1
|
Lee S, Yoo I, Cheon Y, Ka H. Complement system molecules: Expression and regulation at the maternal-conceptus interface during pregnancy in pigs. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 159:105229. [PMID: 39004297 DOI: 10.1016/j.dci.2024.105229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/11/2024] [Accepted: 07/11/2024] [Indexed: 07/16/2024]
Abstract
The complement system, composed of complement components and complement control proteins, plays an essential role in innate immunity. Complement system molecules are expressed at the maternal-conceptus interface, and inappropriate activation of the complement system is associated with various adverse pregnancy outcomes in humans and rodents. However, the expression, regulation, and function of the complement system at the maternal-conceptus interface in pigs have not been studied. In this study, we investigated the expression, localization, and regulation of complement system molecules at the maternal-conceptus interface in pigs. Complement components and complement control proteins were expressed in the endometrium, early-stage conceptus, and chorioallantoic tissues during pregnancy. The expression of complement components acting on the early stage of complement activation increased in the endometrium on Day 15 of pregnancy, with greater levels on that day compared with the estrous cycle. Localization of several complement components and complement control proteins was cell-type specific in the endometrium. The expression of C1QC, C2, C3, C4A, CFI, ITGB2, MASP1, and SERPING1 was increased by IFNG in endometrial explant tissues. Furthermore, cleaved C3 fragments were detected in endometrial tissues and uterine flushings on Day 15 of the estrous cycle and Day 15 of pregnancy, with greater levels on Day 15 of pregnancy. These results suggest that complement system molecules in pigs expressed at the maternal-conceptus interface play important roles in the establishment and maintenance of pregnancy by regulating innate immunity and modulating the maternal immune environment during pregnancy.
Collapse
Affiliation(s)
- Soohyung Lee
- Division of Biological Science and Technology, Yonsei University, Wonju, 26493, Republic of Korea.
| | - Inkyu Yoo
- Division of Biological Science and Technology, Yonsei University, Wonju, 26493, Republic of Korea
| | - Yugyeong Cheon
- Division of Biological Science and Technology, Yonsei University, Wonju, 26493, Republic of Korea
| | - Hakhyun Ka
- Division of Biological Science and Technology, Yonsei University, Wonju, 26493, Republic of Korea.
| |
Collapse
|
2
|
Bi Y, Li T, Zhang S, Yang Y, Dong M. Bioinformatics-based analysis of the dialog between COVID-19 and RSA. Heliyon 2024; 10:e30371. [PMID: 38737245 PMCID: PMC11088317 DOI: 10.1016/j.heliyon.2024.e30371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 04/24/2024] [Accepted: 04/24/2024] [Indexed: 05/14/2024] Open
Abstract
Pregnant women infected with SARS-CoV-2 in early pregnancy may face an increased risk of miscarriage due to immune imbalance at the maternal-fetal interface. However, the molecular mechanisms underlying the crosstalk between COVID-19 infection and recurrent spontaneous abortion (RSA) remain poorly understood. This study aimed to elucidate the transcriptomic molecular dialog between COVID-19 and RSA. Based on bioinformatics analysis, 307 common differentially expressed genes were found between COVID-19 (GSE171110) and RSA (GSE165004). Common DEGs were mainly enriched in ribosome-related and cell cycle-related signaling pathways. Using degree algorithm, the top 10 hub genes (RPS27A, RPL5, RPS8, RPL4, RPS2, RPL30, RPL23A, RPL31, RPL26, RPL37A) were selected from the common DEGs based on their scores. The results of the qPCR were in general agreement with the results of the raw letter analysis. The top 10 candidate drugs were also selected based on P-values. In this study, we provide molecular markers, signaling pathways, and small molecule compounds that may associate COVID-19. These findings may increase the accurate diagnosis and treatment of COVID-19 patients.
Collapse
Affiliation(s)
- Yin Bi
- Guangxi Reproductive Medical Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Guangxi Medical University, Nanning, 530000, China
- The Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning, 530000, China
| | - Ting Li
- Guangxi Reproductive Medical Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Guangxi Medical University, Nanning, 530000, China
- The Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning, 530000, China
| | - Shun Zhang
- Department of Reproductive Medical Center, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, China
| | - Yihua Yang
- Guangxi Reproductive Medical Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Guangxi Medical University, Nanning, 530000, China
- The Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning, 530000, China
| | - Mingyou Dong
- Guangxi Reproductive Medical Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China
- The Key Laboratory of Molecular Pathology (For Hepatobiliary Diseases) of Guangxi, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, China
| |
Collapse
|
3
|
Yasmin H, Agostinis C, Toffoli M, Roy T, Pegoraro S, Balduit A, Zito G, Di Simone N, Ricci G, Madan T, Kishore U, Bulla R. Protective role of complement factor H against the development of preeclampsia. Front Immunol 2024; 15:1351898. [PMID: 38464530 PMCID: PMC10920295 DOI: 10.3389/fimmu.2024.1351898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 01/30/2024] [Indexed: 03/12/2024] Open
Abstract
Pregnancy is an immunologically regulated, complex process. A tightly controlled complement system plays a crucial role in the successful establishment of pregnancy and parturition. Complement inhibitors at the feto-maternal interface are likely to prevent inappropriate complement activation to protect the fetus. In the present study, we aimed to understand the role of Factor H (FH), a negative regulator of complement activation, in normal pregnancy and in a model of pathological pregnancy, i.e. preeclampsia (PE). The distribution and expression of FH was investigated in placental tissues, various placental cells, and in the sera of healthy (CTRL) or PE pregnant women via immunohistochemistry, RT-qPCR, ELISA, and Western blot. Our results showed a differential expression of FH among the placental cell types, decidual stromal cells (DSCs), decidual endothelial cells (DECs), and extravillous trophoblasts (EVTs). Interestingly, FH was found to be considerably less expressed in the placental tissues of PE patients compared to normal placental tissue both at mRNA and protein levels. Similar results were obtained by measuring circulating FH levels in the sera of third trimester CTRL and PE mothers. Syncytiotrophoblast microvesicles, isolated from the placental tissues of PE and CTRL women, downregulated FH expression by DECs. The present study appears to suggest that FH is ubiquitously present in the normal placenta and plays a homeostatic role during pregnancy.
Collapse
Affiliation(s)
- Hadida Yasmin
- Immunology and Cell Biology Laboratory, Department of Zoology, Cooch Behar Panchanan Barma University, Cooch Behar, West Bengal, India
| | - Chiara Agostinis
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Miriam Toffoli
- Department of Medical, Surgical and Health Science, University of Trieste, Trieste, Italy
| | - Tamali Roy
- Immunology and Cell Biology Laboratory, Department of Zoology, Cooch Behar Panchanan Barma University, Cooch Behar, West Bengal, India
| | - Silvia Pegoraro
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Andrea Balduit
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Gabriella Zito
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Nicoletta Di Simone
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Milan, Italy
| | - Giuseppe Ricci
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
- Department of Medical, Surgical and Health Science, University of Trieste, Trieste, Italy
| | - Taruna Madan
- Department of Innate Immunity, ICMR-National Institute for Research in Reproductive and Child Health (NIRRCH), Mumbai, India
| | - Uday Kishore
- Department of Veterinary Medicine, U.A.E. University, Al Ain, United Arab Emirates
- Zayed Centre for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Roberta Bulla
- Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
4
|
Govender S, Naicker T. The Contribution of Complement Protein C1q in COVID-19 and HIV Infection Comorbid with Preeclampsia: A Review. Int Arch Allergy Immunol 2022; 183:1114-1126. [PMID: 35661665 PMCID: PMC9393774 DOI: 10.1159/000524976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/21/2022] [Indexed: 11/19/2022] Open
Abstract
Dysregulation in component 1q (C1q) levels is associated with weak placental development in preeclampsia (PE). Human immunodeficiency virus infection (HIV-1) triggers the C1q complex, resulting in opsonization of healthy host cells, contributing to their removal, and augmented progression of HIV disease. In coronavirus disease 2019 (COVID-19)-infected patients, the deposition of C1q activates the complement. Considering the paucity of data, this review highlights a significant gap in the potential of C1q in the immunocompromised state of preeclamptic HIV-infected women and COVID-19 infection. In PE, C1q is downregulated; while in antiretroviral treatment-treated HIV/COVID-19 infected patients, C1q is upregulated. It is plausible that C1q is augmented in the triad and may exacerbate severity of disease. This thereby provides a foundation for future intended research which involves the investigation of single nucleotide polymorphism expression of the C1q gene, specifically in these diseases.
Collapse
Affiliation(s)
- Sumeshree Govender
- Optics & Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Thajasvarie Naicker
- Optics & Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
5
|
Yang Y, Liu H, Zhao Y, Geng C, Chao L, Hao A. Grim-19 deficiency promotes decidual macrophage autophagy in recurrent spontaneous abortion. Front Endocrinol (Lausanne) 2022; 13:1023194. [PMID: 36387896 PMCID: PMC9641028 DOI: 10.3389/fendo.2022.1023194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 10/11/2022] [Indexed: 11/13/2022] Open
Abstract
Dysregulation of decidual macrophages leads to the occurrence of recurrent spontaneous abortion (RSA). However, the role of macrophages in RSA occurrence remains unclear. In this study, we found that the expression of Grim-19 was decreased, and the expression of autophagy related proteins Beclin1, LC3B II/I and BNIP3 was markedly upregulated in decidual macrophages of RSA patients compared with the normal pregnancy group. Furthermore, we demonstrated that downregulation of GRIM-19 increased the expression of autophagy related proteins Beclin1, LC3B II/I, BNIP3 and the proinflammatory cytokines IL1B, IL6 and TNFa in uterine mononuclear cells of GRIM-19+/- mice. The proportion of CD45+CD11b+F4/80+LC3B+ cells in GRIM-19+/- mouse uteri was significantly higher than that in WT mouse uteri. In addition, we confirmed that inhibition of Grim-19 by siRNA enhanced the expression of autophagy related proteins in RAW264.7 cells and THP-1 cells. More importantly, downregulation of Grim-19 in RAW264.7 cells promoted the release of proinflammatory cytokines and promoted phagocytic activity, which could be reversed by autophagy blockade. For THP-1-derived macrophages, the results of RNA-seq suggested that Grim-19 mainly modulates immune and inflammatory-related pathways, leading to cytokine production, and thus contributing to inflammation. Therefore, our data reveal that Grim-19 deficiency influences macrophage function, characterized by enhanced proinflammatory cytokines and phagocytic activity, and this might be regulated by autophagy. This may represent a novel mechanism for the occurrence of RSA.
Collapse
Affiliation(s)
- Yang Yang
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Haoran Liu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Yue Zhao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Chen Geng
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Lan Chao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Aijun Hao
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Correspondence: Aijun Hao,
| |
Collapse
|
6
|
Circulating C1q levels in health and disease, more than just a biomarker. Mol Immunol 2021; 140:206-216. [PMID: 34735869 DOI: 10.1016/j.molimm.2021.10.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 10/03/2021] [Accepted: 10/11/2021] [Indexed: 12/21/2022]
Abstract
C1q is the recognition molecule of the classical pathway of the complement system. By binding to its targets, such as antigen-bound immunoglobulins or C-reactive protein, C1q contributes to the innate defense against infections. However, C1q also plays several other roles beyond its traditional role in complement activation. Circulating levels of C1q are determined in routine diagnostics as biomarker in several diseases. Decreased C1q levels are present in several autoimmune conditions. The decreased levels reflect the consumption of C1q by complement activation and serves as a biomarker for disease activity. In contrast, increased C1q levels are present in infectious and inflammatory diseases and may serve as a diagnostic biomarker. The increased levels of C1q are still incompletely understood but are suggested to modulate the adaptive immune response as C1q is known to impact on the maturation status of antigen-presenting cells and C1q impacts directly on T cells leading to decreased T-cell activity in high C1q conditions. In this review, we provide a comprehensive overview of the current literature on circulating levels of C1q in health and disease, and discuss how C1q can both protect against infections as well as maintain tolerance by regulating adaptive immunity.
Collapse
|
7
|
Zeng W, Wu AG, Zhou XG, Khan I, Zhang RL, Lo HH, Qu LQ, Song LL, Yun XY, Wang HM, Chen J, Ng JPL, Ren F, Yuan SY, Yu L, Tang Y, Huang GX, Wong VKW, Chung SK, Mok SWF, Qin DL, Sun HL, Liu L, Hsiao WLW, Law BYK. Saponins isolated from Radix polygalae extent lifespan by modulating complement C3 and gut microbiota. Pharmacol Res 2021; 170:105697. [PMID: 34062240 DOI: 10.1016/j.phrs.2021.105697] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/26/2021] [Accepted: 05/26/2021] [Indexed: 02/07/2023]
Abstract
With the increase in human lifespan, population aging is one of the major problems worldwide. Aging is an irreversible progressive process that affects humans via multiple factors including genetic, immunity, cellular oxidation and inflammation. Progressive neuroinflammation contributes to aging, cognitive malfunction, and neurodegenerative diseases. However, precise mechanisms or drugs targeting age-related neuroinflammation and cognitive impairment remain un-elucidated. Traditional herbal plants have been prescribed in many Asian countries for anti-aging and the modulation of aging-related symptoms. In general, herbal plants' efficacy is attributed to their safety and polypharmacological potency via the systemic manipulation of the body system. Radix polygalae (RP) is a herbal plant prescribed for anti-aging and the relief of age-related symptoms; however, its active components and biological functions remained un-elucidated. In this study, an active methanol fraction of RP containing 17 RP saponins (RPS), was identified. RPS attenuates the elevated C3 complement protein in aged mice to a level comparable to the young control mice. The active RPS also restates the aging gut microbiota by enhancing beneficial bacteria and suppressing harmful bacteria. In addition, RPS treatment improve spatial reference memory in aged mice, with the attenuation of multiple molecular markers related to neuroinflammation and aging. Finally, the RPS improves the behavior and extends the lifespan of C. elegans, confirming the herbal plant's anti-aging ability. In conclusion, through the mouse and C. elegas models, we have identified the beneficial RPS that can modulate the aging process, gut microbiota diversity and rectify several aging-related phenotypes.
Collapse
Affiliation(s)
- Wu Zeng
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau; Department of Center for Neuro-metabolism and Regeneration Research, Bioland Laboratory, Guangzhou, China
| | - An Guo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drug Ability Evaluation, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiao-Gang Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Drug Ability Evaluation, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Imran Khan
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau
| | - Rui Long Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau
| | - Hang Hong Lo
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau
| | - Li Qun Qu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau
| | - Lin Lin Song
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau
| | - Xiao Yun Yun
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau
| | - Hui Miao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau
| | - Juan Chen
- The Key Laboratory of Molecular Biology of Infectious Diseases designated by the Chinese Ministry of Education, Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Jerome P L Ng
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau
| | - Fang Ren
- The Key Laboratory of Molecular Biology of Infectious Diseases designated by the Chinese Ministry of Education, Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Si Yu Yuan
- The Key Laboratory of Molecular Biology of Infectious Diseases designated by the Chinese Ministry of Education, Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Lu Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drug Ability Evaluation, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yong Tang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau; Sichuan Key Medical Laboratory of New Drug Discovery and Drug Ability Evaluation, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Guo Xin Huang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau
| | - Vincent Kam Wai Wong
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau
| | - Sookja Kim Chung
- Department of Medicine, Macau University of Science and Technology, Taipa, Macau
| | - Simon Wing Fai Mok
- Department of Medicine, Macau University of Science and Technology, Taipa, Macau
| | - Da Lian Qin
- Sichuan Key Medical Laboratory of New Drug Discovery and Drug Ability Evaluation, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Hua Lin Sun
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau
| | - Liang Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau.
| | - W L Wendy Hsiao
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau.
| | - Betty Yuen Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau.
| |
Collapse
|
8
|
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
|
9
|
Wang J, Zhu D, Yin J, Ma C, Peng X, Zou H, Cao Y, Xu X. Upregulated HMGB1 levels in maternal-fetal interface of patients with unexplained recurrent spontaneous abortion from different sources. J Matern Fetal Neonatal Med 2021; 35:6542-6549. [PMID: 33944653 DOI: 10.1080/14767058.2021.1918084] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE To investigate the expression and sources of high mobility group box 1 (HMGB1) protein in the maternal-fetal interface of patients with unexplained recurrent spontaneous abortion (URSA), and further to verify the role of HMGB1 in the etiology of URSA. METHODS 55 women at early pregnancy with URSA and 55 women undergoing selective termination of normal early pregnancy as control were included. The abortion tissues including villi and decidua were collected. The expression of HMGB1, CD45, CK7, and vimentin in abortion tissues was detected, and the localization and sources of HMGB1 were analyzed. RESULTS Infiltrating immune cells and expression of HMGB1 were significantly increased in villi and decidua in URSA group compared with those in the control group. In the URSA group, HMGB1 was colocalized with the CD45-labeled immune cells, and it was more obvious in decidua than in villi; in addition, HMGB1 was colocalized with the vimentin-labeled decidual stromal cells, but not with the CK7- labeled villous epithelial cells. CONCLUSION High expression of HMGB1 in the maternal-fetal interface in URSA patients was actively secreted by the infiltrating immune cells, and decidual stromal cells may passively release HMGB1 during necrosis.
Collapse
Affiliation(s)
- Jing Wang
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, China.,Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, China
| | - Damin Zhu
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, China.,Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, China.,Department of Obstetrics and Gynecology, Chaohu Hospital of Anhui Medical University, Chaohu, China
| | - Jiaqian Yin
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, China.,Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, China
| | - Cong Ma
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, China.,Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, China
| | - Xiaoqing Peng
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, China.,Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, China
| | - Huijuan Zou
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, China.,Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, China
| | - Yunxia Cao
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, China.,Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, China
| | - Xiaofeng Xu
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, China.,Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, China
| |
Collapse
|
10
|
Gao P, Zha Y, Gong X, Qiao F, Liu H. The role of maternal-foetal interface inflammation mediated by NLRP3 inflammasome in the pathogenesis of recurrent spontaneous abortion. Placenta 2020; 101:221-229. [PMID: 33022545 DOI: 10.1016/j.placenta.2020.09.067] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 09/16/2020] [Accepted: 09/28/2020] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Approximately half of the recurrent spontaneous abortions (RSAs) that remain unidentified to date may be closely related to inflammation. Our previous study found excessive NLRP3 inflammasomes in RSA patients. Here, we investigated further the role of inflammasomes in the maternal-foetal interface of RSA patients. METHODS Villous and decidual tissues were collected during uterine curettage. The trophoblast cell line TEV-1 was cultured with lipopolysaccharide (LPS) or low molecular weight heparin (LMWH), and then the macrophage cell line RAW264.7 was treated with trophoblast media. The expression and localisation of inflammasomes in tissues and cells were detected, and the migration and proliferation of cells were analysed. RESULTS A significantly increased expression of inflammasomes was observed in RSA tissues compared with those in the normal group, and it was more obvious in villous tissues than in decidual tissues. In TEV-1 cells, after LPS stimulation, the expression of inflammasomes was increased, but the cell activity was decreased, whereas in RAW264.7, both expression of inflammasomes and cell activity were increased in the LPS group. In addition, LMWH could inhibit the action of LPS in above cells. DISCUSSION In patients experiencing RSA, abnormal inflammatory response might be mediated by NLRP3 inflammasomes on the maternal-foetal interface, which may reduce trophoblast activity and promote macrophage activity, leading to early embryo implantation failure. LMWH is expected to treat RSA patients by blocking this process.
Collapse
Affiliation(s)
- Peng Gao
- Department of Obstetrics, Department of Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Ying Zha
- Department of Obstetrics, Department of Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Xun Gong
- Department of Obstetrics, Department of Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Fuyuan Qiao
- Department of Obstetrics, Department of Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Haiyi Liu
- Department of Obstetrics, Department of Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China.
| |
Collapse
|
11
|
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
|
12
|
Banadakoppa M, Pennington K, Balakrishnan M, Yallampalli C. Complement inhibitor Crry expression in mouse placenta is essential for maintaining normal blood pressure and fetal growth. PLoS One 2020; 15:e0236968. [PMID: 32745140 PMCID: PMC7398533 DOI: 10.1371/journal.pone.0236968] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 07/16/2020] [Indexed: 11/18/2022] Open
Abstract
Many circumstantial evidences from human and animal studies suggest that complement cascade dysregulation may play an important role in pregnancy associated complications including preeclampsia. Deletion of rodent specific complement inhibitor gene, Complement Receptor 1-related Gene/Protein y (Crry) produces embryonic lethal phenotype due to complement activation. It is not clear if decreased expression of Crry during pregnancy produces hypertensive phenotype. We downregulated Crry in placenta by injecting inducible lentivialshRNA vectors into uterine horn of pregnant C57BL/6 mice at the time of blastocyst hatching. Placenta specific downregulation of Crry without significant loss of embryos was achieved upon induction of shRNA using an optimal doxycycline dose at mid gestation. Crry downregulation resulted in placental complement deposition. Late-gestation measurements showed that fetal weights were reduced and blood pressure increased in pregnant mice upon downregulation of Crry suggesting a critical role for Crry in fetal growth and blood pressure regulation.
Collapse
Affiliation(s)
- Manu Banadakoppa
- Department of Obstetrics & Gynecology, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail: (MB); (CY)
| | - Kathleen Pennington
- Department of Obstetrics & Gynecology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Meena Balakrishnan
- Department of Obstetrics & Gynecology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Chandra Yallampalli
- Department of Obstetrics & Gynecology, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail: (MB); (CY)
| |
Collapse
|
13
|
Koucký M, Malíčková K, Kopřivová H, Cindrová-Davies T, Čapek V, Pařízek A. Serum mannose-binding lectin (MBL) concentrations are reduced in non-pregnant women with previous adverse pregnancy outcomes. Scand J Immunol 2020; 92:e12892. [PMID: 32335925 DOI: 10.1111/sji.12892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 03/27/2020] [Accepted: 04/20/2020] [Indexed: 11/28/2022]
Abstract
Mannose-binding lectin (MBL) is an important component of the innate immunity, and it is responsible not only for opsonization of micro-organisms, but also for efferocytosis. The aim of this study was to investigate whether MBL concentrations and lectin complement pathway activity are altered in non-pregnant women with previous adverse pregnancy outcomes. Patients were divided into four groups on the basis of their history of pregnancy complications, including control patients who had uncomplicated pregnancies and term deliveries (control, n = 33), and three groups of patients with a history of pregnancy complications, including preterm labour (n = 29), recurrent miscarriage (n = 19) or unexplained intrauterine foetal death (IUFD; n = 17). All women enrolled in the study had an interval of three to six months following their previous pregnancy, and they agreed to have a blood sample taken. We found significantly higher MBL concentrations and functional activity of the lectin complement pathway in healthy controls who had previous uneventful term pregnancies (1341 ng/mL; activity 100% (IQR: 62%-100%)), compared to women with the history of IUFD (684 ng/mL, P = .008; activity 8.5% (IQR: 0%-97.8%), P = .011), recurrent miscarriage (524 ng/mL, P = .022; activity 44% (IQR: 4%-83%), P = .011) or preterm labour (799 ng/mL, P = .022; activity 62.5% (IQR: 0%-83%), P = .003). Our results suggest that inadequate function of the complement lectin pathway is associated with a higher risk of preterm labour, recurrent miscarriage and unexplained intrauterine foetal death.
Collapse
Affiliation(s)
- Michal Koucký
- Department of Gynecology and Obstetrics of the First Faculty of Medicine and General Teaching Hospital, Charles University in Prague, Prague, Czech Republic
| | - Karin Malíčková
- Institute of Medical Biochemistry and Laboratory Diagnostics of the First Faculty of Medicine and General Teaching Hospital, Charles University in Prague, Prague, Czech Republic
| | - Helena Kopřivová
- Institute of Medical Biochemistry and Laboratory Diagnostics of the First Faculty of Medicine and General Teaching Hospital, Charles University in Prague, Prague, Czech Republic
| | - Tereza Cindrová-Davies
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | | | - Antonín Pařízek
- Department of Gynecology and Obstetrics of the First Faculty of Medicine and General Teaching Hospital, Charles University in Prague, Prague, Czech Republic
| |
Collapse
|
14
|
Sharma M, Vignesh P, Tiewsoh K, Rawat A. Revisiting the complement system in systemic lupus erythematosus. Expert Rev Clin Immunol 2020; 16:397-408. [PMID: 32228236 DOI: 10.1080/1744666x.2020.1745063] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Introduction: Systemic lupus erythematosus (SLE) is a multi-system autoimmune disease, characterized by the production of autoantibodies. Numerous mechanisms contribute to the pathogenesis and autoimmunity in SLE. One of the most important mechanisms is the defective function of the early complement components that are involved in clearing the immune-complexes and apoptotic debris. Major evidence supporting this hypothesis is the development of severe lupus in individuals with monogenic defects in any one of the early complement components such as C1q, C1 s, C1 r, C2, or C4.Areas covered: In this review, we discuss hereditary defects in classical complement components and their clinical manifestations, acquired defects of complements in lupus, the role of complements in the pathogenesis of antiphospholipid antibody syndrome and lupus nephritis, and laboratory assessment of complement components and their functions. Articles from the last 20 years were retrieved from PubMed for this purpose.Expert opinion: Complements have a dual role in the pathogenesis of SLE. On one hand, deficiency of complement components predisposes to lupus, while, on the other, excess complement activation plays a role in the organ damage. Understanding the intricacies of the role of complements in SLE can pave way for the development of targeted therapies.
Collapse
Affiliation(s)
- Madhubala Sharma
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Pandiarajan Vignesh
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Karalanglin Tiewsoh
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Amit Rawat
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
15
|
De Lorenzo R, Ramirez GA, Punzo D, Lorioli L, Rovelli R, Canti V, Barera G, Rovere-Querini P. Neonatal outcomes of children born to mothers on biological agents during pregnancy: State of the art and perspectives. Pharmacol Res 2019; 152:104583. [PMID: 31816434 DOI: 10.1016/j.phrs.2019.104583] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 11/29/2019] [Accepted: 11/30/2019] [Indexed: 12/19/2022]
Abstract
Biologic disease-modifying anti-rheumatic drugs (bDMARDs) are used in pregnant patients with rheumatic diseases. Long-term follow-up data about newborns exposed to bDMARDs during pregnancy are however scarce. Here we summarize the published evidence and available recommendations for use of bDMARDs during pregnancy. We analyse clinical features at birth and at follow-up of 84 children, including: 16 consecutive children born to mothers with autoimmune diseases exposed to bDMARDs in utero; 32 children born to mothers with autoimmune diseases who did not receive bDMARDs; 36 children born to healthy mothers. In our monocentric cohort, children born to mothers with autoimmune diseases had lower gestational age at birth compared to those born to healthy mothers, independently of exposure to bDMARDs. At multivariate analysis, prematurity was an independent predictor of the need for antibiotic treatment, but not for hospitalisation or neonatal intensive care unit (ICU) stay during the neonatal period. Exposure to bDMARDs during pregnancy does not seem to interfere with post-natal development up to infancy. Prospective studies are needed in larger cohorts of pregnant patients to confirm that bDMARDs do not have a negative impact on psychomotor achievements in newborns.
Collapse
Affiliation(s)
- Rebecca De Lorenzo
- Università Vita-Salute San Raffaele, Milan, Italy; Department of Medicine and Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Giuseppe A Ramirez
- Università Vita-Salute San Raffaele, Milan, Italy; Department of Medicine and Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Daniele Punzo
- Università Vita-Salute San Raffaele, Milan, Italy; Department of Medicine and Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Laura Lorioli
- Unit of Paediatrics, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Rosanna Rovelli
- Unit of Paediatrics, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Valentina Canti
- Università Vita-Salute San Raffaele, Milan, Italy; Department of Medicine and Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Graziano Barera
- Unit of Paediatrics, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Patrizia Rovere-Querini
- Università Vita-Salute San Raffaele, Milan, Italy; Department of Medicine and Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy.
| |
Collapse
|
16
|
Regulation of the complement system and immunological tolerance in pregnancy. Semin Immunol 2019; 45:101337. [PMID: 31757607 DOI: 10.1016/j.smim.2019.101337] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 10/24/2019] [Indexed: 12/18/2022]
Abstract
Preeclampsia is a serious vascular complication of the human pregnancy, whose etiology is still poorly understood. In preeclampsia, exacerbated apoptosis and fragmentation of the placental tissue occurs due to developmental qualities of the placental trophoblast cells and/or mechanical and oxidative distress to the syncytiotrophoblast, which lines the placental villi. Dysregulation of the complement system is recognized as one of the mechanisms of the disease pathology. Complement has the ability to promote inflammation and facilitate phagocytosis of placenta-derived particles and apoptotic cells by macrophages. In preeclampsia, an overload of placental cell damage or dysregulated complement system may lead to insufficient clearance of apoptotic particles and placenta-derived debris. Excess placental damage may lead to sequestration of microparticles, such as placental vesicles, to capillaries in the glomeruli of the kidney and other vulnerable tissues. This phenomenon could contribute to the manifestations of typical diagnostic symptoms of preeclampsia: proteinuria and new-onset hypertension. In this review we propose that the complement system may serve as a regulator of the complex tolerance and clearance processes that are fundamental in healthy pregnancy. It is therefore recommended that further research be conducted to elucidate the interactions between components of the complement system and immune responses in the context of complicated and healthy pregnancy.
Collapse
|
17
|
Lee M, Lee HA, Park M, Park HK, Kim YS, Yang SC, Kim HR, Kim J, Song H. Oestrogen-induced expression of decay accelerating factor is spatiotemporally antagonised by progesterone-progesterone receptor signalling in mouse uterus. Reprod Fertil Dev 2019; 30:1532-1540. [PMID: 29852923 DOI: 10.1071/rd18031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 04/23/2018] [Indexed: 12/23/2022] Open
Abstract
Decay accelerating factor (DAF) is upregulated in the fetoplacental trophoblast, which protects the fetus from maternal complement injury. DAF was found to be downregulated in the endometrium of patients with repeated implantation failure. Thus, we examined the molecular mechanisms of DAF expression regulation by ovarian steroid hormones in the mouse uterus. Immunofluorescence staining demonstrated its exclusive localisation in the apical region of the epithelium in the uterus. Oestrogen (E2) significantly induced Daf mRNA in a time-dependent manner. Progesterone (P4) did not have any significant effect on Daf expression; however, it negatively modulated E2-induced DAF expression and RU486 effectively interfered with the inhibitory action of P4 in the uterus. During early pregnancy DAF was higher on Day 1 of pregnancy, but significantly decreased from Day 3, which is consistent with its E2-dependent regulation. Interestingly, DAF expression seemed to be influenced by the implanting blastocyst on Day 5 and it was gradually increased during preimplantation embryo development with peak levels at blastocyst stages. We demonstrated that E2-dependent DAF expression is antagonised by P4-progesterone receptor signalling in the uterine epithelium. Spatiotemporal regulation of DAF in the uterus and preimplantation embryos suggest that DAF functions as an immune modulator for embryo implantation and early pregnancy in mice.
Collapse
Affiliation(s)
- Miji Lee
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Hyang Ah Lee
- Department of Obstetrics and Gynaecology, Kangwon National University School of Medicine, Chuncheon-si, Gangwon-do, 24341, Republic of Korea
| | - Mira Park
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Hee Kyoung Park
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Yeon Sun Kim
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Seung Chel Yang
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Hye-Ryun Kim
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Jayeon Kim
- CHA Fertility Centre Seoul Station, CHA University, Seoul, 04637, Republic of Korea
| | - Haengseok Song
- Department of Biomedical Science, CHA University, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| |
Collapse
|
18
|
Di Nicuolo F, D'Ippolito S, Castellani R, Rossi ED, Masciullo V, Specchia M, Mariani M, Pontecorvi A, Scambia G, Di Simone N. Effect of alpha-lipoic acid and myoinositol on endometrial inflammasome from recurrent pregnancy loss women. Am J Reprod Immunol 2019; 82:e13153. [PMID: 31148259 DOI: 10.1111/aji.13153] [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: 11/03/2018] [Revised: 04/05/2019] [Accepted: 05/24/2019] [Indexed: 12/22/2022] Open
Abstract
PROBLEM A significant increased expression/activation of one of the most well-characterized inflammasomes, the NAcht leucine-rich-repeat protein-3 (NALP-3), in the endometrium from idiopathic recurrent pregnancy loss women (RPL) has been previously found by our research group. We therefore, suggested this event as being one of the molecular mechanisms altering endometrial inflammatory status during early pregnancy. In the present research, we attempt to investigate whether molecules with anti-inflammatory activity, alpha-lipoic acid (ALA), and/or myoinositol affect the endometrial NALP-3 expression and activation. METHOD OF STUDY Women with a history of idiopathic RPL (n = 30) were included in the study and compared to a control group (n = 15). Endometrial tissues were collected by hysteroscopy during the mid-luteal phase. RPL women underwent a three-month prescription of tablets containing ALA plus myoinositol (Sinopol® ). After treatment, hysteroscopic biopsies were repeated in RPL patients. Inflammasome expression was evaluated by immunohistochemical and Western blot analysis. NALP-3 activation was studied by quantifying the secretion of both caspase-1 and interleukin (IL)-1ß and IL-18 through ELISA. In ex vivo experiments, the effects of each molecule on endometrial inflammasome were studied. RESULTS Sinopol® significantly reduced the RPL endometrial inflammasome expression and activation. ALA, but not myoinositol, significantly reduced the endometrial inflammasome expression and activity. CONCLUSION Our data suggest a role for ALA on RPL inflammasome. Understanding the mechanisms involved in RPL and the observation that specific molecules are able to interfere with such complex at the endometrium might provide new rational design approaches to a personalized evaluation of endometrial status and, ultimately, a targeted medicine.
Collapse
Affiliation(s)
- Fiorella Di Nicuolo
- Paolo VI International Scientific Institute, Università Cattolica del Sacro Cuore, Roma, Italia
| | - Silvia D'Ippolito
- U.O.C. di Ostetricia e Patologia Ostetrica, Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia.,Istituto di Clinica Ostetrica e Ginecologica, Università Cattolica del Sacro Cuore, Roma, Italia
| | - Roberta Castellani
- Istituto di Clinica Ostetrica e Ginecologica, Università Cattolica del Sacro Cuore, Roma, Italia
| | - Esther Diana Rossi
- U.O.C. di Anatomia Patologica, Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia
| | - Valeria Masciullo
- Istituto di Clinica Ostetrica e Ginecologica, Università Cattolica del Sacro Cuore, Roma, Italia.,U.O.C. di Ginecologia Oncologica, Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia
| | - Monia Specchia
- Istituto di Clinica Ostetrica e Ginecologica, Università Cattolica del Sacro Cuore, Roma, Italia
| | - Marco Mariani
- Istituto di Sanità Pubblica, Sezione di Igiene, Università Cattolica Del Sacro Cuore, Roma, Italia
| | - Alfredo Pontecorvi
- Paolo VI International Scientific Institute, Università Cattolica del Sacro Cuore, Roma, Italia.,U.O.C di Endocrinologia e Diabetologia, Dipartimento di Scienze Gastroenterologiche, Endocrino-Metaboliche e Nefro-Urologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia.,Istituto di Patologia Medica, Università Cattolica del Sacro Cuore, Roma, Italia
| | - Giovanni Scambia
- Istituto di Clinica Ostetrica e Ginecologica, Università Cattolica del Sacro Cuore, Roma, Italia.,U.O.C. di Ginecologia Oncologica, Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia
| | - Nicoletta Di Simone
- U.O.C. di Ostetricia e Patologia Ostetrica, Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia.,Istituto di Clinica Ostetrica e Ginecologica, Università Cattolica del Sacro Cuore, Roma, Italia
| |
Collapse
|
19
|
Kumar V. The complement system, toll-like receptors and inflammasomes in host defense: three musketeers’ one target. Int Rev Immunol 2019; 38:131-156. [PMID: 31066339 DOI: 10.1080/08830185.2019.1609962] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Vijay Kumar
- Children’s Health Queensland Clinical Unit, School of Clinical Medicine, Faculty of Medicine, Mater Research, University of Queensland, St Lucia, Brisbane, QLD, Australia
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, ST Lucia, Brisbane, QLD, Australia
| |
Collapse
|
20
|
Pillay Y, Moodley J, Naicker T. The role of the complement system in HIV infection and preeclampsia. Inflamm Res 2019; 68:459-469. [PMID: 31028431 DOI: 10.1007/s00011-019-01240-0] [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: 03/28/2019] [Revised: 04/18/2019] [Accepted: 04/22/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND The complement system is a key component of the innate immune system that plays a vital role in host defense, maintains homeostasis and acts as a mediator of the adaptive immune response. The complement system could possibly play a role in the pathogenesis of HIV infection and preeclampsia (PE), both of which represent major causes of maternal death in South Africa. RECENT FINDINGS The relationship between PE and HIV infection is unclear as PE represents an exaggerated immune response, while HIV infection is associated with a decline in immune activity. Although the complement system works to clear and neutralize HIV, it could also enhance the infectivity of HIV by various other mechanisms. It has been suggested that the dysregulation of the complement system is associated with the development of PE. CONCLUSION There is currently a paucity of information on the combined effect of the complement system in HIV-associated PE. This review highlights the role of the complement system in the duality of HIV infection and PE and provides new insights into this relationship whilst also elucidating potential therapeutic targets.
Collapse
Affiliation(s)
- Yazira Pillay
- Optics and Imaging Centre, University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa.
| | - Jagidesa Moodley
- Women's Health and HIV Research Group, University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa
| | - Thajasvarie Naicker
- Optics and Imaging Centre, University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa
| |
Collapse
|
21
|
Bastu E, Demiral I, Gunel T, Ulgen E, Gumusoglu E, Hosseini MK, Sezerman U, Buyru F, Yeh J. Potential Marker Pathways in the Endometrium That May Cause Recurrent Implantation Failure. Reprod Sci 2018; 26:879-890. [PMID: 30081718 DOI: 10.1177/1933719118792104] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The aim of this prospective cohort study was to identify altered biologic processes in the endometrium that may be potential markers of receptive endometrium in patients with repeated implantation failure (RIF) as compared with fertile controls. The study was conducted in a university-affiliated in vitro fertilization (IVF) gynecology clinic and molecular biology and genetics laboratory. Healthy fertile controls (n = 24) and patients with RIF (n = 24) were recruited. Window of implantation gene profiling associated with RIF was performed. Six hundred forty-one differentially expressed genes were identified, and 44 pathways were found enriched. Upon clustering of the enriched pathways, 9 representative pathways were established. The important pathways that were identified included circadian rhythm, pathways in cancer, proteasome, complement and coagulation cascades, citrate cycle, adherens junction, immune system and inflammation, cell cycle, and renin-angiotensin system. The involvement of the circadian rhythm pathway and other related pathways may alter the endometrium's functioning to ultimately cause RIF. Furthermore, we found that the pathogenesis of RIF was multifaceted and that numerous processes were involved. We believe that a better understanding of the underlying mechanisms of RIF will ultimately give rise to better treatment opportunities and to better outcomes in IVF.
Collapse
Affiliation(s)
- Ercan Bastu
- Department of Obstetrics and Gynecology, Acibadem University School of Medicine, Ataşehir, Istanbul, Turkey.
| | - Irem Demiral
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard University, Boston, MA, USA
| | - Tuba Gunel
- Department of Molecular Biology and Genetics, Istanbul University, Istanbul, Turkey
| | - Ege Ulgen
- Department of Biostatistics and Medical Informatics, Acibadem University School of Medicine, Istanbul, Turkey
| | - Ece Gumusoglu
- Department of Molecular Biology and Genetics, Istanbul University, Istanbul, Turkey
| | | | - Ugur Sezerman
- Department of Biostatistics and Medical Informatics, Acibadem University School of Medicine, Istanbul, Turkey
| | - Faruk Buyru
- Department of Obstetrics and Gynecology, Acibadem University School of Medicine, Ataşehir, Istanbul, Turkey
| | - John Yeh
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard University, Boston, MA, USA
| |
Collapse
|
22
|
Abdulrahman Ahmad H, Muhammd salih MM, Ahmed Khidir K. Complement protein and Immunoglobulins Serum levels in Normal Pregnant and Spontaneous Aborted Women. KURDISTAN JOURNAL OF APPLIED RESEARCH 2018:129-133. [DOI: 3.https:/doi.org/10.24017/science.2018.2.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/01/2024] Open
Abstract
Disorder of maternal immune responses during pregnancy triggers immunological rejection of fetus antigens by maternal immune components, contribute to spontaneous abortion or miscarriage. The study was designed to concentrated on immunoglobulins (IgM, IgG and IgA) and complement elements (C3 and C4) serum levels changes in normal pregnant and abortion women. Study groups were classified into normal pregnant women (20), spontaneous abortion (30) and non-pregnant women (16) as a control group, attending to Shahid Dr. Khalid Hospital/Department of Gynecology and Obstetrics/Koya city. Serum levels of immunoglobulins (IgG, IgM, and IgA), complement proteins (C3 and C4) were determined and analyzed for normal pregnant, abortion and control groups by using Single Radial Immunodiffusion (SRID) technique. The results demonstrated that concentration of IgG levels in abortion differed significantly in compare to normal pregnancy (p ≤0.05), while there were no significant differences in IgM and IgA serum levels among groups (p >0.05). Also, statistical analysis revealed that serum levels of C3 and C4 significantly decreased in abortion group compared to normal pregnant and non-pregnant groups (p ≤0.05). Concluded that complement proteins (C3 andC4) are a good defense line during normal pregnancy, sometime activation (hyper-consuming) of complement elements may provoke spontaneous abortion, while immunoglobulins are a little role in inducing of miscarriage in pregnant women.
Collapse
|
23
|
Tedesco F, Borghi MO, Gerosa M, Chighizola CB, Macor P, Lonati PA, Gulino A, Belmonte B, Meroni PL. Pathogenic Role of Complement in Antiphospholipid Syndrome and Therapeutic Implications. Front Immunol 2018; 9:1388. [PMID: 29971066 PMCID: PMC6018396 DOI: 10.3389/fimmu.2018.01388] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 06/05/2018] [Indexed: 12/17/2022] Open
Abstract
Antiphospholipid syndrome (APS) is an acquired autoimmune disease characterized by thromboembolic events, pregnancy morbidity, and the presence of antiphospholipid (aPL) antibodies. There is sound evidence that aPL act as pathogenic autoantibodies being responsible for vascular clots and miscarriages. However, the exact mechanisms involved in the clinical manifestations of the syndrome are still a matter of investigation. In particular, while vascular thrombosis is apparently not associated with inflammation, the pathogenesis of miscarriages can be explained only in part by the aPL-mediated hypercoagulable state and additional non-thrombotic effects, including placental inflammation, have been described. Despite this difference, evidence obtained from animal models and studies in APS patients support the conclusion that complement activation is a common denominator in both vascular and obstetric APS. Tissue-bound aPL rather than circulating aPL-beta2 glycoprotein I immune complexes seem to be responsible for the activation of the classical and the alternative complement pathways. The critical role of complement is supported by the finding that complement-deficient animals are protected from the pathogenic effect of passively infused aPL and similar results have been obtained blocking complement activation. Moreover, elevated levels of complement activation products in the absence of abnormalities in regulatory molecules have been found in the plasma of APS patients, strongly suggesting that the activation of complement cascade is the result of aPL binding to the target antigen rather than of a defective regulation. Placental complement deposits represent a further marker of complement activation both in animals and in patients, and there is also some suggestive evidence that complement activation products are deposited in the affected vessels. The aim of this review is to analyze the state of the art of complement involvement in the pathogenesis of APS in order to provide insights into the role of this system as predictive biomarker for the clinical manifestations and as therapeutic target.
Collapse
Affiliation(s)
- Francesco Tedesco
- Immunorheumatology Research Laboratory, Istituto Auxologico Italiano, Milan, Italy
| | - Maria Orietta Borghi
- Immunorheumatology Research Laboratory, Istituto Auxologico Italiano, Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Maria Gerosa
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Cecilia Beatrice Chighizola
- Immunorheumatology Research Laboratory, Istituto Auxologico Italiano, Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Paolo Macor
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Paola Adele Lonati
- Immunorheumatology Research Laboratory, Istituto Auxologico Italiano, Milan, Italy
| | - Alessandro Gulino
- Tumor Immunology Unit, Human Pathology Section, Department of Health Sciences, University of Palermo, Palermo, Italy
| | - Beatrice Belmonte
- Tumor Immunology Unit, Human Pathology Section, Department of Health Sciences, University of Palermo, Palermo, Italy
| | - Pier Luigi Meroni
- Immunorheumatology Research Laboratory, Istituto Auxologico Italiano, Milan, Italy
| |
Collapse
|
24
|
Garrett N, Pombo J, Umpierrez M, Clark JE, Simmons M, Girardi G. Pravastatin therapy during preeclampsia prevents long-term adverse health effects in mice. JCI Insight 2018; 3:120147. [PMID: 29669946 DOI: 10.1172/jci.insight.120147] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 03/09/2018] [Indexed: 12/30/2022] Open
Abstract
Preeclampsia (PE), associates with long-term increased risk for cardiovascular disease in women, suggesting that PE is not an isolated disease of pregnancy. It is not known if increased risk for long-term diseases is due to PE-specific factors or to prepregnancy renal and cardiovascular risk factors. We used a mouse model in which a WT female with normal prepregnancy health develops PE to investigate if preeclampsia causes long-term cardiovascular consequences after pregnancy for mothers and offspring. Mothers exhibited endothelial dysfunction and hypertension after PE and had glomerular injury that not only persisted but deteriorated, leading to fibrosis. Left ventricular (LV) remodeling characterized by increased collagen deposition and MMP-9 expression and enlarged cardiomyocytes were also detected after PE. Increased LV internal wall thickness and mass, increased end diastolic and end systolic volumes, and increased stroke volume were observed after PE in the mothers. Placenta-derived bioactive factors that modulate vascular function, markers of metabolic disease, vasoconstrictor isoprostane-8, and proinflammatory mediators were increased in sera during and after a preeclamptic pregnancy in the mother. Offspring of PE mice developed endothelial dysfunction, hypertension, and signs of metabolic disease. Microglia activation was increased in the neonatal brains after PE, suggesting neurogenic hypertension in offspring. Prevention of placental insufficiency with pravastatin prevented PE-associated cardiovascular complications in both mothers and offspring. In conclusion, factors that develop during PE have long-term, cardiovascular effects in the mother and offspring independent of prepregnancy risk factors.
Collapse
Affiliation(s)
- Nicola Garrett
- Pregnancy Laboratory, Department of Women and Children's Health, Rayne Institute, St Thomas' Hospital, King's College London, London, United Kingdom
| | - Joaquim Pombo
- Pregnancy Laboratory, Department of Women and Children's Health, Rayne Institute, St Thomas' Hospital, King's College London, London, United Kingdom
| | - Michelle Umpierrez
- Pregnancy Laboratory, Department of Women and Children's Health, Rayne Institute, St Thomas' Hospital, King's College London, London, United Kingdom
| | - James E Clark
- King's College London BHF Cardiovascular Centre, Rayne Institute, St Thomas' Hospital, London, United Kingdom
| | - Mark Simmons
- Pregnancy Laboratory, Department of Women and Children's Health, Rayne Institute, St Thomas' Hospital, King's College London, London, United Kingdom
| | - Guillermina Girardi
- Pregnancy Laboratory, Department of Women and Children's Health, Rayne Institute, St Thomas' Hospital, King's College London, London, United Kingdom.,Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
25
|
Thielens NM, Tedesco F, Bohlson SS, Gaboriaud C, Tenner AJ. C1q: A fresh look upon an old molecule. Mol Immunol 2017; 89:73-83. [PMID: 28601358 DOI: 10.1016/j.molimm.2017.05.025] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 05/27/2017] [Accepted: 05/29/2017] [Indexed: 12/14/2022]
Abstract
Originally discovered as part of C1, the initiation component of the classical complement pathway, it is now appreciated that C1q regulates a variety of cellular processes independent of complement activation. C1q is a complex glycoprotein assembled from 18 polypeptide chains, with a C-terminal globular head region that mediates recognition of diverse molecular structures, and an N-terminal collagen-like tail that mediates immune effector mechanisms. C1q mediates a variety of immunoregulatory functions considered important in the prevention of autoimmunity such as the enhancement of phagocytosis, regulation of cytokine production by antigen presenting cells, and subsequent alteration in T-lymphocyte maturation. Furthermore, recent advances indicate additional roles for C1q in diverse physiologic and pathologic processes including pregnancy, tissue repair, and cancer. Finally, C1q is emerging as a critical component of neuronal network refinement and homeostatic regulation within the central nervous system. This review summarizes the classical functions of C1q and reviews novel discoveries within the field.
Collapse
Affiliation(s)
| | - Francesco Tedesco
- Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Auxologico Italiano, Milan, Italy
| | | | | | | |
Collapse
|
26
|
Clark DA. The importance of being a regulatory T cell in pregnancy. J Reprod Immunol 2016; 116:60-9. [DOI: 10.1016/j.jri.2016.04.288] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 04/18/2016] [Accepted: 04/19/2016] [Indexed: 12/16/2022]
|
27
|
C1q acts in the tumour microenvironment as a cancer-promoting factor independently of complement activation. Nat Commun 2016; 7:10346. [PMID: 26831747 PMCID: PMC4740357 DOI: 10.1038/ncomms10346] [Citation(s) in RCA: 210] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 12/02/2015] [Indexed: 02/06/2023] Open
Abstract
Complement C1q is the activator of the classical pathway. However, it is now recognized that C1q can exert functions unrelated to complement activation. Here we show that C1q, but not C4, is expressed in the stroma and vascular endothelium of several human malignant tumours. Compared with wild-type (WT) or C3- or C5-deficient mice, C1q-deficient (C1qa−/−) mice bearing a syngeneic B16 melanoma exhibit a slower tumour growth and prolonged survival. This effect is not attributable to differences in the tumour-infiltrating immune cells. Tumours developing in WT mice display early deposition of C1q, higher vascular density and an increase in the number of lung metastases compared with C1qa−/− mice. Bone marrow (BM) chimeras between C1qa−/− and WT mice identify non-BM-derived cells as the main local source of C1q that can promote cancer cell adhesion, migration and proliferation. Together these findings support a role for locally synthesized C1q in promoting tumour growth. C1q is known to initiate the activation of the complement classical pathway. Here, the authors show the C1q is expressed in the tumour microenvironment and can promote cancer cell migration and adhesion in a complement activation-independent manner.
Collapse
|
28
|
Lu J, Zhou WH, Ren L, Zhang YZ. CXCR4, CXCR7, and CXCL12 are associated with trophoblastic cells apoptosis and linked to pathophysiology of severe preeclampsia. Exp Mol Pathol 2015; 100:184-91. [PMID: 26721717 DOI: 10.1016/j.yexmp.2015.12.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 12/20/2015] [Accepted: 12/20/2015] [Indexed: 11/26/2022]
Abstract
Preeclampsia is a pregnancy disorder with sudden onset of maternal hypertension and proteinuria, which is characterized by defective cytotrophoblast invasion, increased apoptosis in cytotrophoblast, and diminished syncytial differentiation. In this study, samples from 11 mild preeclamptic patients, 18 severe preeclamptic patients, and 21 normal pregnant women were collected. The expression level of CXCL12 and its two receptors (CXCR4 and CXCR7) in these samples and their relationship with apoptosis were investigated. Morphological change of trophoblast cells that was observed by scanning electron microscope (SEM) indicated a significant tendency of apoptosis in the preeclamptic placenta. Immunohistochemical staining showed that expression level of three proteins was significantly lower in severe preeclamptic placentas compared with normal placentas (P<0.05), whereas no significant difference was found between mild preeclamptic and normal placentas (P>0.05). Real time quantitative PCR (RT-qPCR) and Western blot showed that both mRNA and protein expression level of CXCR4, CXCR7, and CXCL12 of trophoblasts were lower in the severe preeclampsia group than that in the normal group (P<0.05 for mRNA, P<0.01 for protein). In conclusion, our data revealed that the roles of CXCR4, CXCR7, and CXCL12 are associated with trophoblastic cells apoptosis and may be linked to the occurrence and development of severe preeclampsia.
Collapse
Affiliation(s)
- Jing Lu
- Department of Obstetrics and Gynecology, Medicine Center for Human Reproduction, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, 430071, People's Republic of China
| | - Wen-Hui Zhou
- Medicine Center for Human Reproduction, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China
| | - Liang Ren
- Department of Obstetrics and Gynecology, Medicine Center for Human Reproduction, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, 430071, People's Republic of China
| | - Yuan-Zhen Zhang
- Department of Obstetrics and Gynecology, Medicine Center for Human Reproduction, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, 430071, People's Republic of China.
| |
Collapse
|
29
|
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]
|
30
|
D'Ippolito S, Tersigni C, Marana R, Di Nicuolo F, Gaglione R, Rossi ED, Castellani R, Scambia G, Di Simone N. Inflammosome in the human endometrium: further step in the evaluation of the "maternal side". Fertil Steril 2015; 105:111-8.e1-4. [PMID: 26474737 DOI: 10.1016/j.fertnstert.2015.09.027] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 09/11/2015] [Accepted: 09/18/2015] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To investigate the expression of inflammosome components (NALP-3, associated speck-like protein containing a CARD [ASC]) and their activation (caspase-1, interleukin [IL]-1β, and IL-18 secretion) in the human endometrium from fertile and women with history of recurrent pregnancy loss (RPL). DESIGN Experimental study. SETTING University hospital. PATIENT(S) Ten fertile women (control group [CTR]) and 30 women with RPL. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) Endometrial samples were collected by hysteroscopy during the putative window of implantation and evaluated for chronic endometrial inflammation by hystopathological analysis. Inflammosome expression was analysed by immunohystochemical staining (27 RPL and 10 CTR women). The expression of NALP-3 and ASC protein was quantified by Western blot (30 RPL and 10 CTR women). Caspase-1 activation and IL-1β and IL-18 secretion was quantified by ELISA (30 RPL and 10 CTR women). RESULT(S) We observed a significantly increased expression of inflammasome NALP-3 and ASC protein, an increased activation of caspase-1, and increased levels of IL-1β and IL-18 in RPL endometrium compared with CTR. CONCLUSION(S) Abnormal activation of endometrial innate immunity by means of inflammosome, stimulated by pathogen- or damage-associated molecular patterns, may represent an additional mechanism, currently not investigated, negatively interfering with endometrial receptivity. More studies are required [1] to identify the primary trigger of endometrial inflammosome activation and its clinical impact in the occurrence of RPL; and [2] to validate the inflammosome components as a novel family of endometrial biomarkers and promising therapeutic targets in RPL.
Collapse
Affiliation(s)
- Silvia D'Ippolito
- Department of Obstetrics and Gynecology, Università Cattolica Del Sacro Cuore, A. Gemelli Universitary Hospital, Rome, Italy
| | - Chiara Tersigni
- Department of Obstetrics and Gynecology, Università Cattolica Del Sacro Cuore, A. Gemelli Universitary Hospital, Rome, Italy
| | - Riccardo Marana
- Department of Obstetrics and Gynecology, Università Cattolica Del Sacro Cuore, A. Gemelli Universitary Hospital, Rome, Italy; International Scientific Institute Paolo VI, ISI, Università Cattolica Del Sacro Cuore, A. Gemelli Universitary Hospital, Rome, Italy
| | - Fiorella Di Nicuolo
- Department of Obstetrics and Gynecology, Università Cattolica Del Sacro Cuore, A. Gemelli Universitary Hospital, Rome, Italy
| | - Raffaele Gaglione
- Department of Obstetrics and Gynecology, Università Cattolica Del Sacro Cuore, A. Gemelli Universitary Hospital, Rome, Italy
| | - Esther Diana Rossi
- Division of Anatomic Pathology and Histology, Università Cattolica Del Sacro Cuore, A. Gemelli Universitary Hospital, Rome, Italy
| | - Roberta Castellani
- Department of Obstetrics and Gynecology, Università Cattolica Del Sacro Cuore, A. Gemelli Universitary Hospital, Rome, Italy
| | - Giovanni Scambia
- Department of Obstetrics and Gynecology, Università Cattolica Del Sacro Cuore, A. Gemelli Universitary Hospital, Rome, Italy
| | - Nicoletta Di Simone
- Department of Obstetrics and Gynecology, Università Cattolica Del Sacro Cuore, A. Gemelli Universitary Hospital, Rome, Italy.
| |
Collapse
|
31
|
Weghofer A, Himaya E, Kushnir VA, Barad DH, Lazzaroni-Tealdi E, Yu Y, Wu YG, Gleicher N. Some aspects of interactivity between endocrine and immune systems required for successful reproduction. Reprod Biol Endocrinol 2015; 13:29. [PMID: 25889294 PMCID: PMC4399251 DOI: 10.1186/s12958-015-0020-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 03/15/2015] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND In successful reproduction, endocrine and immune systems closely interact. We here attempt to further elucidate the relationship between androgen levels, systemic activation of the immune system and reproductive success in infertile women, utilizing 2 distinct infertile patient cohorts. METHODS In Group 1, we investigated 322 women (ages 38.6+/-5.4 years) at initial presentation; in Group 2 125 women undergoing in vitro fertilization (169 IVF cycles, ages 38.9+/-5.5 years). In Group 1, we assessed androgens and an immune panel, previously demonstrated to discriminate between activated quiescent immune systems; in Group 2, utilizing the same immune panel, we investigated whether immune system activation relates to embryo quality in IVF cycles. RESULTS No individual immune test within the immune panel was associated with androgen levels. The total/free testosterone ratio (TT/FT) was, however, significantly associated with presence of gammopathies (in IgG, IgM, IgA, IgE; P=0.026). Surprisingly, immune system activation was associated with significantly improved embryo quality (P=0.008), a finding persistent after adjustment for age and repeat IVF cycles (P=0.006). CONCLUSIONS Association of immune system activation with improved embryo quality concurs with previously reported immune activation in association with normal functional ovarian reserve (FOR) and normal androgen levels, while, counter intuitively, hypoandrogenism and low FOR are associated with lack of immune system activation. Mild immune system activation, therefore, likely appears essential for establishment of pregnancy, and may be regulated by androgens.
Collapse
Affiliation(s)
- Andrea Weghofer
- Center for Human Reproduction, New York, NY, USA.
- Department of Obstetrics and Gynecology, Medical University Vienna, Vienna, Austria.
| | - Eric Himaya
- Center for Human Reproduction, New York, NY, USA.
- Centre Hospitalier de l'Université de Montréal, Montreal, Canada.
| | | | - David H Barad
- Center for Human Reproduction, New York, NY, USA.
- Foundation for Reproductive Medicine, New York, NY, USA.
| | | | - Yao Yu
- Center for Human Reproduction, New York, NY, USA.
| | - Yan-Guang Wu
- Center for Human Reproduction, New York, NY, USA.
| | - Norbert Gleicher
- Center for Human Reproduction, New York, NY, USA.
- Foundation for Reproductive Medicine, New York, NY, USA.
| |
Collapse
|
32
|
Regal JF, Gilbert JS, Burwick RM. The complement system and adverse pregnancy outcomes. Mol Immunol 2015; 67:56-70. [PMID: 25802092 DOI: 10.1016/j.molimm.2015.02.030] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 02/20/2015] [Accepted: 02/24/2015] [Indexed: 02/08/2023]
Abstract
Adverse pregnancy outcomes significantly contribute to morbidity and mortality for mother and child, with lifelong health consequences for both. The innate and adaptive immune system must be regulated to insure survival of the fetal allograft, and the complement system is no exception. An intact complement system optimizes placental development and function and is essential to maintain host defense and fetal survival. Complement regulation is apparent at the placental interface from early pregnancy with some degree of complement activation occurring normally throughout gestation. However, a number of pregnancy complications including early pregnancy loss, fetal growth restriction, hypertensive disorders of pregnancy and preterm birth are associated with excessive or misdirected complement activation, and are more frequent in women with inherited or acquired complement system disorders or complement gene mutations. Clinical studies employing complement biomarkers in plasma and urine implicate dysregulated complement activation in components of each of the adverse pregnancy outcomes. In addition, mechanistic studies in rat and mouse models of adverse pregnancy outcomes address the complement pathways or activation products of importance and allow critical analysis of the pathophysiology. Targeted complement therapeutics are already in use to control adverse pregnancy outcomes in select situations. A clearer understanding of the role of the complement system in both normal pregnancy and complicated or failed pregnancy will allow a rational approach to future therapeutic strategies for manipulating complement with the goal of mitigating adverse pregnancy outcomes, preserving host defense, and improving long term outcomes for both mother and child.
Collapse
Affiliation(s)
- Jean F Regal
- Department of Biomedical Sciences, University of Minnesota Medical School, 1035 University Drive, Duluth, MN 55812, USA.
| | - Jeffrey S Gilbert
- Department of Biomedical Sciences, University of Minnesota Medical School, 1035 University Drive, Duluth, MN 55812, USA.
| | - Richard M Burwick
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, Oregon Health & Science University, Mail Code: L-458, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.
| |
Collapse
|
33
|
Severance EG, Gressitt KL, Buka SL, Cannon TD, Yolken RH. Maternal complement C1q and increased odds for psychosis in adult offspring. Schizophr Res 2014; 159:14-9. [PMID: 25195065 PMCID: PMC4177507 DOI: 10.1016/j.schres.2014.07.053] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 07/22/2014] [Accepted: 07/29/2014] [Indexed: 01/15/2023]
Abstract
The presence of maternal antibodies to food and infectious antigens may confer an increased risk of developing schizophrenia and psychosis in adult offspring. Complement factor C1q is an immune molecule with multiple functions including clearance of antigen-antibody complexes from circulation and mediation of synaptic pruning during fetal brain development. To determine if maternal C1q was associated with offspring schizophrenia and psychosis, we evaluated 55 matched case-control maternal serum pairs from the National Collaborative Perinatal Project. Sample pairs were composed of mothers whose offspring developed psychoses as adults and those whose offspring were free from psychiatric disease. Matching criteria for offspring included birth date, delivery hospital, race, and gender, with further matching based on mother's age. IgG markers of C1q, bovine milk casein, egg ovalbumin, and wheat gluten were measured with enzyme-linked immunosorbent assays. C1q levels were compared to food antigen IgG and to previously generated data for C-reactive protein, adenovirus, herpes simplex viruses, influenza viruses, measles virus, and Toxoplasma gondii. C1q was significantly elevated in case mothers with odds ratios of 2.66-6.31 (conditional logistic regressions, p ≤ 0.008-0.05). In case mothers only, C1q was significantly correlated with antibodies to both food and infectious antigens: gluten (R(2)=0.26, p ≤ 0.004), herpes simplex virus type 2 (R(2)=0.21, p ≤ 0.02), and adenovirus (R(2)=0.25, p ≤ 0.006). In conclusion, exposure to maternal C1q activity during pregnancy may be a risk factor for the development of schizophrenia and psychosis in offspring. Prenatal measurement of maternal C1q may be an important and convergent screening tool to identify potentially deleterious immune activation from multiple sources.
Collapse
Affiliation(s)
- Emily G. Severance
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Blalock 1105, Baltimore, MD 21287-4933 U.S.A,Correspondence: Emily G. Severance, , tel: +1 410-614-3918, fax: +1 410-955-3723
| | - Kristin L. Gressitt
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Blalock 1105, Baltimore, MD 21287-4933 U.S.A
| | - Stephen L. Buka
- Department of Epidemiology, School of Public Health, Brown University, Providence, Rhode Island, U.S.A
| | - Tyrone D. Cannon
- Department of Psychology, 2 Hillhouse Avenue, Yale University, New Haven, CT, U.S.A
| | - Robert H. Yolken
- Stanley Division of Developmental Neurovirology, Department of Pediatrics, Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Blalock 1105, Baltimore, MD 21287-4933 U.S.A
| |
Collapse
|
34
|
Schminkey DL, Groer M. Imitating a stress response: a new hypothesis about the innate immune system's role in pregnancy. Med Hypotheses 2014; 82:721-9. [PMID: 24698849 DOI: 10.1016/j.mehy.2014.03.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 03/08/2014] [Indexed: 11/24/2022]
Abstract
Recent research challenges long-held hypotheses about mechanisms through which pregnancy induces maternal immune suppression or tolerance of the embryo/fetus. It is now understood that normal pregnancy engages the immune system and that the immune milieu changes with advancing gestation. We suggest that pregnancy mimics the innate immune system's response to stress, causing a sterile inflammatory response that is necessary for successful reproduction. The relationship between external stressors and immunomodulation in pregnancy has been acknowledged, but the specific mechanisms are still being explicated. Implantation and the first trimester are times of immune activation and intensive inflammation in the uterine environment. A period of immune quiescence during the second trimester allows for the growth and development of the maturing fetus. Labor is also an inflammatory event. The length of gestation and timing of parturition can be influenced by environmental stressors. These stressors affect pregnancy through neuroendocrine interaction with the immune system, specifically through the hypothalamic-pituitary-adrenal (HPA) axis and the hypothalamic-pituitary-ovarian axis. Trophoblastic cells that constitute the maternal-fetal interface appear to harness the maternal immune system to promote and maximize the reproductive success of the mother and fetus. Pregnancy is a time of upregulated innate immune responses and decreased adaptive, cell-mediated responses. The inflammatory processes of pregnancy resemble an immune response to brief naturalistic stressors: there is a shift from T helper (Th) 1 to T helper (Th) 2 dominant adaptive immunity with a concomitant shift in cytokine production, decreased proliferation of T cells, and decreased cytotoxicity of natural killer (NK) cells. Inclusion of both murine and human studies, allows an exploration of insights into how trophoblasts influence the activity of the maternal innate immune system during gestation.
Collapse
|
35
|
The first trimester gravid serum regulates procalcitonin expression in human macrophages skewing their phenotype in vitro. Mediators Inflamm 2014; 2014:248963. [PMID: 24733960 PMCID: PMC3964843 DOI: 10.1155/2014/248963] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 01/29/2014] [Indexed: 12/11/2022] Open
Abstract
Procalcitonin (PCT) is one of the best diagnostic and prognostic markers in clinical practice, widely used to evaluate the evolution of bacterial infections. Although it is mainly produced by thyroid, during sepsis almost all the peripheral tissues are involved in PCT production. Parenchymal cells have been suggested as the main source of PCT expression; however the contribution of macrophages is not clear yet. In response to environmental cues, tissue macrophages acquire distinct functional phenotypes, ranging from proinflammatory (M1) to anti-inflammatory (M2) phenotype. Macrophages at the fetal-maternal interface show immunosuppressive M2-like activities required for the maintenance of immunological homeostasis during pregnancy. This study aims to clarify the ability to synthesise PCT of fully differentiated (M0), polarized (M1/M2) macrophages and those cultured either in the presence of first trimester gravid serum (GS) or pregnancy hormones. We found out that M1 macrophages upregulate PCT expression following LPS stimulation compared to M0 and M2. The GS downregulates PCT expression in macrophages, skewing them towards an M2-like phenotype. This effect seems only partially mediated by the hormonal milieu. Our findings strengthen the key role of macrophages in counteracting inflammatory stimuli during pregnancy, suggesting PCT as a possible new marker of M1-like macrophages.
Collapse
|
36
|
Li X, Xu WM, Yin TL, Zhao QH, Peng LY, Yang J. Temporal and spatial regulation of ezrin-radixin-moesin-binding phosphoprotein-50-kDa (EBP50) during embryo implantation in mouse uterus. Int J Mol Sci 2012. [PMID: 23208378 PMCID: PMC3546698 DOI: 10.3390/ijms131216418] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Embryo implantation is a crucial process for successful pregnancy. To date, the mechanism of embryo implantation remains unclear. Ezrin-radixin-moesin-binding protein-50-kDa (EBP50) is a scaffold protein, which has been shown to play an important role in cancer development. Embryo implantation and cancer follow a similar progression. Thus, in this article, we utilized immunohistochemical staining and western blot analyses to examine the spatiotemporal expression and regulation of EBP50 both in the mouse uterus during embryo implantation as well as in other related models. We found that EBP50 was detected in epithelial cells in all of the groups used in our study. During the peri-implantation period, EBP50 mainly localized in apical membranes. At the implantation site (IS) on day 5 (D5) of pregnancy, EBP50 was mainly expressed in the nuclei of stroma cells, whereas from day 6 to day 8 (D6–D8) of pregnancy, the expression of EBP50 was noted in the cytoplasm of decidual cells. The expression of EBP50 was not significantly different in the pseudopregnant uterus and decreased in the uteri subjected to activation of delayed implantation. Artificial decidualization also decreased EBP50 expression. Thus, the expression levels and location were affected by active blastocysts and decidualization during the window of implantation.
Collapse
Affiliation(s)
- Xing Li
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China.
| | | | | | | | | | | |
Collapse
|
37
|
AP-2α–dependent regulation of Bcl-2/Bax expression affects apoptosis in the trophoblast. J Mol Histol 2012; 43:681-9. [DOI: 10.1007/s10735-012-9439-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 07/31/2012] [Indexed: 12/18/2022]
|