1
|
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
|
2
|
Bangi EF, Yousuf MH, Upadhyay S, Jain P, Jain R. Comprehensive Review of Hypertensive Disorders Related to Pregnancy. South Med J 2023; 116:482-489. [PMID: 37263611 DOI: 10.14423/smj.0000000000001571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Hypertensive disorder of pregnancy is a common complication during pregnancy that affects approximately 10% of pregnancies and is responsible for nearly 14% of maternal deaths worldwide. It affects the mother and the fetus simultaneously, sometimes putting the health of the mother and the fetus at odds with each other. It may present with only hypertension and proteinuria or with life-threatening complications in the mother such as eclampsia; stroke; acute pulmonary edema; acute renal failure; disseminated intravascular coagulation; placental abruption; hemolysis, elevated liver enzymes, and low platelet syndrome; pregnancy loss; and fetal growth restriction and prematurity resulting from the frequent need of delivering preterm in the fetus. In this review, we aimed to describe hypertensive disorders of pregnancy, mainly preeclampsia and chronic hypertension in pregnancy, by discussing the pathophysiology, the central role of abnormal placentation, the release of antiangiogenic factors in the circulation and immunological factors, the clinical outcome in the mother and the fetus, and the diagnostic criteria and principles of management of both the conditions. We also discuss possible screening methods and prevention of preeclampsia using low-dose aspirin and eclampsia prophylaxis.
Collapse
Affiliation(s)
- Eera Fatima Bangi
- From the Seth Gordhandas Sunderdas Medical College and King Edward Memorial Hospital, Mumbai, India
| | | | | | | | - Rohit Jain
- Department of Internal Medicine, Penn State Milton S. Hershey Medical Center, Hershey, Pennsylvania
| |
Collapse
|
3
|
Kurokawa M, Takeshita A, Hashimoto S, Koyama M, Morimoto Y, Tachibana D. Prevention of intrauterine fetal growth restriction by administrating C1q/TNF-related protein 6, a specific inhibitor of the alternative complement pathway. J Assist Reprod Genet 2022; 39:2191-2199. [PMID: 35907048 PMCID: PMC9474761 DOI: 10.1007/s10815-022-02582-1] [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: 01/11/2022] [Accepted: 07/19/2022] [Indexed: 10/16/2022] Open
Abstract
PURPOSE The latest treatments do not sufficiently prevent miscarriage and fetal growth restriction (FGR) in pregnant women. Here, we assessed the effects of a human protein, CTRP6, that specifically inhibits the activation of the alternative complement pathway on miscarriage, fetal and placental development. METHODS Pregnant CBA/J mice mated with DBA/2 male mice as a model of spontaneous abortion and FGR were randomly divided into the control and CTRP6 groups. In the CTRP6 group, the mice were intravenously administered CTRP6 on days 4.5 and 6.5 post-conception (dpc). The abortion rate and fetal and placental weights on 14.5 dpc were examined. Remodeling of the spiral artery was also assessed. RESULTS The abortion rate in the CTRP6 group (13%) was reduced compared to the control group (21%), but there was no statistical difference. The placental and fetal weights in the CTRP6 group were also heavier than those in the control (P < 0.05). Moreover, the thickness of the blood vessel wall in the CTRP6 group was significantly thinner than that in the control (P < 0.05) and comparable to that in the non-abortion model (CBA/J x BALB). The ratio of the inner-per-the-outer diameter of the spiral artery increased more in the CTRP6 group than that in the control (P < 0.05). As well, the Th1/Th2 cytokine ratio was significantly reduced by CTRP6 treatment. CONCLUSIONS Taken together, the supplementation with a protein that regulates the alternative complement pathway in vivo improves FGR and promotes spiral artery remodeling in a mouse model of miscarriage and FGR.
Collapse
Affiliation(s)
- Mayu Kurokawa
- Women's Lifecare Medicine, Obstetrics and Gynecology, School of Medicine, Osaka City University, 545-8585, Osaka, Japan
- Graduate School of Medicine, Reproductive Science, Osaka City University, Osaka, 545-8585, Japan
| | - Ai Takeshita
- Graduate School of Medicine, Reproductive Science, Osaka City University, Osaka, 545-8585, Japan
- Department of Anatomy and Neurobiology, Faculty of Medicine, Kindai University, Osaka, 589-8511, Japan
| | - Shu Hashimoto
- Graduate School of Medicine, Reproductive Science, Osaka City University, Osaka, 545-8585, Japan.
| | - Masayasu Koyama
- Women's Lifecare Medicine, Obstetrics and Gynecology, School of Medicine, Osaka City University, 545-8585, Osaka, Japan
| | | | - Daisuke Tachibana
- Women's Lifecare Medicine, Obstetrics and Gynecology, School of Medicine, Osaka City University, 545-8585, Osaka, Japan
| |
Collapse
|
4
|
Manning JE, Anderson RM, Hill A, Zeidan D, Ciantar E. Pregnancy outcomes in women receiving eculizumab for the management of paroxysmal nocturnal haemoglobinuria. Obstet Med 2022; 15:45-49. [PMID: 35444730 PMCID: PMC9014543 DOI: 10.1177/1753495x211019899] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 04/30/2021] [Accepted: 04/30/2021] [Indexed: 11/15/2022] Open
Abstract
Aims To report pregnancy outcomes and complications in women receiving eculizumab for the management of paroxysmal nocturnal haemoglobinuria. Methods A service evaluation of routinely collected medical records across 49 pregnancies in 21 women. Results Eculizumab was used in 37 pregnancies, 31 of which (83.8%) ended in live birth. Eight infants (25.8%) were born prematurely. Over half (54%) of women required increases in their dose of eculizumab to control their haemolysis. There were no reported cases of maternal thrombosis. Major ante/postpartum bleeding necessitating urgent intervention was reported in 10.8% of pregnancies. There were two cases of intrauterine death and three miscarriages. There were no maternal or neonatal deaths. Three newborns required prolonged hospital stays. Conclusions Eculizumab appears to benefit pregnant women with paroxysmal nocturnal haemoglobinuria and pregnancy outcomes following its use are largely good.
Collapse
Affiliation(s)
| | | | - Anita Hill
- Department of Haematology, Leeds
Teaching Hospitals NHS Trust, Leeds, UK
| | - Doaa Zeidan
- Department of Obstetrics &
Gynaecology, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Etienne Ciantar
- Department of Obstetrics &
Gynaecology, Leeds Teaching Hospitals NHS Trust, Leeds, UK,Etienne Ciantar, Division of Women's and
Children's Health, Leeds Teaching Hospitals NHS Trust, Room 9.88, Worsley
Building, Leeds LS29NL, UK.
| |
Collapse
|
5
|
Liu T, Guo X, Liao Y, Liu Y, Zhu Y, Chen X. Correlation Between the Presence of Antinuclear Antibodies and Recurrent Pregnancy Loss: A Mini Review. Front Endocrinol (Lausanne) 2022; 13:873286. [PMID: 35600596 PMCID: PMC9114698 DOI: 10.3389/fendo.2022.873286] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/29/2022] [Indexed: 12/15/2022] Open
Abstract
In the past decade, the incidence of recurrent pregnancy loss (RPL) has increased significantly, and immunological disorders have been considered as one of the possible causes contributing to RPL. The presence of antinuclear antibodies (ANAs) is regarded as a typical antibody of autoimmunity. However, the relationship between the presence of ANAs and RPL, the underlying mechanism, and the possible role of immunotherapy is still controversial. The aim of this mini review is to assess the association between ANAs and RPL and the effects of immunotherapy on pregnancy outcomes in women with positive ANAs and a history of RPL from the available data and to provide a relevant reference basis for clinical application in this group of women.
Collapse
Affiliation(s)
- Ting Liu
- Department of Obstetrics and Gynaecology, Shenzhen Baoan Women’s and Children’s Hospital, Jinan University, Shenzhen, China
| | - Xi Guo
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Ying Liao
- Department of Obstetrics and Gynaecology, Shenzhen Baoan Women’s and Children’s Hospital, Jinan University, Shenzhen, China
| | - Yingyu Liu
- Department of Obstetrics and Gynaecology, Shenzhen Baoan Women’s and Children’s Hospital, Jinan University, Shenzhen, China
| | - Yuanfang Zhu
- Department of Obstetrics and Gynaecology, Shenzhen Baoan Women’s and Children’s Hospital, Jinan University, Shenzhen, China
- *Correspondence: Yuanfang Zhu, ; Xiaoyan Chen, ;
| | - Xiaoyan Chen
- Department of Obstetrics and Gynaecology, Shenzhen Baoan Women’s and Children’s Hospital, Jinan University, Shenzhen, China
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- *Correspondence: Yuanfang Zhu, ; Xiaoyan Chen, ;
| |
Collapse
|
6
|
Low Preconception Complement Levels Are Associated with Adverse Pregnancy Outcomes in a Multicenter Study of 260 Pregnancies in 197 Women with Antiphospholipid Syndrome or Carriers of Antiphospholipid Antibodies. Biomedicines 2021; 9:biomedicines9060671. [PMID: 34208130 PMCID: PMC8230784 DOI: 10.3390/biomedicines9060671] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/04/2021] [Accepted: 06/08/2021] [Indexed: 12/18/2022] Open
Abstract
Antiphospholipid antibodies (aPL) can induce fetal loss in experimental animal models. Human studies did find hypocomplementemia associated with pregnancy complications in patients with antiphospholipid syndrome (APS), but these results are not unanimously confirmed. To investigate if the detection of low C3/C4 could be considered a risk factor for adverse pregnancy outcomes (APO) in APS and aPL carriers' pregnancies we performed a multicenter study including 503 pregnancies from 11 Italian and 1 Russian centers. Data in women with APS and asymptomatic carriers with persistently positive aPL and preconception complement levels were available for 260 pregnancies. In pregnancies with low preconception C3/C4, a significantly higher prevalence of pregnancy losses was observed (p = 0.008). A subgroup analysis focusing on triple aPL-positive patients found that preconception low C3 and/or C4 levels were associated with an increased rate of pregnancy loss (p = 0.05). Our findings confirm that decreased complement levels before pregnancy are associated with increased risk of APO. This has been seen only in women with triple aPL positivity, indeed single or double positivity does not show this trend. Complement levels are cheap and easy to be measured therefore they could represent a useful aid to identify patients at increased risk of pregnancy loss.
Collapse
|
7
|
Van Heertum K, Lam L, Richardson B, Cartwright MJ, Mesiano SA, Cameron MJ, Weinerman R. Blastocyst Vitrification and Trophectoderm Biopsy Cumulatively Alter Embryonic Gene Expression in a Mouse Model. Reprod Sci 2021; 28:2961-2971. [PMID: 33826099 DOI: 10.1007/s43032-021-00560-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/25/2021] [Indexed: 11/25/2022]
Abstract
Although embryo vitrification has been used extensively in human assisted reproductive technology (ART) and animal models, epidemiologic evidence and randomized controlled trials suggest differences in pregnancy/perinatal outcomes (birthweight, risk for preterm birth, and pre-eclampsia) between babies born from fresh versus frozen embryo transfers. To address the uncertainty surrounding the effects of laboratory manipulations of embryos on clinical outcomes, we subjected mouse blastocysts to increasing levels of manipulation for transcriptome analysis. Blastocysts were randomly divided into four groups: no manipulation (control), single vitrification/thaw (1 vit), double vitrification/thaw (2 vit), and single vitrification/thaw plus trophectoderm biopsy and again vitrified/thawed (2 vit + bx). Three sets of 15 blastocysts in each group were pooled for RNA sequencing, and differentially expressed genes (DEGs) and pathways were determined by statistical analysis. Blastocysts were also stained for ZO-1 and F-actin to assess cytoskeletal integrity. Freeze/thaw and biopsy manipulation affected multiple biological pathways. The most significant differences were detected in genes related to innate immunity, apoptosis, and mitochondrial function, with the magnitude of change proportional to the extent to manipulation. Significant disruptions were also seen in cytoskeletal staining, with greater disruptions seen with greater of manipulation. Our data suggests that embryo vitrification and biopsy affect embryo gene transcription, with several identified DEGs that may have plausible mechanisms for the clinical outcomes seen in human offspring following ART. Further study is required to determine whether these alterations in gene expression are associated with clinical differences seen in children born from fresh or frozen embryo transfer.
Collapse
Affiliation(s)
- Kristin Van Heertum
- University Hospitals Fertility Center/Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Lisa Lam
- CCRM New York Fertility, New York, NY, 10019, USA
| | - Brian Richardson
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Michael J Cartwright
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Sam A Mesiano
- Department of Reproductive Biology, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Mark J Cameron
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Rachel Weinerman
- University Hospitals Fertility Center/Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
8
|
Chighizola CB, Lonati PA, Trespidi L, Meroni PL, Tedesco F. The Complement System in the Pathophysiology of Pregnancy and in Systemic Autoimmune Rheumatic Diseases During Pregnancy. Front Immunol 2020; 11:2084. [PMID: 32973817 PMCID: PMC7481445 DOI: 10.3389/fimmu.2020.02084] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 07/30/2020] [Indexed: 12/16/2022] Open
Abstract
The complement system plays a double role in pregnancy exerting both protective and damaging effects at placental level. Complement activation at fetal-maternal interface participates in protection against infectious agents and helps remove apoptotic and necrotic cells. Locally synthesized C1q contributes to the physiologic vascular remodeling of spiral arteries characterized by loss of smooth muscle cells and transformation into large dilated vessels. Complement activation triggered by the inflammatory process induced by embryo implantation can damage trophoblast and other decidual cells that may lead to pregnancy complications if the cells are not protected by the complement regulators CD55, CD46, and CD59 expressed on cell surface. However, uncontrolled complement activation induces placental alterations resulting in adverse pregnancy outcomes. This may occur in pathological conditions characterized by placental localization of complement fixing antibodies directed against beta2-glycoprotein 1, as in patients with anti-phospholipid syndrome, or circulating immune complexes deposited in placenta, as in patients with systemic lupus erythematosus. In other diseases, such as preeclampsia, the mechanism of complement activation responsible for complement deposits in placenta is unclear. Conflicting results have been reported on the relevance of complement assays as diagnostic and prognostic tools to assess complement involvement in pregnant patients with these disorders.
Collapse
Affiliation(s)
- Cecilia Beatrice Chighizola
- Experimental Laboratory of Immunological and Rheumatologic Researches, Istituto Auxologico Italiano, IRCCS, Milan, Italy
| | - Paola Adele Lonati
- Experimental Laboratory of Immunological and Rheumatologic Researches, Istituto Auxologico Italiano, IRCCS, Milan, Italy
| | - Laura Trespidi
- Department of Obstetrics and Gynaecology, Fondazione Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Pier Luigi Meroni
- Experimental Laboratory of Immunological and Rheumatologic Researches, Istituto Auxologico Italiano, IRCCS, Milan, Italy
| | - Francesco Tedesco
- Experimental Laboratory of Immunological and Rheumatologic Researches, Istituto Auxologico Italiano, IRCCS, Milan, Italy
| |
Collapse
|
9
|
Kestlerová A, Krofta L, Žufić A, Hamplová Běhávková K, Račko J, Beneš J, Feyereisl J. Laboratory options for risk assessment of pregnancy pathologies. Physiol Res 2020; 68:S415-S425. [PMID: 32118472 DOI: 10.33549/physiolres.934376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The most effective method of screening for chromosomal abnormalities and evaluating the risk of pregnancy pathologies in the first trimester is combined screening. The algorithm of screening is based on the combination of maternal age, measuring of the nuchal translucency and the fetal heart rate and analysis of the placental products of free ß-hCG and PAPP-A. For the screening of preeclampsia, placental growth factor (PlGF) is added. To distinguish between preeclampsia and other pathologies caused by placental dysfunction it is recommended to also extend the screening with selected immunological markers. We concluded that elevated biochemical and immunological markers can help to predict the threat of preeclampsia in the third trimester. Some markers can probably predict the development of particularly severe pathological conditions.
Collapse
Affiliation(s)
- A Kestlerová
- Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University, Prague, Czech Republic.
| | | | | | | | | | | | | |
Collapse
|
10
|
Lind CM, Agugliaro J, Farrell TM. The metabolic response to an immune challenge in a viviparous snake, Sistrurus miliarius. J Exp Biol 2020; 223:jeb225185. [PMID: 32321747 DOI: 10.1242/jeb.225185] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 04/15/2020] [Indexed: 08/26/2023]
Abstract
Mounting an immune response may be energetically costly and require the diversion of resources away from other physiological processes. Yet, both the metabolic cost of immune responses and the factors that impact investment priorities remain poorly described in many vertebrate groups. For example, although viviparity has evolved many times in vertebrates, the relationship between immune function and pregnancy has been disproportionately studied in placental mammals. To examine the energetic costs of immune activation and the modulation of immune function during pregnancy in a non-mammalian vertebrate, we elicited an immune response in pregnant and non-pregnant pygmy rattlesnakes, Sistrurus miliarius, using lipopolysaccharide (LPS). Resting metabolic rate (RMR) was measured using flow-through respirometry. Immune function was examined using bactericidal assays and leukocyte counts. The RMR of pygmy rattlesnakes increased significantly in response to LPS injection. There was no statistically significant difference in the metabolic response of non-reproductive and pregnant snakes to LPS. Mean metabolic increments for pregnant females, non-reproductive females, and males were 13%, 18% and 26%, respectively. The ratio of heterophils to lymphocytes was elevated in response to LPS across reproductive categories; however, LPS did not impact plasma bactericidal ability in non-reproductive snakes. Although pregnant females had significantly higher plasma bactericidal ability compared with non-reproductive snakes prior to manipulation, their bactericidal ability declined in response to LPS. LPS administration also significantly reduced several litter characteristics, particularly when administrated relatively early in pregnancy. Our results indicate that immune performance is energetically costly and is altered during pregnancy, and that immune activation during pregnancy may result in tradeoffs that affect offspring in a viviparous reptile.
Collapse
Affiliation(s)
- Craig M Lind
- School of Natural Science and Mathematics, Stockton University, Galloway, NJ 08205, USA
| | - Joseph Agugliaro
- Department of Biological Sciences, Fairleigh Dickinson University, Madison, NJ 07940, USA
| | | |
Collapse
|
11
|
Hansen VL, Miller RD. Evidence for regulation of the complement system during pregnancy being ancient and conserved in mammals. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 104:103562. [PMID: 31785265 PMCID: PMC6937380 DOI: 10.1016/j.dci.2019.103562] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 11/26/2019] [Accepted: 11/26/2019] [Indexed: 06/10/2023]
Abstract
Here we demonstrate that regulation of the Complement (C') components of the immune system is an ancient and conserved feature of mammalian pregnancy. Transcript levels were reduced for complement components C3 and C4 throughout pregnancy in a marsupial, Monodelphis domestica. Downstream C' component transcripts were significantly less abundant relative to non-pregnant controls at the start of pregnancy but increased during late pregnancy, in some cases peaking close to parturition. These results are consistent with observations in human pregnancy that deposition of C5 through C9 on fetal membranes is associated with labor and parturition. Complement regulators CD46 and CD59 are present at the fetomaternal interface during M. domestica pregnancy as well, implying regulation of C' effector mechanisms is necessary for maintenance of normal marsupial pregnancy. Collectively these results support regulating the complement system may have contributed to the transition from oviparity to viviparity in mammals over 165 million years ago.
Collapse
Affiliation(s)
- Victoria L Hansen
- Center for Evolutionary and Theoretical Immunology, Department of Biology, University of New Mexico, Albuquerque, NM, USA
| | - Robert D Miller
- Center for Evolutionary and Theoretical Immunology, Department of Biology, University of New Mexico, Albuquerque, NM, USA.
| |
Collapse
|
12
|
Bilibio JP, Gama TB, Nascimento ICM, Meireles AJC, Aguiar ASC, Nascimento FC, Lorenzzoni PL. Causes of recurrent miscarriage after spontaneous pregnancy and after in vitro fertilization. Am J Reprod Immunol 2020; 83:e13226. [DOI: 10.1111/aji.13226] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 01/03/2020] [Accepted: 01/20/2020] [Indexed: 12/21/2022] Open
Affiliation(s)
- João Paolo Bilibio
- Department of Obstetrics and Gynecology Universidade Federal do Pará Belém Pará Brazil
- Clinica de Reprodução Assistida Pronatus Belém Pará Brazil
- Post doctoral Program of the Programa de Pós Graduação de Ciências Médicas da Universidade Federal do Rio Grande do Sul Porto Alegre Rio Grande do Su Brazil
- Grupo de Pesquisa Bilibio Universidade Federal do Pará Belém Pará Brazil
| | - Thiago Belém Gama
- Department of Obstetrics and Gynecology Universidade Federal do Pará Belém Pará Brazil
- Grupo de Pesquisa Bilibio Universidade Federal do Pará Belém Pará Brazil
| | - Izabel Cristina Magalhães Nascimento
- Department of Obstetrics and Gynecology Universidade Federal do Pará Belém Pará Brazil
- Grupo de Pesquisa Bilibio Universidade Federal do Pará Belém Pará Brazil
| | - Arivaldo José Conceição Meireles
- Clinica de Reprodução Assistida Pronatus Belém Pará Brazil
- Grupo de Pesquisa Bilibio Universidade Federal do Pará Belém Pará Brazil
| | - Ariene Simona Cohen Aguiar
- Department of Obstetrics and Gynecology Universidade Federal do Pará Belém Pará Brazil
- Grupo de Pesquisa Bilibio Universidade Federal do Pará Belém Pará Brazil
- Programa de Residência Médica em Ginecologia e Obstetrícia Hospital Bettina Ferro de Souza Universidade Federal do Pará Belém Pará Brazil
| | - Fábio Costa Nascimento
- Clinica de Reprodução Assistida Pronatus Belém Pará Brazil
- Grupo de Pesquisa Bilibio Universidade Federal do Pará Belém Pará Brazil
| | - Pânila Longhi Lorenzzoni
- Clinica de Reprodução Assistida Pronatus Belém Pará Brazil
- Grupo de Pesquisa Bilibio Universidade Federal do Pará Belém Pará Brazil
| |
Collapse
|
13
|
Pierik E, Prins JR, van Goor H, Dekker GA, Daha MR, Seelen MAJ, Scherjon SA. Dysregulation of Complement Activation and Placental Dysfunction: A Potential Target to Treat Preeclampsia? Front Immunol 2020; 10:3098. [PMID: 32010144 PMCID: PMC6974484 DOI: 10.3389/fimmu.2019.03098] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 12/18/2019] [Indexed: 12/16/2022] Open
Abstract
Preeclampsia is one of the leading causes of maternal and neonatal mortality and morbidity worldwide, affecting 2–8% of all pregnancies. Studies suggest a link between complement activation and preeclampsia. The complement system plays an essential role in the innate immunity, leading to opsonization, inflammation, and elimination of potential pathogens. The complement system also provides a link between innate and adaptive immunity and clearance of immune complexes and apoptotic cells. During pregnancy there is increased activity of the complement system systemically. However, locally at the placenta, complement inhibition is crucial for the maintenance of a normal pregnancy. Inappropriate or excessive activation of the complement system at the placenta is likely involved in placental dysfunction, and is in turn associated with pregnancy complications like preeclampsia. Therefore, modulation of the complement system could be a potential therapeutic target to prevent pregnancy complications such as preeclampsia. This review, based on a systematic literature search, gives an overview of the complement system and its activation locally in the placenta and systemically during healthy pregnancies and during complicated pregnancies, with a focus on preeclampsia. Furthermore, this review describes results of animal and human studies with a focus on the complement system in pregnancy, and the role of the complement system in placental dysfunction. Various clinical and animal studies provide evidence that dysregulation of the complement system is associated with placental dysfunction and therefore with preeclampsia. Several drugs are used for prevention and treatment of preeclampsia in humans and animal models, and some of these drugs work through complement modulation. Therefore, this review further discusses these studies examining pharmaceutical interventions as treatment for preeclampsia. These observations will help direct research to generate new target options for prevention and treatment of preeclampsia, which include direct and indirect modulation of the complement system.
Collapse
Affiliation(s)
- E Pierik
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Jelmer R Prins
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Harry van Goor
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Gustaaf A Dekker
- Department of Obstetrics and Gynecology, Lyell McEwin Hospital, University of Adelaide, Adelaide, SA, Australia
| | - Mohamed R Daha
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen and University of Groningen, Groningen, Netherlands
| | - Marc A J Seelen
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen and University of Groningen, Groningen, Netherlands
| | - Sicco A Scherjon
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
14
|
The role of hepatic sinusoidal obstruction in the pathogenesis of the hepatic involvement in HELLP syndrome: Exploring the literature. Pregnancy Hypertens 2019; 19:37-43. [PMID: 31877439 DOI: 10.1016/j.preghy.2019.11.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 10/11/2019] [Accepted: 11/25/2019] [Indexed: 01/17/2023]
Abstract
AIM This study aims to determine, based on existing data, whether the mechanism resulting in liver dysfunction in HELLP syndrome resembles that in Sinusoidal Obstruction Syndrome (SOS). BACKGROUND HELLP syndrome is a serious pregnancy disorder with high maternal and perinatal morbidity and mortality rates. Because of poor insight in its pathophysiology, particularly that of the liver involvement, clinical management is limited to symptomatic treatment, often followed by termination of pregnancy. SOS is a rare, potentially life-threatening complication of radio and/ or chemotherapy in the preparation of hematopoietic cell transplantation. The etiology of liver dysfunction in SOS is - unlike that in HELLP syndrome - better-understood and seems to be initiated by direct toxic damage and demise of endothelial cells, causing hepatic sinusoidal obstruction and ischemia. METHODS We searched Pubmed, Embase and Cochrane for reports on the etiology of HELLP and SOS. This yielded 73 articles, with 14 additional reports from the references listed in these articles. RESULTS The dysfunctional placenta in women developing HELLP initiates a cascade of events that eventually results in liver dysfunction. The placenta releases, besides anti-angiogenetic factors, also necrotic debris and cell-free DNA, a mixture that not only induces systemic endothelial dysfunction as in preeclampsia, but also a systemic inflammatory response. The latter aggravates the endothelio-toxic effects in the systemic cardiovascular bed, amplifying the already increased pro-thrombotic conditions. Particularly in microcirculations with extremely low shear forces, such as in the hepatic sinusoids, this will facilitate microthrombi formation and fibrin deposition eventually resulting in obstruction of the sinusoids similar as in SOS. The latter causes ischemic damage and progressive demise of hepatocytes. CONCLUSION The available information supports the concept that the liver damage in HELLP and SOS results from sinusoidal ischemia, presumably resulting from partially overlapping pathophysiological mechanisms.
Collapse
|
15
|
Tabacco S, Giannini A, Garufi C, Botta A, Salvi S, Del Sordo G, Benedetti Panici P, Lanzone A, De Carolis S. Complementemia in pregnancies with antiphospholipid syndrome. Lupus 2019; 28:1503-1509. [PMID: 31623520 DOI: 10.1177/0961203319882507] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Prognosis of pregnancies in women with antiphospholipid syndrome has dramatically improved over the past two decades using conventional treatment with low molecular weight heparin and low-dose aspirin. However, despite this regimen, 10-15% of antiphospholipid syndrome patients experience pregnancy losses. Several studies have been performed in order to identify risk factors predictive of complications. Thrombosis has been generally accepted as the key pathogenetic mechanism underlying pregnancy morbidity. However, the thrombogenic state alone is not able to explain all the different mechanisms leading to pregnancy failure. In fact, emerging evidence shows that complement pathway could play an important role in mediating clinical events in antiphospholipid syndrome. However, the exact mechanism through which complement mediates antiphospholipid syndrome complications remains unknown. Low complement levels (C3 and C4) are associated with poor pregnancy outcome in women with antiphospholipid syndrome in different studies. Hypocomplementemia could be indicated as an early predictor of adverse pregnancy outcome, available at the beginning of pregnancy for starting, if necessary, additional treatment to conventional therapy. However, future studies need to better understand the impact of low complement level on antiphospholipid syndrome pregnancy outcome.
Collapse
Affiliation(s)
- S Tabacco
- Department of Gynaecology Obstetrics and Urology, "Sapienza" University of Rome, Rome, Italy
| | - A Giannini
- Department of Gynaecology Obstetrics and Urology, "Sapienza" University of Rome, Rome, Italy
| | - C Garufi
- Lupus Clinic, "Sapienza" University of Rome, Rome, Italy
| | - A Botta
- UOC di Patologia Ostetrica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia
| | - S Salvi
- UOC di Patologia Ostetrica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia
| | - G Del Sordo
- Istituto di Clinica Ostetrica e Ginecologica, Università Cattolica del Sacro Cuore, Roma, Italia
| | - P Benedetti Panici
- Department of Gynaecology Obstetrics and Urology, "Sapienza" University of Rome, Rome, Italy
| | - A Lanzone
- UOC di Patologia Ostetrica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia.,Istituto di Clinica Ostetrica e Ginecologica, Università Cattolica del Sacro Cuore, Roma, Italia
| | - S De Carolis
- UOC di Patologia Ostetrica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia.,Istituto di Clinica Ostetrica e Ginecologica, Università Cattolica del Sacro Cuore, Roma, Italia
| |
Collapse
|
16
|
Abstract
Preeclampsia (PE) is a devastating adverse outcome of pregnancy. Characterized by maternal hypertension, PE, when left untreated, can result in death of both mother and baby. The cause of PE remains unknown, and there is no way to predict which women will develop PE during pregnancy. The only known treatment is delivery of both the fetus and placenta; therefore, an abnormal placenta is thought to play a causal role. Women with obesity before pregnancy have an increased chance of developing PE. Increased adiposity results in a heightened state of systemic inflammation that can influence placental development. Adipose tissue is a rich source of proinflammatory cytokines and complement proteins, which have been implicated in the pathogenesis of PE by promoting the expression of antiangiogenic factors in the mother. Because an aggravated inflammatory response, angiogenic imbalance, and abnormal placentation are observed in PE, we hypothesize that maternal obesity and complement proteins derived from adipose tissue play an important role in the development of PE.
Collapse
Affiliation(s)
- Kelsey N Olson
- Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University , Baton Rouge, Louisiana.,Reproductive Endocrinology & Women's Health Lab, Pennington Biomedical Research Center , Baton Rouge, Louisiana
| | - Leanne M Redman
- Reproductive Endocrinology & Women's Health Lab, Pennington Biomedical Research Center , Baton Rouge, Louisiana
| | - Jenny L Sones
- Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University , Baton Rouge, Louisiana
| |
Collapse
|
17
|
Than NG, Romero R, Tarca AL, Kekesi KA, Xu Y, Xu Z, Juhasz K, Bhatti G, Leavitt RJ, Gelencser Z, Palhalmi J, Chung TH, Gyorffy BA, Orosz L, Demeter A, Szecsi A, Hunyadi-Gulyas E, Darula Z, Simor A, Eder K, Szabo S, Topping V, El-Azzamy H, LaJeunesse C, Balogh A, Szalai G, Land S, Torok O, Dong Z, Kovalszky I, Falus A, Meiri H, Draghici S, Hassan SS, Chaiworapongsa T, Krispin M, Knöfler M, Erez O, Burton GJ, Kim CJ, Juhasz G, Papp Z. Integrated Systems Biology Approach Identifies Novel Maternal and Placental Pathways of Preeclampsia. Front Immunol 2018; 9:1661. [PMID: 30135684 PMCID: PMC6092567 DOI: 10.3389/fimmu.2018.01661] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 07/04/2018] [Indexed: 12/13/2022] Open
Abstract
Preeclampsia is a disease of the mother, fetus, and placenta, and the gaps in our understanding of the complex interactions among their respective disease pathways preclude successful treatment and prevention. The placenta has a key role in the pathogenesis of the terminal pathway characterized by exaggerated maternal systemic inflammation, generalized endothelial damage, hypertension, and proteinuria. This sine qua non of preeclampsia may be triggered by distinct underlying mechanisms that occur at early stages of pregnancy and induce different phenotypes. To gain insights into these molecular pathways, we employed a systems biology approach and integrated different "omics," clinical, placental, and functional data from patients with distinct phenotypes of preeclampsia. First trimester maternal blood proteomics uncovered an altered abundance of proteins of the renin-angiotensin and immune systems, complement, and coagulation cascades in patients with term or preterm preeclampsia. Moreover, first trimester maternal blood from preterm preeclamptic patients in vitro dysregulated trophoblastic gene expression. Placental transcriptomics of women with preterm preeclampsia identified distinct gene modules associated with maternal or fetal disease. Placental "virtual" liquid biopsy showed that the dysregulation of these disease gene modules originates during the first trimester. In vitro experiments on hub transcription factors of these gene modules demonstrated that DNA hypermethylation in the regulatory region of ZNF554 leads to gene down-regulation and impaired trophoblast invasion, while BCL6 and ARNT2 up-regulation sensitizes the trophoblast to ischemia, hallmarks of preterm preeclampsia. In summary, our data suggest that there are distinct maternal and placental disease pathways, and their interaction influences the clinical presentation of preeclampsia. The activation of maternal disease pathways can be detected in all phenotypes of preeclampsia earlier and upstream of placental dysfunction, not only downstream as described before, and distinct placental disease pathways are superimposed on these maternal pathways. This is a paradigm shift, which, in agreement with epidemiological studies, warrants for the central pathologic role of preexisting maternal diseases or perturbed maternal-fetal-placental immune interactions in preeclampsia. The description of these novel pathways in the "molecular phase" of preeclampsia and the identification of their hub molecules may enable timely molecular characterization of patients with distinct preeclampsia phenotypes.
Collapse
Affiliation(s)
- Nandor Gabor Than
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
- Maternity Private Department, Kutvolgyi Clinical Block, Semmelweis University, Budapest, Hungary
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Roberto Romero
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, United States
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, United States
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, United States
| | - Adi Laurentiu Tarca
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Computer Science, College of Engineering, Wayne State University, Detroit, MI, United States
| | - Katalin Adrienna Kekesi
- Laboratory of Proteomics, Department of Physiology and Neurobiology, ELTE Eotvos Lorand University, Budapest, Hungary
| | - Yi Xu
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States
| | - Zhonghui Xu
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard University, Boston, MA, United States
| | - Kata Juhasz
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Gaurav Bhatti
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States
| | | | - Zsolt Gelencser
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Janos Palhalmi
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | | | - Balazs Andras Gyorffy
- Laboratory of Proteomics, Department of Physiology and Neurobiology, ELTE Eotvos Lorand University, Budapest, Hungary
| | - Laszlo Orosz
- Department of Obstetrics and Gynaecology, University of Debrecen, Debrecen, Hungary
| | - Amanda Demeter
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Anett Szecsi
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Eva Hunyadi-Gulyas
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Zsuzsanna Darula
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Attila Simor
- Laboratory of Proteomics, Department of Physiology and Neurobiology, ELTE Eotvos Lorand University, Budapest, Hungary
| | - Katalin Eder
- Department of Genetics, Cell and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Szilvia Szabo
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
- Department of Morphology and Physiology, Semmelweis University, Budapest, Hungary
| | - Vanessa Topping
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States
| | - Haidy El-Azzamy
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States
| | - Christopher LaJeunesse
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States
| | - Andrea Balogh
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Gabor Szalai
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States
- Systems Biology of Reproduction Lendulet Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Susan Land
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, United States
| | - Olga Torok
- Department of Obstetrics and Gynaecology, University of Debrecen, Debrecen, Hungary
| | - Zhong Dong
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States
| | - Ilona Kovalszky
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Andras Falus
- Department of Genetics, Cell and Immunobiology, Semmelweis University, Budapest, Hungary
| | | | - Sorin Draghici
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, United States
- Department of Clinical and Translational Science, Wayne State University, Detroit, MI, United States
| | - Sonia S. Hassan
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
| | | | - Martin Knöfler
- Department of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
| | - Offer Erez
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Obstetrics and Gynecology, Soroka University Medical Center School of Medicine, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Graham J. Burton
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Chong Jai Kim
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, United States
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Detroit, MI, United States
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Pathology, Asan Medical Center, University of Ulsan, Seoul, South Korea
| | - Gabor Juhasz
- Laboratory of Proteomics, Department of Physiology and Neurobiology, ELTE Eotvos Lorand University, Budapest, Hungary
| | - Zoltan Papp
- Maternity Private Department, Kutvolgyi Clinical Block, Semmelweis University, Budapest, Hungary
| |
Collapse
|
18
|
Sicre de Fontbrune F, Peffault de Latour R. Ten Years of Clinical Experience With Eculizumab in Patients With Paroxysmal Nocturnal Hemoglobinuria. Semin Hematol 2018; 55:124-129. [DOI: 10.1053/j.seminhematol.2018.04.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 04/03/2018] [Indexed: 12/12/2022]
|
19
|
Palomino WA, Tayade C, Argandoña F, Devoto L, Young SL, Lessey BA. The endometria of women with endometriosis exhibit dysfunctional expression of complement regulatory proteins during the mid secretory phase. J Reprod Immunol 2018; 125:1-7. [DOI: 10.1016/j.jri.2017.10.046] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Revised: 08/21/2017] [Accepted: 10/23/2017] [Indexed: 01/19/2023]
|
20
|
Kenny LC, Kell DB. Immunological Tolerance, Pregnancy, and Preeclampsia: The Roles of Semen Microbes and the Father. Front Med (Lausanne) 2018; 4:239. [PMID: 29354635 PMCID: PMC5758600 DOI: 10.3389/fmed.2017.00239] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 12/12/2017] [Indexed: 12/18/2022] Open
Abstract
Although it is widely considered, in many cases, to involve two separable stages (poor placentation followed by oxidative stress/inflammation), the precise originating causes of preeclampsia (PE) remain elusive. We have previously brought together some of the considerable evidence that a (dormant) microbial component is commonly a significant part of its etiology. However, apart from recognizing, consistent with this view, that the many inflammatory markers of PE are also increased in infection, we had little to say about immunity, whether innate or adaptive. In addition, we focused on the gut, oral and female urinary tract microbiomes as the main sources of the infection. We here marshall further evidence for an infectious component in PE, focusing on the immunological tolerance characteristic of pregnancy, and the well-established fact that increased exposure to the father's semen assists this immunological tolerance. As well as these benefits, however, semen is not sterile, microbial tolerance mechanisms may exist, and we also review the evidence that semen may be responsible for inoculating the developing conceptus (and maybe the placenta) with microbes, not all of which are benign. It is suggested that when they are not, this may be a significant cause of PE. A variety of epidemiological and other evidence is entirely consistent with this, not least correlations between semen infection, infertility and PE. Our view also leads to a series of other, testable predictions. Overall, we argue for a significant paternal role in the development of PE through microbial infection of the mother via insemination.
Collapse
Affiliation(s)
- Louise C. Kenny
- The Irish Centre for Fetal and Neonatal Translational Research (INFANT), University College Cork, Cork, Ireland
- Department of Obstetrics and Gynecology, University College Cork, Cork, Ireland
- Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Douglas B. Kell
- School of Chemistry, The University of Manchester, Manchester, United Kingdom
- The Manchester Institute of Biotechnology, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
21
|
Characterization of an anti-fetal AChR monoclonal antibody isolated from a myasthenia gravis patient. Sci Rep 2017; 7:14426. [PMID: 29089519 PMCID: PMC5663942 DOI: 10.1038/s41598-017-14350-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 10/10/2017] [Indexed: 11/24/2022] Open
Abstract
We report here the sequence and functional characterization of a recombinantly expressed autoantibody (mAb 131) previously isolated from a myasthenia gravis patient by immortalization of thymic B cells using Epstein-Barr virus and TLR9 activation. The antibody is characterized by a high degree of somatic mutations as well as a 6 amino acid insertion within the VHCDR2. The recombinant mAb 131 is specific for the γ-subunit of the fetal AChR to which it bound with sub-nanomolar apparent affinity, and detected the presence of fetal AChR on a number of rhabdomyosarcoma cell lines. Mab 131 blocked one of the two α-bungarotoxin binding sites on the fetal AChR, and partially blocked the binding of an antibody (mAb 637) to the α-subunit of the AChR, suggesting that both antibodies bind at or near one ACh binding site at the α/γ subunit interface. However, mAb 131 did not reduce fetal AChR ion channel currents in electrophysiological experiments. These results indicate that mAb 131, although generated from an MG patient, is unlikely to be pathogenic and may make it a potentially useful reagent for studies of myasthenia gravis, rhabdomyosarcoma and arthrogryposis multiplex congenita which can be caused by fetal-specific AChR-blocking autoantibodies.
Collapse
|
22
|
Activated NK cells cause placental dysfunction and miscarriages in fetal alloimmune thrombocytopenia. Nat Commun 2017; 8:224. [PMID: 28794456 PMCID: PMC5550461 DOI: 10.1038/s41467-017-00269-1] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 06/16/2017] [Indexed: 01/29/2023] Open
Abstract
Miscarriage and intrauterine growth restriction (IUGR) are devastating complications in fetal/neonatal alloimmune thrombocytopenia (FNAIT). We previously reported the mechanisms for bleeding diatheses, but it is unknown whether placental, decidual immune cells or other abnormalities at the maternal-fetal interface contribute to FNAIT. Here we show that maternal immune responses to fetal platelet antigens cause miscarriage and IUGR that are associated with vascular and immune pathologies in murine FNAIT models. Uterine natural killer (uNK) cell recruitment and survival beyond mid-gestation lead to elevated NKp46 and CD107 expression, perforin release and trophoblast apoptosis. Depletion of NK cells restores normal spiral artery remodeling and placental function, prevents miscarriage, and rescues hemorrhage in neonates. Blockade of NK activation receptors (NKp46, FcɣRIIIa) also rescues pregnancy loss. These findings shed light on uNK antibody-dependent cell-mediated cytotoxicity of invasive trophoblasts as a pathological mechanism in FNAIT, and suggest that anti-NK cell therapies may prevent immune-mediated pregnancy loss and ameliorate FNAIT.Fetal/neonatal alloimmune thrombocytopenia (FNAIT) is a gestational disease caused by maternal immune responses against fetal platelets. Using a FNAIT mouse model and human trophoblast cell lines, here the authors show that uterine natural killer cell-mediated trophoblast apoptosis contributes to FNAIT pathogenesis.
Collapse
|
23
|
Abheiden CNH, Blomjous BS, Kroese SJ, Bultink IEM, Fritsch-Stork RDE, Lely AT, de Boer MA, de Vries JIP. Low-molecular-weight heparin and aspirin use in relation to pregnancy outcome in women with systemic lupus erythematosus and antiphospholipid syndrome: A cohort study. Hypertens Pregnancy 2016; 36:8-15. [PMID: 27599157 DOI: 10.1080/10641955.2016.1217337] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE To relate anticoagulant use to pregnancy complications in women with systemic lupus erythematosus (SLE) and primary antiphospholipid syndrome (APS). METHODS All ongoing pregnancies, 184, in two Dutch tertiary centers between 2000 and 2015. RESULTS LMWH and aspirin was prescribed in 15/109 SLE women without antiphospholipid antibodies (aPL), 5/14 with aPL, 11/13 with APS, 45/48 with primary APS. Main complications in the four treatment groups (no anticoagulant treatment, aspirin, LMWH, aspirin and LMWH) included hypertensive disorders of pregnancy (9.4%, 23.3%, 50%, 18.4%, respectively, p = 0.12) and preterm birth (16.7%, 34.3%, 75%, 36.8%, respectively, p < 0.001). CONCLUSION Maternal and perinatal complications occurred frequently, despite LMWH and aspirin use.
Collapse
Affiliation(s)
- Carolien N H Abheiden
- a Department of Obstetrics and Gynaecology , VU University Medical Center , Amsterdam , The Netherlands
| | - Birgit S Blomjous
- a Department of Obstetrics and Gynaecology , VU University Medical Center , Amsterdam , The Netherlands.,b Department of Rheumatology , Amsterdam Rheumatology and Immunology Center, VU University Medical Center , Amsterdam , The Netherlands
| | - Sylvia J Kroese
- c Department of Rheumatology , University Medical Center Utrecht , Utrecht , The Netherlands
| | - Irene E M Bultink
- b Department of Rheumatology , Amsterdam Rheumatology and Immunology Center, VU University Medical Center , Amsterdam , The Netherlands
| | - Ruth D E Fritsch-Stork
- c Department of Rheumatology , University Medical Center Utrecht , Utrecht , The Netherlands
| | - A Titia Lely
- d Department of Obstetrics and Gynaecology , University Medical Center Utrecht , Utrecht , The Netherlands
| | - Marjon A de Boer
- a Department of Obstetrics and Gynaecology , VU University Medical Center , Amsterdam , The Netherlands
| | - Johanna I P de Vries
- a Department of Obstetrics and Gynaecology , VU University Medical Center , Amsterdam , The Netherlands
| |
Collapse
|
24
|
Ribeiro ES, Greco LF, Bisinotto RS, Lima FS, Thatcher WW, Santos JE. Biology of Preimplantation Conceptus at the Onset of Elongation in Dairy Cows1. Biol Reprod 2016; 94:97. [DOI: 10.1095/biolreprod.115.134908] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 02/25/2016] [Indexed: 01/11/2023] Open
|
25
|
Girardi G. MRI-based methods to detect placental and fetal brain abnormalities in utero. J Reprod Immunol 2016; 114:86-91. [DOI: 10.1016/j.jri.2015.05.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 05/21/2015] [Accepted: 05/29/2015] [Indexed: 11/29/2022]
|
26
|
Serum Adipsin Levels throughout Normal Pregnancy and Preeclampsia. Sci Rep 2016; 6:20073. [PMID: 26832661 PMCID: PMC4735521 DOI: 10.1038/srep20073] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 12/18/2015] [Indexed: 01/08/2023] Open
Abstract
Adipsin is a protease produced at high levels by adipose tissue. It is involved in complement activation and metabolic control. The objective of this study was to determine the changes in adipsin levels during different stages of normal pregnancy, and its association with obstetric outcomes, such as preeclampsia. This nested case-control study in a longitudinal cohort included normal pregnant (n = 54) and preeclamptic (n = 18) women, both followed throughout pregnancy. Additionally, some of the normal pregnant women were followed up three months postpartum (n = 18). Healthy non-pregnant women were also studied during their menstrual cycle (n = 20). The results of this study show that in healthy non-pregnant women, adipsin levels did not change significantly during the menstrual cycle. In normal pregnant women, adipsin levels were lower (p < 0.01) when compared with non-pregnant healthy women, but these serum levels increased again during postpartum (p < 0.001). Adipsin levels were significantly elevated in preeclamptic women in late pregnancy (P < 0.01). A significant correlation was not found between leptin and adipsin during the three periods of gestation studied in healthy pregnant and preeclamptic women. Our results suggest that adipsin may be involved in pregnancy-associated metabolic changes. Moreover, the increase of adipsin levels towards late gestation in preeclamptic women could be related to the pathophysiology of this disease.
Collapse
|
27
|
Petitbarat M, Durigutto P, Macor P, Bulla R, Palmioli A, Bernardi A, De Simoni MG, Ledee N, Chaouat G, Tedesco F. Critical Role and Therapeutic Control of the Lectin Pathway of Complement Activation in an Abortion-Prone Mouse Mating. THE JOURNAL OF IMMUNOLOGY 2015; 195:5602-7. [DOI: 10.4049/jimmunol.1501361] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 10/14/2015] [Indexed: 11/19/2022]
|
28
|
Veglia M, D'Ippolito S, Marana R, Di Nicuolo F, Castellani R, Bruno V, Fiorelli A, Ria F, Maulucci G, De Spirito M, Migliara G, Scambia G, Di Simone N. Human IgG Antinuclear Antibodies Induce Pregnancy Loss in Mice by Increasing Immune Complex Deposition in Placental Tissue: In Vivo Study. Am J Reprod Immunol 2015; 74:542-52. [PMID: 26388133 DOI: 10.1111/aji.12429] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 08/24/2015] [Indexed: 12/18/2022] Open
Abstract
PROBLEM A threefold higher prevalence of antinuclear antibodies (ANA) has been reported in patients with recurrent pregnancy loss (RPL). Nevertheless, the role of ANA in reproductive failure is still unclear. The aim of this study was to investigate the role of ANA during early pregnancy in vivo. METHOD OF STUDY We used pregnant mice treated with immunoglobulin G (IgG) obtained from normal healthy subjects (NHS); ANA(+) sera of patients with RPL; and ANA(+) sera from women with uncomplicated pregnancies (HW). Placental immunohistochemical/immunofluorescence staining was performed to detect complement and immune complex deposition. ELISA was performed to evaluate complement levels. RESULTS ANA(+) IgG from RPL women significantly increased embryo resorption rate, reduced C3, and increased C3a serum levels compared to NHS IgG or ANA(+) -HW IgG. Increased C3 deposition and increased immune complex staining in placental tissues from mice treated with ANA(+) -RPL IgG fraction compared to NHS- and ANA(+) -HW-IgG-treated mice were found. CONCLUSION ANA(+) IgG injection in mice is able to induce fetal resorption and complement activation. The presence on placental tissues of immune complexes and complement fragments suggests the complement activation as a possible mechanism of placental damage.
Collapse
Affiliation(s)
- Manuela Veglia
- Department of Obstetrics and Gynecology, Policlinico A. Gemelli, Università Cattolica Del Sacro Cuore, Rome, Italy
| | - Silvia D'Ippolito
- Department of Obstetrics and Gynecology, Policlinico A. Gemelli, Università Cattolica Del Sacro Cuore, Rome, Italy
| | - Riccardo Marana
- Department of Obstetrics and Gynecology, Policlinico A. Gemelli, Università Cattolica Del Sacro Cuore, Rome, Italy.,International Scientific Institute Paolo VI, ISI, Università Cattolica Del Sacro Cuore, Policlinico A. Gemelli, Rome, Italy
| | - Fiorella Di Nicuolo
- Department of Obstetrics and Gynecology, Policlinico A. Gemelli, Università Cattolica Del Sacro Cuore, Rome, Italy
| | - Roberta Castellani
- Department of Obstetrics and Gynecology, Policlinico A. Gemelli, Università Cattolica Del Sacro Cuore, Rome, Italy
| | - Valentina Bruno
- Section of Gynecology and Obstetrics, Department of Surgical Sciences, Università di Tor Vergata, Rome, Italy
| | - Alessia Fiorelli
- Department of Obstetrics and Gynecology, Policlinico A. Gemelli, Università Cattolica Del Sacro Cuore, Rome, Italy
| | - Francesco Ria
- Institute of General Pathology, Università Cattolica Del Sacro Cuore, Policlinico A. Gemelli, Rome, Italy
| | - Giuseppe Maulucci
- Institute of Physics, Università Cattolica Del Sacro Cuore, Rome, Italy
| | - Marco De Spirito
- Institute of Physics, Università Cattolica Del Sacro Cuore, Rome, Italy
| | - Giuseppe Migliara
- Institute of General Pathology, Università Cattolica Del Sacro Cuore, Policlinico A. Gemelli, Rome, Italy
| | - Giovanni Scambia
- Department of Obstetrics and Gynecology, Policlinico A. Gemelli, Università Cattolica Del Sacro Cuore, Rome, Italy
| | - Nicoletta Di Simone
- Department of Obstetrics and Gynecology, Policlinico A. Gemelli, Università Cattolica Del Sacro Cuore, Rome, Italy
| |
Collapse
|
29
|
Kelly RJ, Höchsmann B, Szer J, Kulasekararaj A, de Guibert S, Röth A, Weitz IC, Armstrong E, Risitano AM, Patriquin CJ, Terriou L, Muus P, Hill A, Turner MP, Schrezenmeier H, Peffault de Latour R. Eculizumab in Pregnant Patients with Paroxysmal Nocturnal Hemoglobinuria. N Engl J Med 2015; 373:1032-9. [PMID: 26352814 DOI: 10.1056/nejmoa1502950] [Citation(s) in RCA: 190] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Eculizumab, a humanized monoclonal antibody against complement protein C5 that inhibits terminal complement activation, has been shown to prevent complications of paroxysmal nocturnal hemoglobinuria (PNH) and improve quality of life and overall survival, but data on the use of eculizumab in women during pregnancy are scarce. METHODS We designed a questionnaire to solicit data on pregnancies in women with PNH and sent it to the members of the International PNH Interest Group and to the physicians participating in the International PNH Registry. We assessed the safety and efficacy of eculizumab in pregnant patients with PNH by examining the birth and developmental records of the children born and adverse events in the mothers. RESULTS Of the 94 questionnaires that were sent out, 75 were returned, representing a response rate of 80%. Data on 75 pregnancies in 61 women with PNH were evaluated. There were no maternal deaths and three fetal deaths (4%). Six miscarriages (8%) occurred during the first trimester. Requirements for transfusion of red cells increased during pregnancy, from a mean of 0.14 units per month in the 6 months before pregnancy to 0.92 units per month during pregnancy. Platelet transfusions were given in 16 pregnancies. In 54% of pregnancies that progressed past the first trimester, the dose or the frequency of use of eculizumab had to be increased. Low-molecular-weight heparin was used in 88% of the pregnancies. Ten hemorrhagic events and 2 thrombotic events were documented; both thrombotic events occurred during the postpartum period. A total of 22 births (29%) were premature. Twenty cord-blood samples were examined for the presence of eculizumab; the drug was detected in 7 of the samples. A total of 25 babies were breast-fed, and in 10 of these cases, breast milk was examined for the presence of eculizumab; the drug was not detected in any of the 10 breast-milk samples. CONCLUSIONS Eculizumab provided benefit for women with PNH during pregnancy, as evidenced by a high rate of fetal survival and a low rate of maternal complications. (ClinicalTrials.gov number, NCT01374360.).
Collapse
Affiliation(s)
- Richard J Kelly
- From Department of Haematology, St. James's University Hospital, Leeds (R.J.K., A.H.), and Haematological Medicine, Kings College Hospital, London (A.K.) - both in the United Kingdom; University of Ulm and German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen, Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, Ulm (B.H., H.S.), and Department of Hematology, University Hospital of Essen, Essen (A.R.) - both in Germany; PNH Subcommittee of the Severe Aplastic Anemia Working Party of the European Group for Blood and Marrow Transplantation, Leiden (B.H., A.K., A.M.R., L.T., H.S., R.P.L.), and Radboud University Medical Center, Nijmegen (P.M.) - both in the Netherlands; Department of Clinical Haematology, the Royal Melbourne Hospital and University of Melbourne, Parkville, VIC, Australia (J.S.); Department of Hematology, University Hospital, Rennes (S.G.), Clinical Immunology, Department of Internal Medicine, University Hospital of Lille, Lille (L.T.), and Assistance Publique-Hôpitaux de Paris, Service d'Hématologie-Greffe, Hôpital Saint-Louis, University Paris Diderot, Sorbonne Paris Cité, Centre de Référence Aplasie Médullaire-HPN, and Filière de Santé Maladie Rare Immuno-Hématologique (MARIH), Paris (R.P.L.) - all in France; Jane Anne Nohl Division of Hematology, Keck School of Medicine of University of Southern California, Los Angeles (I.C.W.); Department of Hematology, Helsinki University Central Hospital, Helsinki (E.A.); Hematology, Department of Clinical Medicine and Surgery, Federico II, University of Naples, Naples, Italy (A.M.R.); Hematology and Thromboembolism, McMaster University, Hamilton, ON, Canada (C.J.P.); and ICON Clinical Research, San Francisco (M.P.T.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Girardi G, Fraser J, Lennen R, Vontell R, Jansen M, Hutchison G. Imaging of activated complement using ultrasmall superparamagnetic iron oxide particles (USPIO)--conjugated vectors: an in vivo in utero non-invasive method to predict placental insufficiency and abnormal fetal brain development. Mol Psychiatry 2015; 20:1017-26. [PMID: 25245499 PMCID: PMC4288949 DOI: 10.1038/mp.2014.110] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 07/14/2014] [Accepted: 07/21/2014] [Indexed: 01/02/2023]
Abstract
In the current study, we have developed a magnetic resonance imaging-based method for non-invasive detection of complement activation in placenta and foetal brain in vivo in utero. Using this method, we found that anti-complement C3-targeted ultrasmall superparamagnetic iron oxide (USPIO) nanoparticles bind within the inflamed placenta and foetal brain cortical tissue, causing a shortening of the T2* relaxation time. We used two mouse models of pregnancy complications: a mouse model of obstetrics antiphospholipid syndrome (APS) and a mouse model of preterm birth (PTB). We found that detection of C3 deposition in the placenta in the APS model was associated with placental insufficiency characterised by increased oxidative stress, decreased vascular endothelial growth factor and placental growth factor levels and intrauterine growth restriction. We also found that foetal brain C3 deposition was associated with cortical axonal cytoarchitecture disruption and increased neurodegeneration in the mouse model of APS and in the PTB model. In the APS model, foetuses that showed increased C3 in their brains additionally expressed anxiety-related behaviour after birth. Importantly, USPIO did not affect pregnancy outcomes and liver function in the mother and the offspring, suggesting that this method may be useful for detecting complement activation in vivo in utero and predicting placental insufficiency and abnormal foetal neurodevelopment that leads to neuropsychiatric disorders.
Collapse
Affiliation(s)
- G Girardi
- MRC Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK,Lupus Research Unit, The Rayne Institute, King's College London St Thomas' Hospital, London, UK,Women's Health, King's College London, St Thomas' Hospital, London SE1 7EH, UK
| | - J Fraser
- Centre for Nano Safety, Napier University Edinburgh, Edinburgh, UK
| | - R Lennen
- BHF/University Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - R Vontell
- Centrer for the Developing Brain, Division of Imaging Sciences and Biomedical Engineering, The Rayne Institute, King's College London, St Thomas' Hospital, London, UK
| | - M Jansen
- BHF/University Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - G Hutchison
- Centre for Nano Safety, Napier University Edinburgh, Edinburgh, UK
| |
Collapse
|
31
|
Regiani T, Cordeiro FB, da Costa LDVT, Salgueiro J, Cardozo K, Carvalho VM, Perkel KJ, Zylbersztejn DS, Cedenho AP, Lo Turco EG. Follicular fluid alterations in endometriosis: label-free proteomics by MS(E) as a functional tool for endometriosis. Syst Biol Reprod Med 2015; 61:263-76. [PMID: 26114977 DOI: 10.3109/19396368.2015.1037025] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Endometriosis is a chronic gynecological condition that affects 10-32% of women of reproductive age and may lead to infertility. The study of protein profiles in follicular fluid may assist in elucidating possible biomarkers related to this disease. For this, follicular fluid samples were obtained from women with tubal factor or minimal male factor infertility who had pregnancy outcomes after in vitro fertilization (IVF) treatment (control group, n = 10), women with endometriosis (endometriosis group, n = 10), along with the endometrioma from these same patients were included (endometrioma group, n = 10). For proteomic analysis, samples were pooled according to their respective groups and normalized to protein content. Proteins were analyzed by in tandem mass spectrometry (MS(E)) Spectra processing and the ProteinLynx Global Server v.2.5. was used for database searching. Data was submitted to the biological network analysis using Cytoscape 2.8.2 with ClueGO plugin. As a result, 535 proteins were identified among all groups. The control group differentially or uniquely expressed 33 (6%) proteins and equal expression of 98 (18%) proteins was observed in the control and endometriosis groups of which 41 (7%) proteins were further identified and/or quantified. Six (1%) proteins were observed in both the endometriosis and endometrioma groups, but 212 (39%) proteins were exclusively identified and/or quantified in the endometrioma group. There were 9 (1%) proteins observed in both the control and endometrioma groups and there were 139 (25%) proteins common among all three groups. Distinct differences among the protein profiles in the follicular fluid of patients included in this study were found, identifying proteins related to the disease progression and IVF success. Thus, some pathways related to endometriosis are associated with the presence of specific proteins, as well as the absence of others. This study provides a first step to the development of more sensitive diagnostic tests and treatment.
Collapse
Affiliation(s)
- Thais Regiani
- a Department of Surgery, Division of Urology , Human Reproduction Section, Sao Paulo Federal University , Sao Paulo , Brazil
| | - Fernanda Bertuccez Cordeiro
- a Department of Surgery, Division of Urology , Human Reproduction Section, Sao Paulo Federal University , Sao Paulo , Brazil
| | - Lívia do Vale Teixeira da Costa
- a Department of Surgery, Division of Urology , Human Reproduction Section, Sao Paulo Federal University , Sao Paulo , Brazil
| | | | | | | | - Kayla Jane Perkel
- c Department of Biomedical Sciences , Ontario Veterinary College, University of Guelph , Guelph , ON , Canada
| | - Daniel Suslik Zylbersztejn
- a Department of Surgery, Division of Urology , Human Reproduction Section, Sao Paulo Federal University , Sao Paulo , Brazil
| | - Agnaldo Pereira Cedenho
- a Department of Surgery, Division of Urology , Human Reproduction Section, Sao Paulo Federal University , Sao Paulo , Brazil
| | - Edson Guimarães Lo Turco
- a Department of Surgery, Division of Urology , Human Reproduction Section, Sao Paulo Federal University , Sao Paulo , Brazil
| |
Collapse
|
32
|
Song D, Yu XJ, Wang FM, Xu BN, He YD, Chen Q, Wang SX, Yu F, Song WC, Zhao MH. Overactivation of Complement Alternative Pathway in Postpartum Atypical Hemolytic Uremic Syndrome Patients with Renal Involvement. Am J Reprod Immunol 2015; 74:345-56. [PMID: 26011580 DOI: 10.1111/aji.12404] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Accepted: 05/06/2015] [Indexed: 12/17/2022] Open
Affiliation(s)
- Di Song
- Renal Division; Department of Medicine; Peking University First Hospital; Beijing China
- Peking University Institute of Nephrology; Beijing China
- Key laboratory of Renal Disease; Ministry of Health of China; Beijing China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment; Ministry of Education of China; Beijing China
| | - Xiao-juan Yu
- Renal Division; Department of Medicine; Peking University First Hospital; Beijing China
- Peking University Institute of Nephrology; Beijing China
- Key laboratory of Renal Disease; Ministry of Health of China; Beijing China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment; Ministry of Education of China; Beijing China
| | - Feng-mei Wang
- Renal Division; Department of Medicine; Peking University First Hospital; Beijing China
- Peking University Institute of Nephrology; Beijing China
- Key laboratory of Renal Disease; Ministry of Health of China; Beijing China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment; Ministry of Education of China; Beijing China
| | - Bing-ning Xu
- Department of Obstetrics and Gynecology; Peking University First Hospital; Beijing China
| | - Ying-dong He
- Department of Obstetrics and Gynecology; Peking University First Hospital; Beijing China
| | - Qian Chen
- Department of Obstetrics and Gynecology; Peking University First Hospital; Beijing China
| | - Su-xia Wang
- Renal Division; Department of Medicine; Peking University First Hospital; Beijing China
- Peking University Institute of Nephrology; Beijing China
- Key laboratory of Renal Disease; Ministry of Health of China; Beijing China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment; Ministry of Education of China; Beijing China
| | - Feng Yu
- Renal Division; Department of Medicine; Peking University First Hospital; Beijing China
- Peking University Institute of Nephrology; Beijing China
- Key laboratory of Renal Disease; Ministry of Health of China; Beijing China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment; Ministry of Education of China; Beijing China
| | - Wen-chao Song
- Institute for Translational Medicine and Therapeutics and Department of Pharmacology; Perelman School of Medicine; University of Pennsylvania; Philadelphia PA USA
| | - Ming-hui Zhao
- Renal Division; Department of Medicine; Peking University First Hospital; Beijing China
- Peking University Institute of Nephrology; Beijing China
- Key laboratory of Renal Disease; Ministry of Health of China; Beijing China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment; Ministry of Education of China; Beijing China
- Peking-Tsinghua Center for Life Sciences; Beijing China
| |
Collapse
|
33
|
Szalai G, Romero R, Chaiworapongsa T, Xu Y, Wang B, Ahn H, Xu Z, Chiang PJ, Sundell B, Wang R, Jiang Y, Plazyo O, Olive M, Tarca AL, Dong Z, Qureshi F, Papp Z, Hassan SS, Hernandez-Andrade E, Than NG. Full-length human placental sFlt-1-e15a isoform induces distinct maternal phenotypes of preeclampsia in mice. PLoS One 2015; 10:e0119547. [PMID: 25860260 PMCID: PMC4393117 DOI: 10.1371/journal.pone.0119547] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 01/30/2015] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Most anti-angiogenic preeclampsia models in rodents utilized the overexpression of a truncated soluble fms-like tyrosine kinase-1 (sFlt-1) not expressed in any species. Other limitations of mouse preeclampsia models included stressful blood pressure measurements and the lack of postpartum monitoring. We aimed to 1) develop a mouse model of preeclampsia by administering the most abundant human placental sFlt-1 isoform (hsFlt-1-e15a) in preeclampsia; 2) determine blood pressures in non-stressed conditions; and 3) develop a survival surgery that enables the collection of fetuses and placentas and postpartum (PP) monitoring. METHODS Pregnancy status of CD-1 mice was evaluated with high-frequency ultrasound on gestational days (GD) 6 and 7. Telemetry catheters were implanted in the carotid artery on GD7, and their positions were verified by ultrasound on GD13. Mice were injected through tail-vein with adenoviruses expressing hsFlt-1-e15a (n = 11) or green fluorescent protein (GFP; n = 9) on GD8/GD11. Placentas and pups were delivered by cesarean section on GD18 allowing PP monitoring. Urine samples were collected with cystocentesis on GD6/GD7, GD13, GD18, and PPD8, and albumin/creatinine ratios were determined. GFP and hsFlt-1-e15a expression profiles were determined by qRT-PCR. Aortic ring assays were performed to assess the effect of hsFlt-1-e15a on endothelia. RESULTS Ultrasound predicted pregnancy on GD7 in 97% of cases. Cesarean section survival rate was 100%. Mean arterial blood pressure was higher in hsFlt-1-e15a-treated than in GFP-treated mice (∆MAP = 13.2 mmHg, p = 0.00107; GD18). Focal glomerular changes were found in hsFlt-1-e15a -treated mice, which had higher urine albumin/creatinine ratios than controls (109.3 ± 51.7 μg/mg vs. 19.3 ± 5.6 μg/mg, p = 4.4 x 10(-2); GD18). Aortic ring assays showed a 46% lesser microvessel outgrowth in hsFlt-1-e15a-treated than in GFP-treated mice (p = 1.2 x 10(-2)). Placental and fetal weights did not differ between the groups. One mouse with liver disease developed early-onset preeclampsia-like symptoms with intrauterine growth restriction (IUGR). CONCLUSIONS A mouse model of late-onset preeclampsia was developed with the overexpression of hsFlt-1-e15a, verifying the in vivo pathologic effects of this primate-specific, predominant placental sFlt-1 isoform. HsFlt-1-e15a induced early-onset preeclampsia-like symptoms associated with IUGR in a mouse with a liver disease. Our findings support that hsFlt-1-e15a is central to the terminal pathway of preeclampsia, and it can induce the full spectrum of symptoms in this obstetrical syndrome.
Collapse
Affiliation(s)
- Gabor Szalai
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
| | - Roberto Romero
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Yi Xu
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
| | - Bing Wang
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
| | - Hyunyoung Ahn
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
| | - Zhonghui Xu
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
| | - Po Jen Chiang
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
| | - Birgitta Sundell
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
| | - Rona Wang
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
| | - Yang Jiang
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
| | - Olesya Plazyo
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
| | - Mary Olive
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Adi L. Tarca
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
- Department of Computer Science, Wayne State University, Detroit, Michigan, United States of America
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Zhong Dong
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
| | - Faisal Qureshi
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Zoltan Papp
- Maternity Private Department, Kutvolgyi Clinical Block, Semmelweis University, Budapest, Hungary
| | - Sonia S. Hassan
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Edgar Hernandez-Andrade
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Nandor Gabor Than
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, United States of America
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Maternity Private Department, Kutvolgyi Clinical Block, Semmelweis University, Budapest, Hungary
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
- First Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| |
Collapse
|
34
|
Lashley LEELO, Buurma A, Swings GMJS, Eikmans M, Anholts JDH, Bakker JA, Claas FHJ. Preeclampsia in autologous and oocyte donation pregnancy: is there a different pathophysiology? J Reprod Immunol 2015; 109:17-23. [PMID: 25863695 DOI: 10.1016/j.jri.2015.03.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 02/20/2015] [Accepted: 03/03/2015] [Indexed: 12/25/2022]
Abstract
Oocyte donation (OD) is a specific method of artificial reproductive technology that is accompanied by a higher risk of preeclampsia during pregnancy. The pathophysiological mechanism underlying preeclampsia in OD pregnancies is thought to differ from preeclampsia in autologous pregnancies. As preeclampsia in autologous pregnancies is suggested to be associated with complement activation, we studied C4d deposition, circulating complement components and placental complement regulatory proteins in preeclamptic OD pregnancies. Women with uncomplicated and preeclamptic pregnancies after OD or spontaneous conception were selected. We stained the placentas for C4d, marker for complement activation, measured complement factors C1q, C3 and C4 in maternal sera and quantified the placental mRNA expression of complement regulatory proteins CD46, CD55 and CD59. A significantly (p < 0.03) higher incidence of C4d deposition was observed in placentas from women with preeclampsia compared with uncomplicated pregnancies, both OD and autologous. The level of complement factors in serum did not differ between the groups. Children born in the autologous preeclampsia group were significantly lower in birth weight (p < 10th percentile) compared with the preeclamptic OD group. In addition, the placental mRNA expression level of complement regulatory proteins was significantly lower in uncomplicated and preeclamptic OD compared with the autologous pregnancies. In line with autologous preeclampsia pregnancies, there is excessive activation of complement in preeclamptic OD pregnancies. However, in contrast to autologous pregnancies this is not associated with counterbalancing upregulation of complement regulatory proteins. Furthermore, C4d deposition in OD pregnancies is not related to the severity of preeclampsia, suggesting another trigger or regulatory mechanism of placental C4d deposition in preeclamptic OD pregnancies.
Collapse
Affiliation(s)
- Lisa E E L O Lashley
- Department of Gynecology and Obstetrics, Leiden University Medical Centre, The Netherlands.
| | - Aletta Buurma
- Department of Pathology, Leiden University Medical Centre, The Netherlands
| | - Godelieve M J S Swings
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Centre, The Netherlands
| | - Michael Eikmans
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Centre, The Netherlands
| | - Jacqueline D H Anholts
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Centre, The Netherlands
| | - Jaap A Bakker
- Department of Clinical Chemistry and Laboratory Medicine, Leiden University Medical Centre, The Netherlands
| | - Frans H J Claas
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Centre, The Netherlands
| |
Collapse
|
35
|
Velickovic I, Dalloul M, Wong KA, Bakare O, Schweis F, Garala M, Alam A, Medranda G, Lekovic J, Shuaib W, Tedjasukmana A, Little P, Hanono D, Wijetilaka R, Weedon J, Lin J, Toledano RD, Zhang M. Complement factor B activation in patients with preeclampsia. J Reprod Immunol 2015; 109:94-100. [PMID: 25604034 DOI: 10.1016/j.jri.2014.12.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 11/14/2014] [Accepted: 12/08/2014] [Indexed: 01/28/2023]
Abstract
Preeclampsia is a leading cause of maternal and fetal morbidity and mortality. Bb, the active fragment of complement factor B (fB), has been reported to be a predictor of preeclampsia. However, conflicting results have been found by some investigators. We hypothesized that the disagreement in findings may be due to the racial/ethnic differences among various study groups, and that fB activation is significant in women of an ethnic minority with preeclampsia. We investigated the maternal and fetal levels of Bb (the activated fB fragment) in pregnant women of an ethnic minority with or without preeclampsia. We enrolled 291 pregnant women (96% of an ethnic minority, including 78% African-American). Thirteen percent of these were diagnosed with preeclampsia. Maternal venous blood was collected from all participants together with fetal umbilical cord blood samples from 154 deliveries in the 291 women. The results were analyzed using the Mann-Whitney U test and multivariate analyses. Maternal Bb levels were significantly higher in the preeclamptic group than in the nonpreeclamptic group. Levels of Bb in fetal cord blood were similar in both groups. Subgroup analyses of African-American patients' results confirmed the study hypothesis that there would be a significant increase in Bb in the maternal blood of the preeclamptic group and no increase in Bb in the fetal cord blood of this group. These results suggest that a maternal immune response through complement fB might play a role in the development of preeclampsia, particularly in African-American patients.
Collapse
Affiliation(s)
- Ivan Velickovic
- Department of Anesthesiology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Mudar Dalloul
- Department of Obstetrics & Gynecology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Karen A Wong
- Department of Anesthesiology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Olufunke Bakare
- Department of Anesthesiology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Franz Schweis
- Department of Anesthesiology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Maya Garala
- Department of Anesthesiology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Amit Alam
- Department of Anesthesiology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Giorgio Medranda
- Department of Anesthesiology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Jovana Lekovic
- Department of Anesthesiology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Waqas Shuaib
- Department of Anesthesiology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Andreas Tedjasukmana
- Department of Anesthesiology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Perry Little
- Department of Anesthesiology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Daniel Hanono
- Department of Anesthesiology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Ruvini Wijetilaka
- Department of Anesthesiology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA; Department of Anesthesiology, Lutheran Medical Center, Brooklyn, NY 11220, USA
| | - Jeremy Weedon
- Scientific Computing Center, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Jun Lin
- Department of Anesthesiology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA; Department of Anesthesiology, University Hospital of Brooklyn at Long Island College Hospital, Brooklyn, NY 11201, USA
| | - Roulhac d'Arby Toledano
- Department of Anesthesiology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA; Department of Anesthesiology, Lutheran Medical Center, Brooklyn, NY 11220, USA
| | - Ming Zhang
- Department of Anesthesiology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA; Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA.
| |
Collapse
|
36
|
Mayer AE, Parks GD. An AGM model for changes in complement during pregnancy: neutralization of influenza virus by serum is diminished in late third trimester. PLoS One 2014; 9:e112749. [PMID: 25409303 PMCID: PMC4237339 DOI: 10.1371/journal.pone.0112749] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 10/14/2014] [Indexed: 12/28/2022] Open
Abstract
Pregnant women in the third trimester are at increased risk of severe influenza disease relative to the general population, though mechanisms behind this are not completely understood. The immune response to influenza infection employs both complement (C') and antibody (Ab). The relative contributions of these components to the anti-viral response are difficult to dissect because most humans have pre-existing influenza-specific Abs. We developed the African green monkey (AGM) as a tractable nonhuman primate model to study changes in systemic innate immunity to influenza during pregnancy. Because the AGMs were influenza-naïve, we were able to examine the role of C' in influenza virus neutralization using serum from non-pregnant animals before and after influenza infection. We determined that serum from naïve AGMs neutralized influenza via C', while post-infection neutralization did not require C', suggesting an Ab-mediated mechanism. The latter mimicked neutralization using human serum. Further, we found that ex vivo neutralization of influenza with both naïve and influenza-immune AGM serum occurred by virus particle aggregation and lysis, with immune serum lysing virus at a much higher rate than naïve serum. We hypothesized that the anti-influenza C' response would diminish late in AGM pregnancy, corresponding with the time when pregnant women suffer increased influenza severity. We found that influenza neutralization capacity is significantly diminished in serum collected late in the third trimester. Strikingly, we found that circulating levels of C3, C3a, and C4 are diminished late in gestation relative to nonpregnant animals, and while neutralization capacity and serum C3a return to normal shortly after parturition, C3 and C4 levels do not. This AGM model system will enable further studies of the role of physiologic and hormonal changes in downregulating C'-mediated anti-viral immunity during pregnancy, and it will permit the identification of therapeutic targets to improve outcomes of influenza virus infection in pregnant women.
Collapse
Affiliation(s)
- Anne E. Mayer
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27101, United States of America
| | - Griffith D. Parks
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27101, United States of America
| |
Collapse
|
37
|
Szalai G, Xu Y, Romero R, Chaiworapongsa T, Xu Z, Chiang PJ, Ahn H, Sundell B, Plazyo O, Jiang Y, Olive M, Wang B, Jacques SM, Qureshi F, Tarca AL, Erez O, Dong Z, Papp Z, Hassan SS, Hernandez-Andrade E, Than NG. In vivo experiments reveal the good, the bad and the ugly faces of sFlt-1 in pregnancy. PLoS One 2014; 9:e110867. [PMID: 25393290 PMCID: PMC4230935 DOI: 10.1371/journal.pone.0110867] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 09/23/2014] [Indexed: 01/06/2023] Open
Abstract
Objective Soluble fms-like tyrosine kinase (sFlt)-1-e15a, a primate-specific sFlt-1-isoform most abundant in the human placenta in preeclampsia, can induce preeclampsia in mice. This study compared the effects of full-length human (h)sFlt-1-e15a with those of truncated mouse (m)sFlt-1(1-3) used in previous preeclampsia studies on pregnancy outcome and clinical symptoms in preeclampsia. Methods Mice were injected with adenoviruses or fiber-mutant adenoviruses overexpressing hsFlt-1-e15a, msFlt-1(1-3) or control GFP under the CMV or CYP19A1 promoters on gestational day 8 (GD8) and GD11. Placentas and pups were delivered by cesarean section, and dams were monitored postpartum. Blood pressure was telemetrically recorded. Urine samples were collected with cystocentesis and examined for albumin/creatinine ratios. Tissue specimens were evaluated for transgene as well as endogenous mFlt-1 and msFlt-1-i13 expression. H&E-, Jones- and PAS-stained kidney sections were histopathologically examined. Placental GFP expression and aortic ring assays were investigated with confocal microscopy. Results Mean arterial blood pressure (MAP) was elevated before delivery in hsFlt-1-e15a-treated mice compared to controls (GD18: ΔMAP = 7.8 mmHg, p = 0.009), while ΔMAP was 12.8 mmHg (GD18, p = 0.005) in msFlt-1(1-3)-treated mice. Urine albumin/creatinine ratio was higher in hsFlt-1-e15a-treated mice than in controls (GD18, p = 0.04; PPD8, p = 0.03), and msFlt-1(1-3)-treated mice had marked proteinuria postpartum (PPD8, p = 4×10−5). Focal glomerular changes were detected in hsFlt-1-e15a and msFlt-1(1-3)-treated mice. Aortic ring microvessel outgrowth was decreased in hsFlt-1-e15a (p = 0.007) and msFlt-1(1-3)-treated (p = 0.02) mice. Full-length msFlt-1-i13 expression was unique for the placenta. In hsFlt-1-e15a-treated mice, the number of pups (p = 0.046), total weight of living pups (p = 0.04) and maternal weights (p = 0.04) were higher than in controls. These differences were not observed in truncated msFlt-1(1-3)-treated mice. Conclusions Truncated msFlt-1(1-3) simulated the preeclampsia-promoting effects of full-length hsFlt-1. MsFlt-1(1-3) had strong effect on maternal endothelium but not on placentas and embryos. In contrast, hsFlt-1-e15a induced preeclampsia-like symptoms; however, it also increased litter size. In accord with the predominant placental expression of hsFlt-1-e15a and msFlt-1-i13, full-length sFlt-1 may have a role in the regulation of embryonic development. These observations point to the difference in the biological effects of full-length and truncated sFlt-1 and the changes in the effect of full-length sFlt-1 during pregnancy, and may have important implications in the management of preeclampsia.
Collapse
Affiliation(s)
- Gabor Szalai
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
| | - Yi Xu
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
| | - Roberto Romero
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
- * E-mail: (RR); (NGT)
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Zhonghui Xu
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
| | - Po Jen Chiang
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
| | - Hyunyoung Ahn
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
| | - Birgitta Sundell
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
| | - Olesya Plazyo
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
| | - Yang Jiang
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
| | - Mary Olive
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Bing Wang
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
| | - Suzanne M. Jacques
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Faisal Qureshi
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Adi L. Tarca
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
- Department of Computer Science, Wayne State University, Detroit, MI, United States of America
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Offer Erez
- Department of Obstetrics and Gynecology, Soroka University Medical Center, School of Medicine, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Zhong Dong
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
| | - Zoltan Papp
- Maternity Private Department, Kutvolgyi Clinical Block, Semmelweis University, Budapest, Hungary
| | - Sonia S. Hassan
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Edgar Hernandez-Andrade
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Nandor Gabor Than
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States of America
- Maternity Private Department, Kutvolgyi Clinical Block, Semmelweis University, Budapest, Hungary
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
- * E-mail: (RR); (NGT)
| |
Collapse
|
38
|
Takeshita A, Kusakabe KT, Hiyama M, Kuniyoshi N, Kondo T, Kano K, Kiso Y, Okada T. Dynamics and reproductive effects of complement factors in the spontaneous abortion model of CBA/J×DBA/2 mice. Immunobiology 2014; 219:385-91. [DOI: 10.1016/j.imbio.2014.01.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 12/04/2013] [Accepted: 01/03/2014] [Indexed: 11/16/2022]
|
39
|
Voskoboynik A, Newman AM, Corey DM, Sahoo D, Pushkarev D, Neff NF, Passarelli B, Koh W, Ishizuka KJ, Palmeri KJ, Dimov IK, Keasar C, Fan HC, Mantalas GL, Sinha R, Penland L, Quake SR, Weissman IL. Identification of a colonial chordate histocompatibility gene. Science 2013; 341:384-7. [PMID: 23888037 PMCID: PMC3810301 DOI: 10.1126/science.1238036] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Histocompatibility is the basis by which multicellular organisms of the same species distinguish self from nonself. Relatively little is known about the mechanisms underlying histocompatibility reactions in lower organisms. Botryllus schlosseri is a colonial urochordate, a sister group of vertebrates, that exhibits a genetically determined natural transplantation reaction, whereby self-recognition between colonies leads to formation of parabionts with a common vasculature, whereas rejection occurs between incompatible colonies. Using genetically defined lines, whole-transcriptome sequencing, and genomics, we identified a single gene that encodes self-nonself and determines "graft" outcomes in this organism. This gene is significantly up-regulated in colonies poised to undergo fusion and/or rejection, is highly expressed in the vasculature, and is functionally linked to histocompatibility outcomes. These findings establish a platform for advancing the science of allorecognition.
Collapse
Affiliation(s)
- Ayelet Voskoboynik
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Developmental Biology, Stanford University, Hopkins Marine Station, Pacific Grove, CA 93950, USA
| | - Aaron M. Newman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Daniel M. Corey
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Debashis Sahoo
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Dmitry Pushkarev
- Departments of Applied Physics and Bioengineering, Stanford University, and Howard Hughes Medical Institute, Stanford, CA 94305, USA
| | - Norma F. Neff
- Departments of Applied Physics and Bioengineering, Stanford University, and Howard Hughes Medical Institute, Stanford, CA 94305, USA
| | - Benedetto Passarelli
- Departments of Applied Physics and Bioengineering, Stanford University, and Howard Hughes Medical Institute, Stanford, CA 94305, USA
| | - Winston Koh
- Departments of Applied Physics and Bioengineering, Stanford University, and Howard Hughes Medical Institute, Stanford, CA 94305, USA
| | - Katherine J. Ishizuka
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Developmental Biology, Stanford University, Hopkins Marine Station, Pacific Grove, CA 93950, USA
| | - Karla J. Palmeri
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Developmental Biology, Stanford University, Hopkins Marine Station, Pacific Grove, CA 93950, USA
| | - Ivan K. Dimov
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Chen Keasar
- Department of Computer Science, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - H. Christina Fan
- Departments of Applied Physics and Bioengineering, Stanford University, and Howard Hughes Medical Institute, Stanford, CA 94305, USA
| | - Gary L. Mantalas
- Departments of Applied Physics and Bioengineering, Stanford University, and Howard Hughes Medical Institute, Stanford, CA 94305, USA
| | - Rahul Sinha
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lolita Penland
- Departments of Applied Physics and Bioengineering, Stanford University, and Howard Hughes Medical Institute, Stanford, CA 94305, USA
| | - Stephen R. Quake
- Departments of Applied Physics and Bioengineering, Stanford University, and Howard Hughes Medical Institute, Stanford, CA 94305, USA
| | - Irving L. Weissman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Developmental Biology, Stanford University, Hopkins Marine Station, Pacific Grove, CA 93950, USA
- Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
40
|
Khattab A, Kremsner P, Meri S. Complement activation in primiparous women from a malaria endemic area is associated with reduced birthweight. Placenta 2013; 34:162-7. [DOI: 10.1016/j.placenta.2012.11.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Revised: 11/22/2012] [Accepted: 11/22/2012] [Indexed: 10/27/2022]
|
41
|
Chaouat G. Inflammation, NK cells and implantation: friend and foe (the good, the bad and the ugly?): replacing placental viviparity in an evolutionary perspective. J Reprod Immunol 2013; 97:2-13. [PMID: 23347505 DOI: 10.1016/j.jri.2012.10.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Revised: 09/25/2012] [Accepted: 10/01/2012] [Indexed: 01/14/2023]
Abstract
This review summarises an invited talk presented at the 2012 ESRI/ASRI meeting in Hamburg, concerning current views of inflammation in pregnancy, which is timely given that the effects of a local injury in the uterus acts to favour implantation. Recalling that inflammation can be good (it is useful and necessary for implantation), bad (in implantation failure, RSA) and ugly (at the extreme, endometriosis is associated with pain and infertility) leads to consideration of its status in pregnancy. Its role in implantation and the fact that pregnancy maintains some aspects of inflammation throughout, leads to revision of not only concepts of immunosuppression and the Th1/Th2 paradigm, but also the feto-maternal relationship as seen since Medawar's hypotheses were advanced. This is examined from an evolutionary perspective, which should lead to further review of our perception of uterine NK cells, and the emergence of Treg cells to control some aspects of adaptive immunity, which appeared long after placentation.
Collapse
Affiliation(s)
- Gérard Chaouat
- U 976 INSERM, Pavillon Equerre Bazin, Hôpital Saint Louis, Paris, France.
| |
Collapse
|
42
|
Interleukin-15 (IL-15) and anti-C1q antibodies as serum biomarkers for ectopic pregnancy and missed abortion. Clin Dev Immunol 2013; 2013:637513. [PMID: 23401701 PMCID: PMC3562602 DOI: 10.1155/2013/637513] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2012] [Accepted: 11/25/2012] [Indexed: 11/17/2022]
Abstract
Given the present lack of clinically useful tests for the accurate diagnosis of ectopic pregnancy (EP), there is a need to select out those immunological factors measured in the maternal serum, as potential biomarkers. Our assumption was that C1q/anti-C1q antibody complexes and serum levels of interleukin-15 (IL-15) may play a role in differentiating abortions (MAs) and EPs and normal pregnancies. We assessed whether their measurement could set the diagnosis in a case control study at 6–8 weeks consisting of 60 women with failed early pregnancy (30 EPs, 30 MAs) and 33 women with intrauterine pregnancies. Normal pregnancies contain anti-C1q antibodies more frequently compared to women with failed pregnancies, the lowest levels being found in EPs, but this lacked statistical significance and anti-C1q could not serve as a marker. However EP pregnancies had elevated IL-15 levels that could statistically significantly differentiate them from MAs and IUPs. Furthermore, when assessing IL-15 for the clinically important differentiation between IUP and EP, we found at a cut-off of 16 pg/mL a negative predictive value of 99 with a sensitivity for diagnosing an EP of 92%. According to these results, serum IL-15 is a promising marker differentiating an MA from an EP.
Collapse
|
43
|
Denny KJ, Woodruff TM, Taylor SM, Callaway LK. Complement in pregnancy: a delicate balance. Am J Reprod Immunol 2012; 69:3-11. [PMID: 22925193 DOI: 10.1111/aji.12000] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 07/09/2012] [Indexed: 02/06/2023] Open
Abstract
The complement system is a key component of innate host defence that, under normal conditions, is responsible for the opsonization and destruction of potential pathogens. However, inappropriate or excessive activation of complement can have a detrimental effect on the host and has been implicated in the pathophysiology of numerous disease states. Recently, there has been increasing evidence for a role of the complement system and, in particular, the potent pro-inflammatory anaphylatoxin complement component 5a (C5a) in both normal and complicated pregnancy. The following review describes the results of in vitro, animal, and human clinical studies investigating the role of the complement system in healthy pregnancy, recurrent miscarriage, preterm birth, and preeclampsia.
Collapse
Affiliation(s)
- Kerina J Denny
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia.
| | | | | | | |
Collapse
|
44
|
Reggia R, Ziglioli T, Andreoli L, Bellisai F, Iuliano A, Gerosa M, Ramoni V, Tani C, Brucato A, Galeazzi M, Mosca M, Caporali R, Meroni PL, Tincani A. Primary anti-phospholipid syndrome: any role for serum complement levels in predicting pregnancy complications? Rheumatology (Oxford) 2012; 51:2186-90. [PMID: 22923750 DOI: 10.1093/rheumatology/kes225] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE To evaluate the association between serum complement levels (C3 and C4) and obstetric complications. METHODS Fifty-seven pregnancies in primary APS (PAPS) patients were compared with 49 pregnancies in patients with UCTD and SS. A group of 175 healthy pregnant women were studied to calculate a normality range for C3 and C4 during pregnancy. Such a range was applied to define hypocomplementaemia in PAPS and UCTD/SS. RESULTS Both groups of patients (PAPS and UCTD/SS) showed significantly lower levels of C3 and C4 in each trimester as compared with healthy women; conversely, no significant difference was found between PAPS and UCTD/SS. Comparing PAPS pregnancies with and without complications, no difference was found in the prevalence of low C3 or low C4. CONCLUSION No association was found between hypocomplementaemia and obstetric complications in PAPS. However, both cases of pre-eclampsia were characterized by low C3 throughout pregnancy. There is evidence that the complement system is a contributor to the mechanisms of aPL-mediated damage, but its predictive role on the final pregnancy outcome does not seem to be of major impact.
Collapse
Affiliation(s)
- Rossella Reggia
- Rheumatology and Clinical Immunology, Spedali Civili and University of Brescia, Brescia, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Molecular genetics of preeclampsia and HELLP syndrome - a review. Biochim Biophys Acta Mol Basis Dis 2012; 1822:1960-9. [PMID: 22917566 DOI: 10.1016/j.bbadis.2012.08.004] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Revised: 08/06/2012] [Accepted: 08/07/2012] [Indexed: 11/24/2022]
Abstract
Preeclampsia is characterised by new onset hypertension and proteinuria and is a major obstetrical problem for both mother and foetus. Haemolysis elevated liver enzymes and low platelets (HELLP) syndrome is an obstetrical emergency and most cases occur in the presence of preeclampsia. Preeclampsia and HELLP are complicated syndromes with a wide variety in severity of clinical symptoms and gestational age at onset. The pathophysiology depends not only on periconceptional conditions and the foetal and placental genotype, but also on the capability of the maternal system to deal with pregnancy. Genetically, preeclampsia is a complex disorder and despite numerous efforts no clear mode of inheritance has been established. A minor fraction of HELLP cases is caused by foetal homozygous LCHAD deficiency, but for most cases the genetic background has not been elucidated yet. At least 178 genes have been described in relation to preeclampsia or HELLP syndrome. Confined placental mosaicism (CPM) is documented to cause early onset preeclampsia in some cases; the overall contribution of CPM to the occurrence of preeclampsia has not been adequately investigated yet. This article is part of a Special Issue entitled: Molecular Genetics of Human Reproductive Failure.
Collapse
|
46
|
Wang W, Irani RA, Zhang Y, Ramin SM, Blackwell SC, Tao L, Kellems RE, Xia Y. Autoantibody-mediated complement C3a receptor activation contributes to the pathogenesis of preeclampsia. Hypertension 2012; 60:712-21. [PMID: 22868393 DOI: 10.1161/hypertensionaha.112.191817] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Preeclampsia (PE) is a prevalent life-threatening hypertensive disorder of pregnancy associated with increased complement activation. However, the causative factors and pathogenic role of increased complement activation in PE are largely unidentified. Here we report that a circulating maternal autoantibody, the angiotensin II type 1 receptor agonistic autoantibody, which emerged recently as a potential pathogenic contributor to PE, stimulates deposition of complement C3 in placentas and kidneys of pregnant mice via angiotensin II type 1 receptor activation. Next, we provide in vivo evidence that selectively interfering with C3a signaling by a complement C3a receptor-specific antagonist significantly reduces hypertension from 167±7 to 143±5 mm Hg and proteinuria from 223.5±7.5 to 78.8±14.0 μg of albumin per milligram creatinine (both P<0.05) in angiotensin II type 1 receptor agonistic autoantibody-injected pregnant mice. In addition, we demonstrated that complement C3a receptor antagonist significantly inhibited autoantibody-induced circulating soluble fms-like tyrosine kinase 1, a known antiangiogenic protein associated with PE, and reduced small placental size with impaired angiogenesis and intrauterine growth restriction. Similarly, in humans, we demonstrate that C3 deposition is significantly elevated in the placentas of preeclamptic patients compared with normotensive controls. Lastly, we show that complement C3a receptor activation is a key mechanism underlying autoantibody-induced soluble fms-like tyrosine kinase 1 secretion and decreased angiogenesis in cultured human villous explants. Overall, we provide mouse and human evidence that angiotensin II type 1 receptor agonistic autoantibody-mediated activation contributes to elevated C3 and that complement C3a receptor signaling is a key mechanism underlying the pathogenesis of the disease. These studies are the first to link angiotensin II type 1 receptor agonistic autoantibody with complement activation and to provide important new opportunities for therapeutic intervention in PE.
Collapse
Affiliation(s)
- Wei Wang
- Department of Nephrology, Xiangya Hospital, Central South University, Hunan, China
| | | | | | | | | | | | | | | |
Collapse
|
47
|
de la Torre YM, Pregnolato F, D’Amelio F, Grossi C, DiSimone N, Pasqualini F, Nebuloni M, Chen P, Pierangeli S, Bassani N, Ambrogi F, Borghi MO, Vecchi A, Locati M, Meroni PL. Anti-phospholipid induced murine fetal loss: novel protective effect of a peptide targeting the β2 glycoprotein I phospholipid-binding site. Implications for human fetal loss. J Autoimmun 2012; 38:J209-15. [PMID: 22196923 PMCID: PMC3313005 DOI: 10.1016/j.jaut.2011.11.009] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Revised: 11/21/2011] [Accepted: 11/22/2011] [Indexed: 10/14/2022]
Abstract
β2 glycoprotein I (β2GPI)-dependent anti-phospholipid antibodies (aPL) induce thrombosis and affect pregnancy. The CMV-derived synthetic peptide TIFI mimics the PL-binding site of β2GPI and inhibits β2GPI cell-binding in vitro and aPL-mediated thrombosis in vivo. Here we investigated the effect of TIFI on aPL-induced fetal loss in mice. TIFI inhibitory effect on in vitro aPL binding to human trophoblasts was evaluated by indirect immunofluorescence and ELISA. TIFI effect on aPL-induced fetal loss was investigated in pregnant C57BL/6 mice treated with aPL or normal IgG (NHS). Placenta/fetus weight and histology and RNA expression were analyzed. TIFI, but not the control peptide VITT, displayed a dose-dependent inhibition of aPL binding to trophoblasts in vitro. Injection of low doses of aPL at day 0 of pregnancy caused growth retardation and increased fetal loss rate, both significantly reduced by TIFI but not VITT. Consistent with observations in humans, histological analysis showed no evidence of inflammation in this model, as confirmed by the absence of an inflammatory signature in gene expression analysis, which in turn revealed a TIFI-dependent modulation of molecules involved in differentiation and development processes. These findings support the non-inflammatory pathogenic role of aPL and suggest innovative therapeutic approaches to aPL-dependent fetal loss.
Collapse
Affiliation(s)
| | | | - Fabio D’Amelio
- Department of Internal Medicine, University of Milan, Italy
| | | | | | | | - Manuela Nebuloni
- L. Sacco Department of Medical Science, University of Milan, Italy
| | - Pojen Chen
- Department of Medicine, University of Los Angeles, CA -USA
| | - Silvia Pierangeli
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX - USA
| | | | | | - Maria-Orietta Borghi
- Istituto Auxologico Italiano, Milan, Italy
- Department of Internal Medicine, University of Milan, Italy
| | | | - Massimo Locati
- Istituto Clinico Humanitas, Rozzano, Italy
- Department of Translational Medicine, University of Milan, Italy
| | - Pier-Luigi Meroni
- Istituto Auxologico Italiano, Milan, Italy
- Department of Internal Medicine, University of Milan, Italy
- Istituto G. Pini, Milan, Italy
| |
Collapse
|
48
|
Perricone C, de Carolis C, Perricone R. Pregnancy and autoimmunity: A common problem. Best Pract Res Clin Rheumatol 2012; 26:47-60. [DOI: 10.1016/j.berh.2012.01.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2012] [Accepted: 01/12/2012] [Indexed: 12/15/2022]
|
49
|
Berencsi G, Takács M. Barriers of the Human Organism and Their Achilles’ Heels. MATERNAL FETAL TRANSMISSION OF HUMAN VIRUSES AND THEIR INFLUENCE ON TUMORIGENESIS 2012. [PMCID: PMC7121758 DOI: 10.1007/978-94-007-4216-1_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The human body is covered by barriers separating it from the external and internal surroundings. The “milieu enterieur” has to be stabilised in spite of the variable external and internal conditions of toxic, osmotic, microbial and climatic environmental circumstances. This first line of barriers is composed of skin and mucous membranes of complicated structures. A second line of barrier system is present in our organisms. Certain organs have to be separated from the immune system and other parts of the body because of evolutionary reasons (eye-bulb and testicles) because of unique proteins “unknown” for the acquired immune system. The blood-brain barrier (BBB) is providing enhanced safety circumstances for the central nervous system. The second line of barriers is represented by the special properties of the capillary endothelial system. The maternal-fetal barrier is the most complex. At the maternal fetal interface two individuals of two different haplotypes has to be live 9 months separated by a very complicated dynamic barrier. The placenta is the organ, which is separating the maternal and fetal tissues. Similar to others the bidirectional transport of gasses, metabolites, cells, proteins, regulatory substances, are transported by active or passive transcellular and intercellular mechanisms. The fetal immune system develops immunotolerance to all maternal cells and antigens transferred transplacentally. The problem is to mitigate the maternal immune system to tolerate the paternal haplotype of the fetus. In the case of normal pregnancy a complex series of physiological modifications can solve the problem without harmful consequences to the mother and fetus. The outermost contact cells of trophoblasts express instead of HLA-class Ia and class II antigens non-variable HLA-C, HLA-E, HLA-F and HLA-G antigens. The first consequence of this is reduction of the activity of maternal natural killer cells and maternal dendritic cells; Progesteron, micro-RNA and mediators influence the development of T effector-cells. The production of soluble HLA-G(5 and 6) and IL-10 supports the differentiation of Th-2 CD4+ helper cells, reducing the ability of maternal cells to kill fetal cells. Series of receptors and costimulators are expressed by the different lines of semi-allogenic trophoblast cells to bind HLA-G and mitigate maternal immune response; The maternal immunotolerance is further facilitated by the activation of CD4+CD25brightFoxp3+ regulatory T (TREG) cells. Infections have to be prevented during pregnancy. The cells of placenta express 10 Toll-like receptors a group of pattern recognition receptors responsible for innate immunity. The interferon level is also higher in the placental tissues than in the somatic fetal or maternal cells. The complement system is also adapted to the requirements of the pregnancy and fetal damage is inhibited by the production of “assymmetric IgG antibodies” under hormonal and placental-regulation. These modifications prevent the activation of complement, cytotoxic activity, opsonising ability, antigen clearance and precipitating activity of the molecules. The Achilles’ heels of the different barriers are regularly found by virus infections. Lamina cribrosa of the blood-brain barrier, optical nerve of the eyes, etc. the risk factors of the maternal-fetal barrier has been summarised in Table 1.1.
Collapse
|
50
|
MBL interferes with endovascular trophoblast invasion in pre-eclampsia. Clin Dev Immunol 2011; 2012:484321. [PMID: 22203857 PMCID: PMC3235499 DOI: 10.1155/2012/484321] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2011] [Accepted: 09/07/2011] [Indexed: 02/01/2023]
Abstract
The spiral arteries undergo physiologic changes during pregnancy, and the failure of this process may lead to a spectrum of pregnancy disorders, including pre-eclampsia. Our recent data indicate that decidual endothelial cells (DECs), covering the inner side of the spiral arteries, acquire the ability to synthesize C1q, which acts as a link between endovascular trophoblast and DECs favouring the process of vascular remodelling. In this study, we have shown that sera obtained from pre-eclamptic patients strongly inhibit the interaction between extravillous trophoblast (EVT) and DECs, preventing endovascular invasion of trophoblast cells. We further demonstrated that mannose-binding lectin (MBL), one of the factor increased in pre-eclamptic patient sera, strongly inhibits the interaction of EVT with C1q interfering with the process of EVT adhesion to and migration through DECs. These data suggest that the increased level of MBL in pre-eclampsia may contribute to the failure of the endovascular invasion of trophoblast cells.
Collapse
|