1
|
Frimat M, Gnemmi V, Stichelbout M, Provôt F, Fakhouri F. Pregnancy as a susceptible state for thrombotic microangiopathies. Front Med (Lausanne) 2024; 11:1343060. [PMID: 38476448 PMCID: PMC10927739 DOI: 10.3389/fmed.2024.1343060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/12/2024] [Indexed: 03/14/2024] Open
Abstract
Pregnancy and the postpartum period represent phases of heightened vulnerability to thrombotic microangiopathies (TMAs), as evidenced by distinct patterns of pregnancy-specific TMAs (e.g., preeclampsia, HELLP syndrome), as well as a higher incidence of nonspecific TMAs, such as thrombotic thrombocytopenic purpura or hemolytic uremic syndrome, during pregnancy. Significant strides have been taken in understanding the underlying mechanisms of these disorders in the past 40 years. This progress has involved the identification of pivotal factors contributing to TMAs, such as the complement system, ADAMTS13, and the soluble VEGF receptor Flt1. Regardless of the specific causal factor (which is not generally unique in relation to the usual multifactorial origin of TMAs), the endothelial cell stands as a central player in the pathophysiology of TMAs. Pregnancy has a major impact on the physiology of the endothelium. Besides to the development of placenta and its vascular consequences, pregnancy modifies the characteristics of the women's microvascular endothelium and tends to render it more prone to thrombosis. This review aims to delineate the distinct features of pregnancy-related TMAs and explore the contributing mechanisms that lead to this increased susceptibility, particularly influenced by the "gravid endothelium." Furthermore, we will discuss the potential contribution of histopathological studies in facilitating the etiological diagnosis of pregnancy-related TMAs.
Collapse
Affiliation(s)
- Marie Frimat
- CHU Lille, Nephrology Department, Univ. Lille, Lille, France
- Inserm, Institut Pasteur de Lille, Univ. Lille, Lille, France
| | | | | | - François Provôt
- CHU Lille, Nephrology Department, Univ. Lille, Lille, France
| | - Fadi Fakhouri
- Service of Nephrology and Hypertension, CHUV and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
2
|
Understanding the Immune System in Fetal Protection and Maternal Infections during Pregnancy. J Immunol Res 2022; 2022:7567708. [PMID: 35785037 PMCID: PMC9249541 DOI: 10.1155/2022/7567708] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 05/20/2022] [Indexed: 11/18/2022] Open
Abstract
The fetal-maternal immune system determines the fate of pregnancy. The trophoblast cells not only give an active response against external stimuli but are also involved in secreting most of the cytokines. These cells have an essential function in fetal acceptance or fetal rejection. Other immune cells also play a pivotal role in carrying out a successful pregnancy. The disruption in this mechanism may lead to harmful effects on pregnancy. The placenta serves as an immune barrier in fetus protection against invading pathogens. Once the infections prevail, they may localize in placental and fetal tissues, and the presence of inflammation due to cytokines may have detrimental effects on pregnancy. Moreover, some pathogens are responsible for congenital fetal anomalies and affect almost all organs of the developing fetus. This review article is designed to address the bacterial and viral infections that threaten pregnancy and their possible outcomes. Moreover, training of the fetal immune system against the exposure of infections and the role of CD49a + NK cells in embryonic development will also be highlighted.
Collapse
|
3
|
Zhang L, Cao L, Feng P, Han X, Yang L. Complement regulation in ovine lymph nodes during early pregnancy. Exp Ther Med 2021; 23:166. [PMID: 35069847 PMCID: PMC8753979 DOI: 10.3892/etm.2021.11089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 12/08/2021] [Indexed: 11/05/2022] Open
Abstract
A fetus changes immune responses in the uterus and the maternal immune system, and lymph nodes are associated with regulating maternal adaptive immunity. Complement activation is associated with abnormal pregnancy in mice and humans. The aim of the present study was to explore the expression levels of complement components in maternal lymph nodes during early pregnancy in sheep. Maternal inguinal lymph nodes were sampled on day 16 of the estrous cycle, and days 13, 16 and 25 of gestation in ewes. Reverse transcription-quantitative PCR, western blotting and immunohistochemical analyses were used to detect the expression levels of complement components C1q, C1r, C1s, C2, C3, C4a, C5b and C9 in the lymph nodes. The results revealed that the protein and mRNA levels of C1q, C1s and C5b were enhanced during early pregnancy, and that C1r and C4a were upregulated at day 25 of pregnancy. The mRNA and protein levels of C2 and C9 peaked at day 16 of pregnancy, but C3 was decreased at day 25 of pregnancy. C3 protein was located in the subcapsular sinuses and lymph sinuses of the maternal lymph node. In summary, the present study detected changes in the expression levels of complement components in maternal lymph nodes, which may be associated with maternal immune regulation during early pregnancy in sheep.
Collapse
Affiliation(s)
- Leying Zhang
- Department of Animal Science, School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, Hebei 056038, P.R. China
| | - Lidong Cao
- Department of Animal Science, School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, Hebei 056038, P.R. China
| | - Pengfei Feng
- Department of Animal Science, School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, Hebei 056038, P.R. China
| | - Xu Han
- Department of Animal Science, School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, Hebei 056038, P.R. China
| | - Ling Yang
- Department of Animal Science, School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, Hebei 056038, P.R. China
| |
Collapse
|
4
|
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
|
5
|
Zhang L, Zhang Q, Wang H, Feng P, Yang G, Yang L. Effects of early pregnancy on the complement system in the ovine thymus. Vet Res Commun 2021; 46:137-145. [PMID: 34559379 DOI: 10.1007/s11259-021-09837-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 09/17/2021] [Indexed: 11/30/2022]
Abstract
The complement system is crucial for the innate immune system, and complement activation is related to abnormal pregnancy in mice and humans. It is hypothesized that the complement system participates in maternal thymic immune regulation during early pregnancy in sheep. In this study, maternal thymuses were sampled on day 16 of the estrous cycle, and days 13, 16 and 25 of gestation in sheep. Quantitative real-time PCR, Western blot and immunohistochemistry analyses were used to analyze the expression of the complement components C1q, C1r, C1s, C2, C3, C4a, C5b and C9 in the maternal thymus. The results revealed that the mRNA and protein expression of C1r, C1s, C2, C3 and C4a was inhibited by early pregnancy, and the pregnancy recognition signal induced upregulation of C1q, C5b and C9 expression at day 16 of gestation. Furthermore, C3 protein was mostly located in epithelial reticular cells and thymic corpuscles, which may be involved in immune regulation. In summary, early pregnancy inhibits the complement system in the maternal thymus, which may be essential for the maternal immune regulation and successful pregnancy in sheep.
Collapse
Affiliation(s)
- Leying Zhang
- Department of Animal Science, School of Life Sciences and Food Engineering, Hebei University of Engineering, No. 19 Taiji Road, Handan, 056038, China
| | - Qiongao Zhang
- Department of Animal Science, School of Life Sciences and Food Engineering, Hebei University of Engineering, No. 19 Taiji Road, Handan, 056038, China
| | - Haichao Wang
- Department of Animal Science, School of Life Sciences and Food Engineering, Hebei University of Engineering, No. 19 Taiji Road, Handan, 056038, China
| | - Pengfei Feng
- Department of Animal Science, School of Life Sciences and Food Engineering, Hebei University of Engineering, No. 19 Taiji Road, Handan, 056038, China
| | - Gengxin Yang
- Department of Animal Science, School of Life Sciences and Food Engineering, Hebei University of Engineering, No. 19 Taiji Road, Handan, 056038, China
| | - Ling Yang
- Department of Animal Science, School of Life Sciences and Food Engineering, Hebei University of Engineering, No. 19 Taiji Road, Handan, 056038, China.
| |
Collapse
|
6
|
Abstract
Preeclampsia is a medical condition affecting 5-10% of pregnancies. It has serious effects on the health of the pregnant mother and developing fetus. While possible causes of preeclampsia are speculated, there is no consensus on its etiology. The advancement of big data and high-throughput technologies enables to study preeclampsia at the new and systematic level. In this review, we first highlight the recent progress made in the field of preeclampsia research using various omics technology platforms, including epigenetics, genome-wide association studies (GWAS), transcriptomics, proteomics and metabolomics. Next, we integrate the results in individual omic level studies, and show that despite the lack of coherent biomarkers in all omics studies, inhibin is a potential preeclamptic biomarker supported by GWAS, transcriptomics and DNA methylation evidence. Using network analysis on the biomarkers of all the literature reviewed here, we identify four striking sub-networks with clear biological functions supported by previous molecular-biology and clinical observations. In summary, omics integration approach offers the promise to understand molecular mechanisms in preeclampsia.
Collapse
|
7
|
Mei C, Yang W, Wei X, Wu K, Huang D. The Unique Microbiome and Innate Immunity During Pregnancy. Front Immunol 2019; 10:2886. [PMID: 31921149 PMCID: PMC6929482 DOI: 10.3389/fimmu.2019.02886] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 11/25/2019] [Indexed: 12/26/2022] Open
Abstract
A successful pregnancy depends on not only the tolerance of the fetal immune system by the mother but also resistance against the threat of hazardous microorganisms. Infection with pathogenic microorganisms during pregnancy may lead to premature delivery, miscarriage, growth restriction, neonatal morbidity, and other adverse outcomes. Moreover, the host also has an intact immune system to avoid these adverse outcomes. It is important to note the presence of normal bacteria in the maternal reproductive tract and the principal role of the maternal-placental-fetal interaction in antimicrobial immunity. Previous studies mainly focused on maternal infection during pregnancy. However, this review summarizes the new views on the study of the maternal microbiome and expounds the innate immune defense mechanism of the maternal vagina and decidua as well as how cytotrophoblasts and syncytiotrophoblasts recognize and kill bacteria in the placenta. Fetal immune systems, thought to be weak, also exhibit an immune defense function that is indispensable for maintaining the safety of the fetus. The skin, lungs, and intestines of the fetus during pregnancy constitute the main immune barriers. These findings will provide a new understanding of the effects of normal microbial flora and how the host resists harmful microbes during pregnancy. We believe that it may also contribute to the reference on the clinical prevention and treatment of gestational infection to avoid adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Chunlei Mei
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weina Yang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Wei
- Second Affiliated Hospital of Jinlin University, Changchun, China
| | - Kejia Wu
- Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Donghui Huang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
8
|
Smoking alters hydroxyprostaglandin dehydrogenase expression in fetal membranes. Reprod Toxicol 2018; 82:18-24. [PMID: 30248390 DOI: 10.1016/j.reprotox.2018.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 08/15/2018] [Accepted: 09/19/2018] [Indexed: 11/20/2022]
Abstract
INTRODUCTION The way in which tobacco smoking increases the risk of preterm labor remains uncertain. Altered prostaglandin metabolism is one potential mechanism. METHODS Proteins in fetal membrane samples (amniochoriodecidua) from 20 women were relatively quantified using Tandem Mass Tagging nano-liquid chromatography mass spectrometry. RESULTS Prostaglandin synthases and two enzymes involved in prostaglandin degradation, hydroxyprostaglandin dehydrogenase (HPGD) and CBR1, were detected by the mass spectrometer. The expression of HPGD was significantly lower in smokers relative to non-smokers (0.43 fold, p = 0.016). There was no effect of labor, inflammatory status or gestational age on the HPGD levels. DISCUSSION We describe for the first time an association between maternal smoking and HPGD expression. We propose that reduced expression of HPGD is one mechanism through which smoking may contribute to preterm labor. Lower levels of this enzyme, key to metabolising prostaglandins, may result in higher levels of prostaglandins and therefore precipitate labor prematurely.
Collapse
|
9
|
Canda MT, Caglayan LD, Demir N, Ortaç R. Increased C4d and Bb immunoreactivity and decreased MBL immunoreactivity characterise first-time pathologic first-trimester miscarriage: a case-control study. J OBSTET GYNAECOL 2017; 38:90-95. [DOI: 10.1080/01443615.2017.1328589] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- M. Tunc Canda
- Department of Obstetrics and Gynaecology, Kent Hospital, İzmir, Turkey
| | | | - Namik Demir
- Department of Obstetrics and Gynaecology, Kent Hospital, İzmir, Turkey
| | - Ragıp Ortaç
- Department of Pathology, Kent Hospital, İzmir, Turkey
| |
Collapse
|
10
|
Shanmugalingam R, Hsu D, Makris A. Pregnancy-induced atypical haemolytic uremic syndrome: A new era with eculizumab. Obstet Med 2017; 11:28-31. [PMID: 29636811 DOI: 10.1177/1753495x17704563] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 03/12/2017] [Indexed: 11/15/2022] Open
Abstract
Pregnancy is a well-recognised trigger of atypical haemolytic syndrome (P-aHUS) and often occurs in the post-partum period. Similar to atypical haemolytic uremic syndrome, it carries a poor prognosis with high morbidity particularly in the form of renal failure. Early recognition and intervention is crucial in its management particularly with the recent availability of eculizumab, a humanized monoclonal antibody to complement component C5, which has demonstrated drastic improvement in prognosis. The issue, however, is arriving at a timely diagnosis given the considerable amount of overlap in the clinical and biochemical manifestation of P-aHUS, HELLP syndrome (haemolysis, elevated liver enzyme and low platelet count) and other hypertensive disorders of pregnancy. We present a case report and literature review that highlights the clinical conundrum of arriving at the diagnosis. We also highlight the importance of early management of P-aHUS with eculizumab and its impact on improving morbidity.
Collapse
Affiliation(s)
- Renuka Shanmugalingam
- Department of Renal Medicine, Liverpool Hospital, NSW, Australia.,School of Medicine, Western Sydney University, NSW, Australia
| | - Danny Hsu
- Department of Haematology, Liverpool Hospital, NSW, Australia
| | - Angela Makris
- Department of Renal Medicine, Liverpool Hospital, NSW, Australia.,School of Medicine, Western Sydney University, NSW, Australia.,University of New South Wales, NSW, Australia
| |
Collapse
|
11
|
Kouser L, Madhukaran SP, Shastri A, Saraon A, Ferluga J, Al-Mozaini M, Kishore U. Emerging and Novel Functions of Complement Protein C1q. Front Immunol 2015; 6:317. [PMID: 26175731 PMCID: PMC4484229 DOI: 10.3389/fimmu.2015.00317] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 06/02/2015] [Indexed: 02/02/2023] Open
Abstract
Complement protein C1q, the recognition molecule of the classical pathway, performs a diverse range of complement and non-complement functions. It can bind various ligands derived from self, non-self, and altered self and modulate the functions of immune and non-immune cells including dendritic cells and microglia. C1q involvement in the clearance of apoptotic cells and subsequent B cell tolerance is more established now. Recent evidence appears to suggest that C1q plays an important role in pregnancy where its deficiency and dysregulation can have adverse effects, leading to preeclampsia, missed abortion, miscarriage or spontaneous loss, and various infections. C1q is also produced locally in the central nervous system, and has a protective role against pathogens and possible inflammatory functions while interacting with aggregated proteins leading to neurodegenerative diseases. C1q role in synaptic pruning, and thus CNS development, its anti-cancer effects as an immune surveillance molecule, and possibly in aging are currently areas of extensive research.
Collapse
Affiliation(s)
- Lubna Kouser
- Centre for Infection, Immunity and Disease Mechanisms, College of Health and Life Sciences, Brunel University London , Uxbridge , UK
| | - Shanmuga Priyaa Madhukaran
- Centre for Infection, Immunity and Disease Mechanisms, College of Health and Life Sciences, Brunel University London , Uxbridge , UK ; Centre for Biotechnology and Bioinformatics, Jawaharlal Nehru Institute for Advanced Studies, School of Life Sciences , Secunderabad , India
| | - Abhishek Shastri
- St. Ann's Hospital, Dorset Healthcare University NHS Foundation Trust , Poole , UK
| | - Anuvinder Saraon
- Centre for Infection, Immunity and Disease Mechanisms, College of Health and Life Sciences, Brunel University London , Uxbridge , UK
| | - Janez Ferluga
- Centre for Infection, Immunity and Disease Mechanisms, College of Health and Life Sciences, Brunel University London , Uxbridge , UK
| | - Maha Al-Mozaini
- Department of Infection and Immunity, King Faisal Specialist Hospital and Research Centre , Riyadh , Saudi Arabia
| | - Uday Kishore
- Centre for Infection, Immunity and Disease Mechanisms, College of Health and Life Sciences, Brunel University London , Uxbridge , UK
| |
Collapse
|
12
|
Yuan W, Heesom K, Phillips R, Chen L, Trinder J, López Bernal A. Low abundance plasma proteins in labour. Reproduction 2012; 144:505-18. [DOI: 10.1530/rep-12-0114] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Every year, millions of births worldwide are complicated by prematurity or difficult post-term deliveries, resulting in a high incidence of perinatal mortality and morbidity. Our poor understanding of human parturition is a key reason for our inability to improve the management of preterm and post-term birth. In this study, we used proteomic techniques to look into protein changes in placental blood plasma obtained from women before or after spontaneous or induced labour, with vaginal or caesarean section deliveries. Our aim was to understand the basic mechanisms of human parturition regardless of whether the signals that trigger labour are of maternal and/or fetal origin. We found proteins from 33 genes with significantly altered expression profiles in relation to mode of labour and delivery. Most changes in labour occurred in proteins associated with ‘immune and defence responses’. Although the signal transduction and regulation of these pathways varied among modes of delivery, hepatocyte nuclear factor 1 homeobox A emerged as a shared protein in the mechanism of labour. Moreover, several apolipoproteins such as apolipoprotein A-IV and APOE were found to change with labour, and these changes were also confirmed in maternal plasma. This study has identified significant protein changes in placental intervillous plasma with labour and has revealed several pathways related to human parturition.
Collapse
|
13
|
Bulla R, Bossi F, Tedesco F. The complement system at the embryo implantation site: friend or foe? Front Immunol 2012; 3:55. [PMID: 22566936 PMCID: PMC3341982 DOI: 10.3389/fimmu.2012.00055] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Accepted: 03/02/2012] [Indexed: 12/19/2022] Open
Abstract
An inflammatory-like process and vascular remodeling represent the main changes that occur in decidua in the early phase of pregnancy. These changes are partly induced by trophoblast cells that colonize the decidua and are also contributed by the complement system, which can easily be activated as a result of tissue remodeling. Local control by several complement regulators including surface-bound and soluble molecules is critical to prevent complement-mediated tissue damage in normal pregnancy. C7 expressed on the endothelial cells (ECs) surface has been recognized as a novel complement regulator involved in the control of the proinflammatory effect of the terminal complement complex. The protective role of placental complement regulators in pregnancy is underscored by the recent finding of an association of preeclampsia with mutations in the genes encoding for some of these proteins. Complement components produced at feto-maternal interface serve an important function in placental development. C1q synthesized by decidual ECs and expressed on the cell surface is particularly important in this regard because it acts as a molecular bridge between endovascular trophoblast and ECs. C1q is also produced by extravillous trophoblast and is used to favor trophoblast migration through the decidua. Defective expression of C1q by trophoblast is associated with impaired trophoblast invasion of decidua and may have important implications in pregnancy disorders such as preeclampsia characterized by reduced vascular remodeling.
Collapse
Affiliation(s)
- R Bulla
- Department of Life Sciences, University of Trieste Trieste, Italy
| | | | | |
Collapse
|
14
|
Fakhouri F, Roumenina L, Provot F, Sallée M, Caillard S, Couzi L, Essig M, Ribes D, Dragon-Durey MA, Bridoux F, Rondeau E, Frémeaux-Bacchi V. Pregnancy-associated hemolytic uremic syndrome revisited in the era of complement gene mutations. J Am Soc Nephrol 2010; 21:859-67. [PMID: 20203157 DOI: 10.1681/asn.2009070706] [Citation(s) in RCA: 293] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In contrast to pregnancy-associated thrombotic thrombocytopenic purpura, the pathogenesis and presentation of pregnancy-associated atypical hemolytic uremic syndrome (P-aHUS) remain ill-defined. We conducted a retrospective study to assess the presentation and outcomes of patients presenting with P-aHUS and the prevalence of alternative C3 convertase dysregulation. P-aHUS occurred in 21 of the 100 adult female patients with atypical HUS, with 79% presenting postpartum. We detected complement abnormalities in 18 of the 21 patients. The outcomes were poor: 62% reached ESRD by 1 month and 76% by last follow-up. The risk for P-aHUS was highest during a second pregnancy. Thirty-five women, 26 (74%) of whom had complement abnormalities, had at least one pregnancy before the onset of a non-pregnancy-related aHUS. Outcomes did not differ between patients with pregnancy-related and non-pregnancy-related aHUS. Mutations in the SCR19-20 domains of factor H were less frequent in P-aHUS patients compared with non-pregnancy-related aHUS. Pregnancies in female patients with complement abnormalities (n = 44) were complicated by fetal loss and preeclampsia in 4.8% and 7.7%, respectively. Better understanding of complement dysregulation in pregnancy complications is essential, especially to guide development of pharmacologic agents to modulate this system.
Collapse
Affiliation(s)
- Fadi Fakhouri
- Department of Nephrology and UMR 643, CHU de Nantes, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Bulla R, Bossi F, Agostinis C, Radillo O, Colombo F, De Seta F, Tedesco F. Complement production by trophoblast cells at the feto-maternal interface. J Reprod Immunol 2009; 82:119-25. [DOI: 10.1016/j.jri.2009.06.124] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2008] [Revised: 05/27/2009] [Accepted: 06/05/2009] [Indexed: 12/21/2022]
|
16
|
Soto E, Romero R, Richani K, Yoon BH, Chaiworapongsa T, Vaisbuch E, Mittal P, Erez O, Gotsch F, Mazor M, Kusanovic JP. Evidence for complement activation in the amniotic fluid of women with spontaneous preterm labor and intra-amniotic infection. J Matern Fetal Neonatal Med 2009; 22:983-92. [PMID: 19900036 PMCID: PMC3437778 DOI: 10.3109/14767050902994747] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE The complement system plays an important role in host defense against infection. Concentrations of complement split products or anaphylatoxins (C3a, C4a, and C5a) in biological fluids are considered to reflect complement activation. The purpose of this study was to determine if term and preterm parturition are associated with evidence of complement activation in the amniotic fluid. STUDY DESIGN Amniotic fluid (AF) samples were collected from 270 women in the following groups: (1) normal pregnant women in midtrimester (n = 70), (2) term not in labor (n = 23), (3) term in labor (n = 48), and (4) preterm labor (PTL) (n = 129). PTL was categorized into: (a) PTL without microbial invasion of the amniotic cavity (MIAC) who delivered at term (n = 42), (b) PTL who delivered preterm without MIAC (n = 57), and (c) PTL with MIAC (n = 30). C5a, C4a, and C3a concentrations in amniotic fluid were determined by ELISA. Nonparametric tests were used for statistical analysis. RESULTS (1) The median AF C5a concentration was higher in women at term than that of those in the midtrimester (p = 0.02); (2) Spontaneous labor at term was not associated with changes in AF concentrations of anaphylatoxins C3a, C4a, and C5a (all p > 0.05); (3) Among patients with PTL who delivered preterm, those with MIAC had higher AF C4a and C5a concentrations than those without infection (p < 0.01); and (4) AF C3a, C4a, and C5a concentrations were higher in patients with PTL with MIAC than in those with PTL without MIAC who delivered at term. CONCLUSION Patients with spontaneous preterm labor and intact membranes with microbial invasion of the amniotic cavity had higher median amniotic fluid concentration of complement split products C3a, C4a, and C5a than patients without intra-amniotic infection. These findings suggest that preterm labor in the context of infection is associated with activation of the complement system.
Collapse
Affiliation(s)
- Eleazar Soto
- Perinatology Research Branch, NICHD/NIH/DHHS, Hutzel Women’s Hospital, Bethesda, Maryland and Detroit, Michigan, USA
- Wayne State University School of Medicine, Department of Obstetrics and Gynecology, Detroit, Michigan, USA
| | - Roberto Romero
- Perinatology Research Branch, NICHD/NIH/DHHS, Hutzel Women’s Hospital, Bethesda, Maryland and Detroit, Michigan, USA
- Wayne State University School of Medicine, Department of Obstetrics and Gynecology, Detroit, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
| | - Karina Richani
- Perinatology Research Branch, NICHD/NIH/DHHS, Hutzel Women’s Hospital, Bethesda, Maryland and Detroit, Michigan, USA
| | - Bo Hyun Yoon
- Department of Obstetrics and Gynecology, Seoul National University, Seoul, Korea
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, NICHD/NIH/DHHS, Hutzel Women’s Hospital, Bethesda, Maryland and Detroit, Michigan, USA
- Wayne State University School of Medicine, Department of Obstetrics and Gynecology, Detroit, Michigan, USA
| | - Edi Vaisbuch
- Perinatology Research Branch, NICHD/NIH/DHHS, Hutzel Women’s Hospital, Bethesda, Maryland and Detroit, Michigan, USA
- Wayne State University School of Medicine, Department of Obstetrics and Gynecology, Detroit, Michigan, USA
| | - Pooja Mittal
- Perinatology Research Branch, NICHD/NIH/DHHS, Hutzel Women’s Hospital, Bethesda, Maryland and Detroit, Michigan, USA
- Wayne State University School of Medicine, Department of Obstetrics and Gynecology, Detroit, Michigan, USA
| | - Offer Erez
- Perinatology Research Branch, NICHD/NIH/DHHS, Hutzel Women’s Hospital, Bethesda, Maryland and Detroit, Michigan, USA
- Wayne State University School of Medicine, Department of Obstetrics and Gynecology, Detroit, Michigan, USA
| | - Francesca Gotsch
- Perinatology Research Branch, NICHD/NIH/DHHS, Hutzel Women’s Hospital, Bethesda, Maryland and Detroit, Michigan, USA
| | - Moshe Mazor
- Department of Obstetrics and Gynecology, Ben Gurion University of the Negev, Beer-Sheva, Israel
| | - Juan Pedro Kusanovic
- Perinatology Research Branch, NICHD/NIH/DHHS, Hutzel Women’s Hospital, Bethesda, Maryland and Detroit, Michigan, USA
- Wayne State University School of Medicine, Department of Obstetrics and Gynecology, Detroit, Michigan, USA
| |
Collapse
|
17
|
Vaisbuch E, Romero R, Erez O, Tovi SM, Pedro KJ, Soto E, Gotsch F, Dong Z, Chaiworapongsa T, Kim SK, Mittal P, Pacora P, Yeo L, Hassan SS. Fragment Bb in amniotic fluid: evidence for complement activation by the alternative pathway in women with intra-amniotic infection/inflammation. J Matern Fetal Neonatal Med 2009; 22:905-16. [PMID: 19603351 PMCID: PMC3609549 DOI: 10.1080/14767050902994663] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Fragment Bb is an activator of the alternative pathway of the complement system. Recently, increased first trimester maternal plasma concentrations of this fragment were reported in patients destined to have a spontaneous preterm delivery before 34 weeks of gestation. The aim of this study was to determine whether the amniotic fluid (AF) concentrations of fragment Bb change with gestational age, spontaneous labor (term and preterm) and in the presence of intra-amniotic infection/inflammation (IAI). STUDY DESIGN This cross-sectional study included patients in the following groups: (1) mid-trimester (n = 64); (2) term in spontaneous labor (n = 70); (3) term not in labor (n = 43); (4) spontaneous preterm labor (PTL) who delivered at term (n = 76); (5) PTL without IAI who delivered preterm (n = 73); (6) PTL with IAI (n = 76); (7) preterm prelabor rupture of membranes (PROM) without IAI (n = 71); and (8) preterm PROM with IAI (n = 71). Fragment Bb concentration in AF was determined by an enzyme-linked immunoassay. Non-parametric statistics were used for analyses. RESULTS (1) Fragment Bb was detected in all AF samples (n = 544); (2) The median AF concentration of fragment Bb in patients at term not in labor was significantly higher than that of those in the mid-trimester [2.42 microg/ml, interquartile range (IQR) 1.78-3.22 vs. 1.64 microg/ml, IQR 1.06-3.49; p < 0.001]; (3) Among patients with PTL, those with IAI had a higher median AF fragment Bb concentration than that of woman without IAI, who delivered preterm (4.82 microg/ml, IQR 3.32-6.08 vs. 3.67 microg/ml, IQR 2.35-4.57; p < 0.001) and than that of women with an episode of PTL, who delivered at term (3.21 microg/ml, IQR 2.39-4.16; p < 0.001); (4) Similarly, among patients with preterm PROM, the median AF fragment Bb concentration was higher in individuals with IAI than in those without IAI (4.24 microg/ml, IQR 2.58-5.79 vs. 2.79 microg/ml, IQR 2.09-3.89; p < 0.001). (5) Among patients at term, the median AF fragment Bb concentration did not differ between women with spontaneous labor and those without labor (term in labor: 2.47 microg/ml, IQR 1.86-3.22; p = 0.97). CONCLUSIONS (1) Fragment Bb, an activator of the alternative complement pathway, is a physiologic constituent of the AF, and its concentration increases with advancing gestational age; (2) AF concentrations of fragment Bb are higher in pregnancies complicated with IAI; and (3) labor at term is not associated with changes in the AF concentrations of fragment Bb. These findings suggest a role for fragment Bb in the host immune response against IAI.
Collapse
MESH Headings
- Adult
- Amniotic Fluid/chemistry
- Amniotic Fluid/metabolism
- Bacterial Infections/immunology
- Bacterial Infections/metabolism
- Chorioamnionitis/immunology
- Chorioamnionitis/metabolism
- Complement Activation/physiology
- Complement Factor B/analysis
- Complement Factor B/metabolism
- Complement Factor B/physiology
- Complement Pathway, Alternative/physiology
- Cross-Sectional Studies
- Female
- Fetal Membranes, Premature Rupture/immunology
- Fetal Membranes, Premature Rupture/metabolism
- Humans
- Inflammation/immunology
- Inflammation/metabolism
- Obstetric Labor, Premature/immunology
- Obstetric Labor, Premature/metabolism
- Osmolar Concentration
- Pregnancy
- Pregnancy Complications, Infectious/immunology
- Pregnancy Complications, Infectious/metabolism
- Pregnancy Trimester, Second/immunology
- Pregnancy Trimester, Second/metabolism
- Pregnancy Trimester, Third/immunology
- Pregnancy Trimester, Third/metabolism
- Young Adult
Collapse
Affiliation(s)
- Edi Vaisbuch
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland and Detroit, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
| | - Roberto Romero
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland and Detroit, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
- Wayne State University School of Medicine, Department of Obstetrics and Gynecology, Detroit, Michigan, USA
| | - Offer Erez
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland and Detroit, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
| | - Shali Mazaki Tovi
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland and Detroit, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
| | - Kusanovic Juan Pedro
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland and Detroit, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
| | - Eleazar Soto
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland and Detroit, Michigan, USA
| | - Francesca Gotsch
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland and Detroit, Michigan, USA
| | - Zhong Dong
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland and Detroit, Michigan, USA
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland and Detroit, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
| | - Sun Kwon Kim
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland and Detroit, Michigan, USA
| | - Pooja Mittal
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland and Detroit, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
| | - Percy Pacora
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland and Detroit, Michigan, USA
| | - Lami Yeo
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland and Detroit, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
| | - Sonia S. Hassan
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, Maryland and Detroit, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|