant women with PE, providing a resource for exploring the contribution of lysine acetylation modification to PE.
Collapse
Affiliation(s)
- Yu Shangguan
- Clinical Medical Research CenterGuangdong Provincial Engineering Research Center of Autoimmune Disease Precision MedicineShenzhen Engineering Research Center of Autoimmune DiseaseThe Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and TechnologyShenzhen People's HospitalShenzhenChina
- Guangxi Key Laboratory of Metabolic Disease ResearchNephrology Department924st HospitalGuilinChina
- College of Life ScienceGuangxi Normal UniversityGuilinChina
| | - Yinglan Wang
- Clinical Medical Research CenterGuangdong Provincial Engineering Research Center of Autoimmune Disease Precision MedicineShenzhen Engineering Research Center of Autoimmune DiseaseThe Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and TechnologyShenzhen People's HospitalShenzhenChina
| | - Wei Shi
- Clinical Medical Research CenterGuangdong Provincial Engineering Research Center of Autoimmune Disease Precision MedicineShenzhen Engineering Research Center of Autoimmune DiseaseThe Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and TechnologyShenzhen People's HospitalShenzhenChina
| | - Ruonan Guo
- Clinical Medical Research CenterGuangdong Provincial Engineering Research Center of Autoimmune Disease Precision MedicineShenzhen Engineering Research Center of Autoimmune DiseaseThe Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and TechnologyShenzhen People's HospitalShenzhenChina
| | - Zhipeng Zeng
- Clinical Medical Research CenterGuangdong Provincial Engineering Research Center of Autoimmune Disease Precision MedicineShenzhen Engineering Research Center of Autoimmune DiseaseThe Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and TechnologyShenzhen People's HospitalShenzhenChina
| | - Wenlong Hu
- Clinical Medical Research CenterGuangdong Provincial Engineering Research Center of Autoimmune Disease Precision MedicineShenzhen Engineering Research Center of Autoimmune DiseaseThe Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and TechnologyShenzhen People's HospitalShenzhenChina
| | - Wanxia Cai
- Clinical Medical Research CenterGuangdong Provincial Engineering Research Center of Autoimmune Disease Precision MedicineShenzhen Engineering Research Center of Autoimmune DiseaseThe Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and TechnologyShenzhen People's HospitalShenzhenChina
| | - Qiang Yan
- Guangxi Key Laboratory of Metabolic Disease ResearchNephrology Department924st HospitalGuilinChina
- College of Life ScienceGuangxi Normal UniversityGuilinChina
| | - Yong Xu
- Clinical Medical Research CenterGuangdong Provincial Engineering Research Center of Autoimmune Disease Precision MedicineShenzhen Engineering Research Center of Autoimmune DiseaseThe Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and TechnologyShenzhen People's HospitalShenzhenChina
| | - Donge Tang
- Clinical Medical Research CenterGuangdong Provincial Engineering Research Center of Autoimmune Disease Precision MedicineShenzhen Engineering Research Center of Autoimmune DiseaseThe Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and TechnologyShenzhen People's HospitalShenzhenChina
| | - Yong Dai
- Clinical Medical Research CenterGuangdong Provincial Engineering Research Center of Autoimmune Disease Precision MedicineShenzhen Engineering Research Center of Autoimmune DiseaseThe Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and TechnologyShenzhen People's HospitalShenzhenChina
- Guangxi Key Laboratory of Metabolic Disease ResearchNephrology Department924st HospitalGuilinChina
| |
Collapse
|
27
|
Chen S, Li Z, He Y, Chen Q. Dysregulation of complement system in HELLP syndrome. Hypertens Pregnancy 2021; 40:303-311. [PMID: 34697959 DOI: 10.1080/10641955.2021.1983593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE To investigate the expression of complement system's activation factors in patients with HELLP syndrome. METHODS A case-control study was performed. Sixteen HELLP syndrome patients, 32 severe preeclampsia patients, and 48 normal pregnancy women were involved in this studyELISA was used to test C1q, C4d, MBL, Bb, C3a, C5a, sC5b-9, s-Endoglin, and sflt-1 in the plasma. RESULTS The levels of C5a (P < 0.01) and sC5b-9 (P = 0.014) in HELLP syndrome were higher than those in severe preeclampsia patients. CONCLUSIONS The abnormal activation of the complement system is more significant in the pathogenesis of HELLP syndrome than in severe preeclampsia.
Collapse
Affiliation(s)
- Shi Chen
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, P.R. China
| | - Zheng Li
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, P.R. China
| | - Yingdong He
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, P.R. China
| | - Qian Chen
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, P.R. China
| |
Collapse
|
28
|
Gurung S, Greening DW, Rai A, Poh QH, Evans J, Salamonsen LA. The proteomes of endometrial stromal cell-derived extracellular vesicles following a decidualizing stimulus define the cells' potential for decidualization success. Mol Hum Reprod 2021; 27:6370708. [PMID: 34524461 DOI: 10.1093/molehr/gaab057] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 08/26/2021] [Indexed: 12/14/2022] Open
Abstract
Adequate endometrial stromal cell (ESC) decidualization is vital for endometrial health. Given the importance of extracellular vesicles (EVs) in intercellular communication, we investigated how their protein landscape is reprogrammed and dysregulated during decidual response. Small EVs (sEVs) from human ESC-conditioned media at Day-2 and -14 following decidual stimuli were grouped as well- (WD) or poorly decidualized (PD) based on their prolactin secretion and subjected to mass spectrometry-based quantitative proteomics. On Day 2, in PD- versus WD-ESC-sEVs, 17 sEV- proteins were down-regulated (C5, C6; complement/coagulation cascades, and SERPING1, HRG; platelet degranulation and fibrinolysis) and 39 up-regulated (FLNA, COL1A1; focal adhesion, ENO1, PKM; glycolysis/gluconeogenesis, and RAP1B, MSN; leukocyte transendothelial migration). On Day 14, in PD- versus WD-ESC-sEVs, FLNA was down-regulated while 21 proteins were up-regulated involved in complement/coagulation cascades (C3, C6), platelet degranulation (SERPINA4, ITIH4), B-cell receptor signalling and innate immune response (immunoglobulins). Changes from Days 2 to 14 suggested a subsequent response in PD-ESC-sEVs with 89 differentially expressed proteins mostly involved in complement and coagulation cascades (C3, C6, C5), but no change in WD-ESC-sEVs ESC. Poor decidualization was also associated with loss of crucial sEV-proteins for cell adhesion and invasion (ITGA5, PFN1), glycolysis (ALDOA, PGK1) and cytoskeletal reorganization (VCL, RAC1). Overall, this study indicates varied ESC response even prior to decidualization and provides insight into sEVs-proteomes as a benchmark of well-decidualized ESC. It shows distinct variation in sEV-protein composition depending on the ESC decidual response that is critical for embryo implantation, enabling and limiting trophoblast invasion during placentation and sensing a healthy embryo.
Collapse
Affiliation(s)
- Shanti Gurung
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Obstetrics and Gynaecology, Monash Health, Monash University, Victoria, Australia
| | - David W Greening
- Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Australia.,Central Clinical School, Faulty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia.,Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Alin Rai
- Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Central Clinical School, Faulty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia.,Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Qi Hui Poh
- Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Australia
| | - Jemma Evans
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Lois A Salamonsen
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Molecular and Translational Medicine, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| |
Collapse
|
29
|
Morales-Prieto DM, Fuentes-Zacarías P, Murrieta-Coxca JM, Gutierrez-Samudio RN, Favaro RR, Fitzgerald JS, Markert UR. Smoking for two- effects of tobacco consumption on placenta. Mol Aspects Med 2021; 87:101023. [PMID: 34521556 DOI: 10.1016/j.mam.2021.101023] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 08/22/2021] [Accepted: 09/07/2021] [Indexed: 12/14/2022]
Abstract
Tobacco smoking is an important public health issue recognized by the world health organization as one of the most serious, preventable risk factors for developing a series of pregnancy pathologies. Maternal smoking is positively associated with intrauterine growth restriction (IUGR) and gestational diabetes (GDM), but negatively associated with preeclampsia (PE). In this review, we examine epidemiological, clinical and laboratory studies of smoking effects on immunoregulation during pregnancy, trophoblast function, and placental vasculature development and metabolism. We aim to identify effects of tobacco smoke components on specific placental compartments or cells, which may contribute to the understanding of the influences of maternal smoking on placenta function in normal and pathological pregnancies. Data corroborates that in any trimester, smoking is unsafe for pregnancy and that its detrimental effects outweigh questionable benefits. The effects of maternal smoking on the maternal immune regulation throughout pregnancy and the impact of different tobacco products on fetal growth have not yet been fully understood. Smoking cessation rather than treatment with replacement therapies is recommended for future mothers because also single components of tobacco and its smoke may have detrimental effects on placental function.
Collapse
Affiliation(s)
| | | | | | | | - Rodolfo R Favaro
- Placenta Lab, Department of Obstetrics, University Hospital Jena, Jena, Germany
| | - Justine S Fitzgerald
- Placenta Lab, Department of Obstetrics, University Hospital Jena, Jena, Germany; Zentrum für ambulante Medizin, University Hospital Jena, Jena, Germany
| | - Udo R Markert
- Placenta Lab, Department of Obstetrics, University Hospital Jena, Jena, Germany.
| |
Collapse
|
30
|
Low Molecular Weight Heparins (LMWH) and Implications along Pregnancy: a Focus on the Placenta. Reprod Sci 2021; 29:1414-1423. [PMID: 34231172 DOI: 10.1007/s43032-021-00678-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 06/18/2021] [Indexed: 10/20/2022]
Abstract
Low molecular weight heparins (LMWH) have been largely studied for their use during pregnancy. The biology and the pharmacology of these molecules are well known and may be summarized in three main mechanisms of action: anti-coagulant, anti-inflammatory, and immunomodulant. The clinical implications of these drugs during pregnancy are mainly related to their action on the placenta, because of the presence of specific molecular and cellular targets, particularly at the trophoblast-endometrial interface. As well as for the prevention and treatment of thromboembolism, LMWH have been largely investigated for the improvement of embryo implantation and for the prevention of placenta-related complications such as preeclampsia, fetal growth restriction, and intrauterine fetal death. However, data on this topic are still unclear. The present review discusses the biological features, the mechanisms of action, and the possible contribution of LMWH to the success of placentation along pregnancy, pointing out the need for future basic science and clinical researches in this important field with the final aim to improve clinical practice in high-risk pregnancies.
Collapse
|
31
|
Waker CA, Kaufman MR, Brown TL. Current State of Preeclampsia Mouse Models: Approaches, Relevance, and Standardization. Front Physiol 2021; 12:681632. [PMID: 34276401 PMCID: PMC8284253 DOI: 10.3389/fphys.2021.681632] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/24/2021] [Indexed: 12/14/2022] Open
Abstract
Preeclampsia (PE) is a multisystemic, pregnancy-specific disorder and a leading cause of maternal and fetal death. PE is also associated with an increased risk for chronic morbidities later in life for mother and offspring. Abnormal placentation or placental function has been well-established as central to the genesis of PE; yet much remains to be determined about the factors involved in the development of this condition. Despite decades of investigation and many clinical trials, the only definitive treatment is parturition. To better understand the condition and identify potential targets preclinically, many approaches to simulate PE in mice have been developed and include mixed mouse strain crosses, genetic overexpression and knockout, exogenous agent administration, surgical manipulation, systemic adenoviral infection, and trophoblast-specific gene transfer. These models have been useful to investigate how biological perturbations identified in human PE are involved in the generation of PE-like symptoms and have improved the understanding of the molecular mechanisms underpinning the human condition. However, these approaches were characterized by a wide variety of physiological endpoints, which can make it difficult to compare effects across models and many of these approaches have aspects that lack physiological relevance to this human disorder and may interfere with therapeutic development. This report provides a comprehensive review of mouse models that exhibit PE-like symptoms and a proposed standardization of physiological characteristics for analysis in murine models of PE.
Collapse
Affiliation(s)
- Christopher A Waker
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Melissa R Kaufman
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Thomas L Brown
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| |
Collapse
|
32
|
Alrahmani L, Gonzalez Suarez ML, Cousin MA, Moyer AM, Willrich MAV, White WM, Wick MJ, Tostrud LJ, Narang K, Garovic VD. Quantitative Alterations in Complement Alternative Pathway and Related Genetic Analysis in Severe Phenotype Preeclampsia. KIDNEY360 2021; 2:1463-1472. [PMID: 35373096 PMCID: PMC8786143 DOI: 10.34067/kid.0000992021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 06/29/2021] [Indexed: 02/04/2023]
Abstract
Background Preeclampsia and hemolysis, elevated liver enzymes, and low platelets (HELLP) syndrome share many clinical and biologic features with thrombotic microangiopathy syndromes caused by complement abnormalities. Our hypothesis was that similar functional and genetic alterations in the complement alternative pathway (CAP) are present in these disorders of pregnancy. Methods We conducted quantitative analysis of proteins involved in CAP using ELISA and nephelometry on prospectively collected blood samples from patients with severe phenotype preeclampsia (defined as delivery ≤34 weeks due to preeclampsia), HELLP syndrome, or eclampsia, and matched normotensive controls (n=25 in each arm) between 2011 and 2016. Sequencing was performed to interrogate 14 genes encoding CAP components. Results Both groups were similar in age, gravidity, parity, marital status, and race. The study group had a higher BMI (mean±SD, 32±8 versus 25±4 kg/m2; P=0.002) and earlier gestational age at delivery (32.5±3.6 versus 40.3±1 weeks; P<0.001). Serologic studies demonstrated elevated Bb subunit (median [range], 1.2 [0.5-4.3] versus 0.6 [0.5-1] μg/ml; P<0.001), complement C5 concentration (28 [18-33] versus 24 [15-34] mg/dl; P=0.03), and sMAC (371 [167-761] versus 184 [112-249] ng/ml; P<0.001) concentrations in patients with preeclampsia. Two thirds of patients with preeclampsia had at least one nonsynonymous sequence variant in CAP genes. Conclusion Patients with severe phenotype preeclampsia manifest functional alterations in CAP activation. Genetic variants in the CAP genes were detected in several patients, but a larger population study is necessary to fully evaluate genetic risk. Genetic screening and complement-targeted treatment may be useful in risk stratification and novel therapeutic approaches.
Collapse
Affiliation(s)
- Layan Alrahmani
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota,Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Loyola University Medical Center, Chicago, Illinois
| | | | - Margot A. Cousin
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota,Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota
| | - Ann M. Moyer
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | | | - Wendy M. White
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota
| | - Myra J. Wick
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota,Department of Medical Genetics, Mayo Clinic, Rochester, Minnesota
| | - Linda J. Tostrud
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Kavita Narang
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota
| | - Vesna D. Garovic
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota,Division of Nephrology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
33
|
Placental Complement Activation in Fetal and Neonatal Alloimmune Thrombocytopenia: An Observational Study. Int J Mol Sci 2021; 22:ijms22136763. [PMID: 34201864 PMCID: PMC8267834 DOI: 10.3390/ijms22136763] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 11/16/2022] Open
Abstract
Fetal and neonatal alloimmune thrombocytopenia (FNAIT) is a disease that causes thrombocytopenia and a risk of bleeding in the (unborn) child that result from maternal alloantibodies directed against fetal, paternally inherited, human platelet antigens (HPA). It is hypothesized that these alloantibodies can also bind to the placenta, causing placental damage. This study aims to explore signs of antibody-mediated placental damage in FNAIT. We performed a retrospective study that included pregnant women, their newborns, and placentas. It comprised 23 FNAIT cases, of which nine were newly diagnosed (14 samples) and 14 were antenatally treated with intravenous immune globulins (IVIg) (21 samples), and 20 controls, of which 10 had anti-HLA-class I antibodies. Clinical information was collected from medical records. Placental samples were stained for complement activation markers (C1q, C4d, SC5b-9, and mannose-binding lectin) using immunohistochemistry. Histopathology was examined according to the Amsterdam criteria. A higher degree of C4d deposition was present in the newly diagnosed FNAIT cases (10/14 samples), as compared to the IVIg-treated FNAIT cases (2/21 samples, p = 0.002) and anti-HLA-negative controls (3/20 samples, p = 0.006). A histopathological examination showed delayed maturation in four (44%) placentas in the newly diagnosed FNAIT cases, five (36%) in the IVIg-treated FNAIT cases, and one in the controls (NS). C4d deposition at the syncytiotrophoblast was present in combination with low-grade villitis of unknown etiology in three newly diagnosed FNAIT cases that were born SGA. We conclude that a higher degree of classical pathway-induced complement activation is present in placentas from pregnancies with untreated FNAIT. This may affect placental function and fetal growth.
Collapse
|
34
|
Situmorang PC, Ilyas S, Hutahaean S, Rosidah R. Histological changes in placental rat apoptosis via FasL and cytochrome c by the nano-herbal Zanthoxylum acanthopodium. Saudi J Biol Sci 2021; 28:3060-3068. [PMID: 34025182 PMCID: PMC8117027 DOI: 10.1016/j.sjbs.2021.02.047] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/15/2020] [Accepted: 02/10/2021] [Indexed: 12/03/2022] Open
Abstract
The administration of nanoherbal andaliman reduced apoptosis via cytochrome c and FasL. EVOO reduces apoptosis via cytochrome c and FasL better than andaliman. Combination Nano herbal andaliman and EVOO reduce reduced apoptosis via cytochrome c and FasL on placental histology of hypertension rats. Combination Nano herbal andaliman and EVOO reduce MDA levels in hypertension rats. Nano herbal andaliman and combined with EVOO increase HSP-70 expression in hypertension rats.
Zanthoxylum acanthopodium has several biological activities, such as antimicrobial, anti-inflammatory, and antioxidant properties. This strong antioxidant herb can be used as a drug for hypertension. FasL and cytochrome c expression play roles in the apoptotic pathway in the placenta. This study focused on the histological change in apoptosis via cytochrome c and Fas ligand expression by investigating whether Zanthoxylum acanthopodium (ZA) fruits affect apoptosis. The present study consisted of five treatments: Normal pregnant rats (C), Hypertension rats (C + ), hypertension rats + extra virgin olive oil (EVOO) (T1), Hypertension rats + ZA (T2), and hypertension rats + EVOO + ZA (T3). Hypertension was induced in rats by injecting 3 mlml of 6% NaCl. Nanoherbal of ZA (100 mg/kg BW) and EVOO (1 ml) were given on the 13th–19th days of pregnancy. Pregnant rats were dissected on the 20th day of pregnancy by cervical dislocation. ELISA assays were performed for the analysis of HSP-70 expression. Immunohistochemistry and TUNEL assays were used to analyse the histological changes in placental tissue. The results showed that cytochrome c and FasL protein exposure levels in the labyrinth, basal, and yolk sac zones were increased during hypertensive pregnancy (P < 0.0001) in rats. The administration of nanoherbal of ZA decreased the expression of cytochrome c and FasL. A significant difference was found in the combination of nanoherbal of ZA and EVOO.
Collapse
Affiliation(s)
- Putri Cahaya Situmorang
- Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Sumatera Utara, Medan, Indonesia
| | - Syafruddin Ilyas
- Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Sumatera Utara, Medan, Indonesia
| | - Salomo Hutahaean
- Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Sumatera Utara, Medan, Indonesia
| | - Rosidah Rosidah
- Faculty of Pharmacy, Department of Pharmacology, Universitas Sumatera Utara, Medan, Indonesia
| |
Collapse
|
35
|
Kedziora SM, Kräker K, Markó L, Binder J, Sugulle M, Gauster M, Müller DN, Dechend R, Haase N, Herse F. Kidney Injury Caused by Preeclamptic Pregnancy Recovers Postpartum in a Transgenic Rat Model. Int J Mol Sci 2021; 22:ijms22073762. [PMID: 33916404 PMCID: PMC8038582 DOI: 10.3390/ijms22073762] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/26/2021] [Accepted: 03/31/2021] [Indexed: 11/21/2022] Open
Abstract
Preeclampsia (PE) is characterized by the onset of hypertension (≥140/90 mmHg) and presence of proteinuria (>300 mg/L/24 h urine) or other maternal organ dysfunctions. During human PE, renal injuries have been observed. Some studies suggest that women with PE diagnosis have an increased risk to develop renal diseases later in life. However, in human studies PE as a single cause of this development cannot be investigated. Here, we aimed to investigate the effect of PE on postpartum renal damage in an established transgenic PE rat model. Female rats harboring the human-angiotensinogen gene develop a preeclamptic phenotype after mating with male rats harboring the human-renin gene, but are normotensive before and after pregnancy. During pregnancy PE rats developed mild tubular and glomerular changes assessed by histologic analysis, increased gene expression of renal damage markers such as kidney injury marker 1 and connective-tissue growth factor, and albuminuria compared to female wild-type rats (WT). However, four weeks postpartum, most PE-related renal pathologies were absent, including albuminuria and elevated biomarker expression. Only mild enlargement of the glomerular tuft could be detected. Overall, the glomerular and tubular function were affected during pregnancy in the transgenic PE rat. However, almost all these pathologies observed during PE recovered postpartum.
Collapse
Affiliation(s)
- Sarah M. Kedziora
- Experimental and Clinical Research Center (ECRC), A Joint Cooperation of Charité—Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, 13125 Berlin, Germany; (S.M.K.); (K.K.); (L.M.); (D.N.M.); (R.D.); (N.H.)
- Max Delbruck Center for Molecular Medicine (MDC) in the Helmholtz Association, 13125 Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, 10117 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 13353 Berlin, Germany
| | - Kristin Kräker
- Experimental and Clinical Research Center (ECRC), A Joint Cooperation of Charité—Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, 13125 Berlin, Germany; (S.M.K.); (K.K.); (L.M.); (D.N.M.); (R.D.); (N.H.)
- Max Delbruck Center for Molecular Medicine (MDC) in the Helmholtz Association, 13125 Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, 10117 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 13353 Berlin, Germany
| | - Lajos Markó
- Experimental and Clinical Research Center (ECRC), A Joint Cooperation of Charité—Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, 13125 Berlin, Germany; (S.M.K.); (K.K.); (L.M.); (D.N.M.); (R.D.); (N.H.)
- Max Delbruck Center for Molecular Medicine (MDC) in the Helmholtz Association, 13125 Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, 10117 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 13353 Berlin, Germany
| | - Julia Binder
- Department of Obstetrics and Gynecology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Meryam Sugulle
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, 0318 Oslo, Norway;
- Division of Obstetrics and Gynaecology, Oslo University Hospital, 0450 Oslo, Norway
| | - Martin Gauster
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Centre, Medical University of Graz, 8036 Graz, Austria;
| | - Dominik N. Müller
- Experimental and Clinical Research Center (ECRC), A Joint Cooperation of Charité—Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, 13125 Berlin, Germany; (S.M.K.); (K.K.); (L.M.); (D.N.M.); (R.D.); (N.H.)
- Max Delbruck Center for Molecular Medicine (MDC) in the Helmholtz Association, 13125 Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, 10117 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 13353 Berlin, Germany
| | - Ralf Dechend
- Experimental and Clinical Research Center (ECRC), A Joint Cooperation of Charité—Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, 13125 Berlin, Germany; (S.M.K.); (K.K.); (L.M.); (D.N.M.); (R.D.); (N.H.)
- Max Delbruck Center for Molecular Medicine (MDC) in the Helmholtz Association, 13125 Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, 10117 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 13353 Berlin, Germany
- Helios Klinikum, 13125 Berlin, Germany
| | - Nadine Haase
- Experimental and Clinical Research Center (ECRC), A Joint Cooperation of Charité—Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, 13125 Berlin, Germany; (S.M.K.); (K.K.); (L.M.); (D.N.M.); (R.D.); (N.H.)
- Max Delbruck Center for Molecular Medicine (MDC) in the Helmholtz Association, 13125 Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, 10117 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, 13353 Berlin, Germany
| | - Florian Herse
- Experimental and Clinical Research Center (ECRC), A Joint Cooperation of Charité—Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, 13125 Berlin, Germany; (S.M.K.); (K.K.); (L.M.); (D.N.M.); (R.D.); (N.H.)
- Max Delbruck Center for Molecular Medicine (MDC) in the Helmholtz Association, 13125 Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, 10117 Berlin, Germany
- Correspondence:
| |
Collapse
|
36
|
Feng X, Liu Y, Zhang Y, Zhang Y, Li H, Zheng Q, Li N, Tang J, Xu Z. New views on endothelial dysfunction in gestational hypertension and potential therapy targets. Drug Discov Today 2021; 26:1420-1436. [PMID: 33677145 DOI: 10.1016/j.drudis.2021.03.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/10/2020] [Accepted: 03/01/2021] [Indexed: 12/14/2022]
Abstract
The placenta has vital roles in metabolite exchange, fetal growth, and pre-eclampsia (PE). In this review, we discuss the pathogenesis of hypertension in pregnancy, focusing on four major theories to explain PE, discussing endothelial roles in those theories. We focus in particular on the roles of nitric oxide (NO) and prostacyclin (PGI2) in placental endothelium, and propose new hypotheses for the influence and mechanisms of endothelial NO and PGI2 signaling pathways in PE.
Collapse
Affiliation(s)
- Xueqin Feng
- First Hospital of Soochow University & Maternal and Child Health Care Hospital of Wuxi, Jiangsu, China; Department of Obstetrics, Affiliated Hospital of Jining Medical University, Shandong, China
| | - Yanping Liu
- First Hospital of Soochow University & Maternal and Child Health Care Hospital of Wuxi, Jiangsu, China
| | - Yingying Zhang
- First Hospital of Soochow University & Maternal and Child Health Care Hospital of Wuxi, Jiangsu, China
| | - Yumeng Zhang
- First Hospital of Soochow University & Maternal and Child Health Care Hospital of Wuxi, Jiangsu, China
| | - Huan Li
- First Hospital of Soochow University & Maternal and Child Health Care Hospital of Wuxi, Jiangsu, China
| | - Qiutong Zheng
- First Hospital of Soochow University & Maternal and Child Health Care Hospital of Wuxi, Jiangsu, China
| | - Na Li
- First Hospital of Soochow University & Maternal and Child Health Care Hospital of Wuxi, Jiangsu, China
| | - Jiaqi Tang
- First Hospital of Soochow University & Maternal and Child Health Care Hospital of Wuxi, Jiangsu, China.
| | - Zhice Xu
- First Hospital of Soochow University & Maternal and Child Health Care Hospital of Wuxi, Jiangsu, China.
| |
Collapse
|
37
|
Jaworska J, Ropka-Molik K, Piórkowska K, Szmatoła T, Kowalczyk-Zięba I, Wocławek-Potocka I, Siemieniuch M. Transcriptome Profiling of the Retained Fetal Membranes-An Insight in the Possible Pathogenesis of the Disease. Animals (Basel) 2021; 11:ani11030675. [PMID: 33802481 PMCID: PMC8000898 DOI: 10.3390/ani11030675] [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: 02/03/2021] [Revised: 02/25/2021] [Accepted: 02/27/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Retained fetal membranes (RFM) in mares is a disease of a multifactorial etiology with not fully understood pathogenesis. Profound analysis of genes expressed in the placenta may reveal pathways and processes which might be comprised in mares with this disease and hence help to explain the pathogenesis of RFM. This work employed RNA sequencing to identify and compare genes differentially expressed (DEGs) in the placenta of mares that retained fetal membranes and those that released them physiologically. Results showed that within DEGs genes important for apoptosis, inflammatory-related processes, and metabolism of extracellular matrix were identified. Abstract Retained fetal membranes (RFM) is one of the most common post-partum diseases of a complex etiology. Moreover, its pathogenesis is still not elucidated. Detailed transcriptomic analysis of physiological and retained placenta may bring profound insight in the pathogenesis of the disease. The aim of the study was to compare the transcriptome of the retained and physiologically released placenta as well as biological pathways and processes in order to determine the possible pathogenesis of the disease. Samples of the endometrium and the allantochorion were taken within 2 h after parturition from control mares (n = 3) and mares with RFM (n = 3). RNA sequencing was performed with the use of all samples and mRNA expression of chosen genes was validated with Real Time PCR. Analysis of RNA-seq identified 487 differentially expressed genes in the allantochorion and 261 in the endometrium of control and RFM mares (p < 0.0001). Within genes that may be important in the release of fetal membranes and were differentially expressed, our report pinpointed BGN, TIMP1, DRB, CD3E, C3, FCN3, CASP3, BCL2L1. Gene ontology analysis showed possible processes which were altered in RFM that are apoptosis, inflammatory-related processes, and extracellular matrix metabolism and might be involved in the pathogenesis of RFM. This is the first report on the transcriptome of RFM and physiologically released placenta in mares.
Collapse
Affiliation(s)
- Joanna Jaworska
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-747 Olsztyn, Poland; (I.K.-Z.); (I.W.-P.)
- Correspondence:
| | - Katarzyna Ropka-Molik
- Department of Animal Molecular Biology, National Research Institute of Animal Production, 32-083 Balice, Poland; (K.R.-M.); (K.P.); (T.S.)
| | - Katarzyna Piórkowska
- Department of Animal Molecular Biology, National Research Institute of Animal Production, 32-083 Balice, Poland; (K.R.-M.); (K.P.); (T.S.)
| | - Tomasz Szmatoła
- Department of Animal Molecular Biology, National Research Institute of Animal Production, 32-083 Balice, Poland; (K.R.-M.); (K.P.); (T.S.)
- University Centre of Veterinary Medicine Krakow, University of Agriculture in Krakow, Al. Mickiewicza 24/28, 30-059 Krakow, Poland
| | - Ilona Kowalczyk-Zięba
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-747 Olsztyn, Poland; (I.K.-Z.); (I.W.-P.)
| | - Izabela Wocławek-Potocka
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, 10-747 Olsztyn, Poland; (I.K.-Z.); (I.W.-P.)
| | - Marta Siemieniuch
- Research Station of the Institute of Reproduction and Food Research, Polish Academy of Sciences in Popielno, 12-220 Ruciane-Nida, Poland;
| |
Collapse
|
38
|
Koopman JJE, van Essen MF, Rennke HG, de Vries APJ, van Kooten C. Deposition of the Membrane Attack Complex in Healthy and Diseased Human Kidneys. Front Immunol 2021; 11:599974. [PMID: 33643288 PMCID: PMC7906018 DOI: 10.3389/fimmu.2020.599974] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 12/21/2020] [Indexed: 12/11/2022] Open
Abstract
The membrane attack complex-also known as C5b-9-is the end-product of the classical, lectin, and alternative complement pathways. It is thought to play an important role in the pathogenesis of various kidney diseases by causing cellular injury and tissue inflammation, resulting in sclerosis and fibrosis. These deleterious effects are, consequently, targeted in the development of novel therapies that inhibit the formation of C5b-9, such as eculizumab. To clarify how C5b-9 contributes to kidney disease and to predict which patients benefit from such therapy, knowledge on deposition of C5b-9 in the kidney is essential. Because immunohistochemical staining of C5b-9 has not been routinely conducted and never been compared across studies, we provide a review of studies on deposition of C5b-9 in healthy and diseased human kidneys. We describe techniques to stain deposits and compare the occurrence of deposits in healthy kidneys and in a wide spectrum of kidney diseases, including hypertensive nephropathy, diabetic nephropathy, membranous nephropathy, IgA nephropathy, lupus nephritis, C3 glomerulopathy, and thrombotic microangiopathies such as the atypical hemolytic uremic syndrome, vasculitis, interstitial nephritis, acute tubular necrosis, kidney tumors, and rejection of kidney transplants. We summarize how these deposits are related with other histological lesions and clinical characteristics. We evaluate the prognostic relevance of these deposits in the light of possible treatment with complement inhibitors.
Collapse
Affiliation(s)
- Jacob J E Koopman
- Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
- Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Mieke F van Essen
- Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Helmut G Rennke
- Division of Renal Pathology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Aiko P J de Vries
- Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Cees van Kooten
- Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
39
|
Collier ARY, Smith LA, Karumanchi SA. Review of the immune mechanisms of preeclampsia and the potential of immune modulating therapy. Hum Immunol 2021; 82:362-370. [PMID: 33551128 DOI: 10.1016/j.humimm.2021.01.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/23/2020] [Accepted: 01/05/2021] [Indexed: 12/14/2022]
Abstract
Successful pregnancy relies on maternal immunologic tolerance mechanisms limit maladaptive immune responses against the semi-allogeneic fetus and placenta and support fetal growth. Preeclampsia is a common disorder of pregnancy that affects 4-10% of pregnancies and is a leading cause of maternal and neonatal morbidity and mortality. Preeclampsia clinically manifests as maternal hypertension, proteinuria, and progressive multi-organ injury likely triggered by hypoxic injury to the placenta, resulting in local and systemic anti-angiogenic and inflammatory factor production. Despite the steady rising rates of preeclampsia in the United States, effective treatment options are limited to delivery, which improves maternal status often at the cost of prematurity in the newborn. Preeclampsia also increases the lifelong risk of cardiovascular disease for both mother and infant. Thus, identifying new therapeutic targets is a high priority area to improve maternal, fetal, and infant health outcomes. Immune abnormalities in the placenta and in the maternal circulation have been reported to precede the clinical onset of disease. In particular, excessive systemic and placental complement activation and impaired adaptive T cell tolerance with Th1/Th2/Th17/Treg imbalance has been reported in humans and in animal models of preeclampsia. In this review, we focus on the evidence for the immune origins of preeclampsia, discuss the promise of immune modulating therapy for prevention or treatment, and highlight key areas for future research.
Collapse
Affiliation(s)
- Ai-Ris Y Collier
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA, USA; Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| | - Laura A Smith
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Obstetrics, Gynecology, and Reproductive Biology, Harvard Medical School, Boston, MA, USA
| | - S Ananth Karumanchi
- Department of Obstetrics and Gynecology, Beth Israel Deaconess Medical Center, Boston, MA, USA; Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Boston, MA, USA; Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
40
|
Navajas R, Corrales F, Paradela A. Quantitative proteomics-based analyses performed on pre-eclampsia samples in the 2004-2020 period: a systematic review. Clin Proteomics 2021; 18:6. [PMID: 33499801 PMCID: PMC7836571 DOI: 10.1186/s12014-021-09313-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 01/15/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Quantitative proteomics is an invaluable tool in biomedicine for the massive comparative analysis of protein component of complex biological samples. In the last two decades, this technique has been used to describe proteins potentially involved in the pathophysiological mechanisms of preeclampsia as well as to identify protein biomarkers that could be used with diagnostic/prognostic purposes in pre-eclampsia. RESULTS We have done a systematic review of all proteomics-based papers describing differentially expressed proteins in this disease. Searching Pubmed with the terms pre-eclampsia and proteomics, restricted to the Title/Abstract and to MeSH fields, and following manual curation of the original list, retrieved 69 original articles corresponding to the 2004-2020 period. We have only considered those results based on quantitative, unbiased proteomics studies conducted in a controlled manner on a cohort of control and pre-eclamptic individuals. The sources of biological material used were serum/plasma (n = 32), placenta (n = 23), urine (n = 9), cerebrospinal fluid (n = 2), amniotic fluid (n = 2) and decidual tissue (n = 1). Overall results were filtered based on two complementary criteria. First, we have only accounted all those proteins described in at least two (urine), three (placenta) and four (serum/plasma) independent studies. Secondly, we considered the consistency of the quantitative data, that is, inter-study agreement in the protein abundance control/pre-eclamptic ratio. The total number of differential proteins in serum/plasma (n = 559), placenta (n = 912), urine (n = 132) and other sources of biological material (n = 26), reached 1631 proteins. Data were highly complementary among studies, resulting from differences on biological sources, sampling strategies, patient stratification, quantitative proteomic analysis methods and statistical data analysis. Therefore, stringent filtering was applied to end up with a cluster of 18, 29 and 16 proteins consistently regulated in pre-eclampsia in placenta, serum/plasma and urine, respectively. The systematic collection, standardization and evaluation of the results, using diverse filtering criteria, provided a panel of 63 proteins whose levels are consistently modified in the context of pre-eclampsia.
Collapse
Affiliation(s)
- Rosana Navajas
- Functional Proteomics Facility, Centro Nacional de Biotecnología (CNB-CSIC), ProteoRed-ISCIII, Madrid, Spain
| | - Fernando Corrales
- Functional Proteomics Facility, Centro Nacional de Biotecnología (CNB-CSIC), ProteoRed-ISCIII, Madrid, Spain
| | - Alberto Paradela
- Functional Proteomics Facility, Centro Nacional de Biotecnología (CNB-CSIC), ProteoRed-ISCIII, Madrid, Spain.
| |
Collapse
|
41
|
Yu H, Yin Y, Zhang J, Zhou R. The impact of particulate matter 2.5 on the risk of preeclampsia: an updated systematic review and meta-analysis. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:37527-37539. [PMID: 32740838 PMCID: PMC7496023 DOI: 10.1007/s11356-020-10112-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 07/10/2020] [Indexed: 05/07/2023]
Abstract
There is increasing and inconsistent evidence of a linkage between maternal exposure to particulate matter 2.5 (PM2.5) and preeclampsia. Therefore, this study was conducted to investigate this relationship. Electronic databases including PubMed, Embase, Web of Science, and Cochrane Library were searched to identify articles published from inception to March 23, 2020, which showed a correlation between PM2.5 and preeclampsia. Finally, 9 of 523 initial studies were deemed eligible for inclusion. A random effect model was adopted to calculate the standardized odds ratio (OR) and 95% confidence interval (CI). Based on potential effect modification, subgroup analyses were further performed. Meta-analysis showed that maternal exposure to PM2.5 (per 10 μg/m3 increment) elevated the risk of preeclampsia (OR = 1.32, 95% CI 1.10 to 1.58%). Compared with other pregnancy trimesters, the third trimester of pregnancy seems to be the period in which women are more susceptible to PM2.5. Significant effect modification of the correlation between PM2.5 exposure and preeclampsia according to multiple pregnancies, pregnancy stage, maternal-related disease history, and sample size was not observed. The results demonstrated that maternal exposure to PM2.5 may predispose pregnant women to develop preeclampsia, especially in the third trimester of pregnancy. Therefore, more efforts should be made to improve air quality to maintain the health of pregnant women.
Collapse
Affiliation(s)
- Hongbiao Yu
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Yangxue Yin
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Jiashuo Zhang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Rong Zhou
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University) of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
42
|
Cornish EF, Filipovic I, Åsenius F, Williams DJ, McDonnell T. Innate Immune Responses to Acute Viral Infection During Pregnancy. Front Immunol 2020; 11:572567. [PMID: 33101294 PMCID: PMC7556209 DOI: 10.3389/fimmu.2020.572567] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 08/31/2020] [Indexed: 02/06/2023] Open
Abstract
Immunological adaptations in pregnancy allow maternal tolerance of the semi-allogeneic fetus but also increase maternal susceptibility to infection. At implantation, the endometrial stroma, glands, arteries and immune cells undergo anatomical and functional transformation to create the decidua, the specialized secretory endometrium of pregnancy. The maternal decidua and the invading fetal trophoblast constitute a dynamic junction that facilitates a complex immunological dialogue between the two. The decidual and peripheral immune systems together assume a pivotal role in regulating the critical balance between tolerance and defense against infection. Throughout pregnancy, this equilibrium is repeatedly subjected to microbial challenge. Acute viral infection in pregnancy is associated with a wide spectrum of adverse consequences for both mother and fetus. Vertical transmission from mother to fetus can cause developmental anomalies, growth restriction, preterm birth and stillbirth, while the mother is predisposed to heightened morbidity and maternal death. A rapid, effective response to invasive pathogens is therefore essential in order to avoid overwhelming maternal infection and consequent fetal compromise. This sentinel response is mediated by the innate immune system: a heritable, highly evolutionarily conserved system comprising physical barriers, antimicrobial peptides (AMP) and a variety of immune cells—principally neutrophils, macrophages, dendritic cells, and natural killer cells—which express pattern-receptors that detect invariant molecular signatures unique to pathogenic micro-organisms. Recognition of these signatures during acute infection triggers signaling cascades that enhance antimicrobial properties such as phagocytosis, secretion of pro-inflammatory cytokines and activation of the complement system. As well as coordinating the initial immune response, macrophages and dendritic cells present microbial antigens to lymphocytes, initiating and influencing the development of specific, long-lasting adaptive immunity. Despite extensive progress in unraveling the immunological adaptations of pregnancy, pregnant women remain particularly susceptible to certain acute viral infections and continue to experience mortality rates equivalent to those observed in pandemics several decades ago. Here, we focus specifically on the pregnancy-induced vulnerabilities in innate immunity that contribute to the disproportionately high maternal mortality observed in the following acute viral infections: Lassa fever, Ebola virus disease (EVD), dengue fever, hepatitis E, influenza, and novel coronavirus infections.
Collapse
Affiliation(s)
- Emily F Cornish
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, United Kingdom
| | - Iva Filipovic
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institute, Stockholm, Sweden
| | - Fredrika Åsenius
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, United Kingdom
| | - David J Williams
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, United Kingdom
| | - Thomas McDonnell
- Department of Biochemical Engineering, University College London, London, United Kingdom
| |
Collapse
|
43
|
Aneman I, Pienaar D, Suvakov S, Simic TP, Garovic VD, McClements L. Mechanisms of Key Innate Immune Cells in Early- and Late-Onset Preeclampsia. Front Immunol 2020; 11:1864. [PMID: 33013837 PMCID: PMC7462000 DOI: 10.3389/fimmu.2020.01864] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/10/2020] [Indexed: 12/23/2022] Open
Abstract
Preeclampsia is a complex cardiovascular disorder of pregnancy with underlying multifactorial pathogeneses; however, its etiology is not fully understood. It is characterized by the new onset of maternal hypertension after 20 weeks of gestation, accompanied by proteinuria, maternal organ damage, and/or uteroplacental dysfunction. Preeclampsia can be subdivided into early- and late-onset phenotypes (EOPE and LOPE), diagnosed before 34 weeks or from 34 weeks of gestation, respectively. Impaired placental development in early pregnancy and subsequent growth restriction is often associated with EOPE, while LOPE is associated with maternal endothelial dysfunction. The innate immune system plays an essential role in normal progression of physiological pregnancy and fetal development. However, inappropriate or excessive activation of this system can lead to placental dysfunction or poor maternal vascular adaptation and contribute to the development of preeclampsia. This review aims to comprehensively outline the mechanisms of key innate immune cells including macrophages, neutrophils, natural killer (NK) cells, and innate B1 cells, in normal physiological pregnancy, EOPE and LOPE. The roles of the complement system, syncytiotrophoblast extracellular vesicles and mesenchymal stem cells (MSCs) are also discussed in the context of innate immune system regulation and preeclampsia. The outlined molecular mechanisms, which represent potential therapeutic targets, and associated emerging treatments, are evaluated as treatments for preeclampsia. Therefore, by addressing the current understanding of innate immunity in the pathogenesis of EOPE and LOPE, this review will contribute to the body of research that could lead to the development of better diagnosis, prevention, and treatment strategies. Importantly, it will delineate the differences in the mechanisms of the innate immune system in two different types of preeclampsia, which is necessary for a more personalized approach to the monitoring and treatment of affected women.
Collapse
Affiliation(s)
- Ingrid Aneman
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
| | - Dillan Pienaar
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
| | - Sonja Suvakov
- Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Tatjana P. Simic
- Faculty of Medicine, Institute of Medical and Clinical Biochemistry, University of Belgrade, Belgrade, Serbia
- Department of Medical Sciences, Serbian Academy of Sciences and Arts, Belgrade, Serbia
| | - Vesna D. Garovic
- Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Lana McClements
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
| |
Collapse
|
44
|
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: 41] [Impact Index Per Article: 10.3] [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
|