1
|
Rizzuto G. B Cell Responses to the Placenta and Fetus. ANNUAL REVIEW OF PATHOLOGY 2025; 20:33-58. [PMID: 39264989 PMCID: PMC11912550 DOI: 10.1146/annurev-pathmechdis-111523-023459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/14/2024]
Abstract
Pregnancy has fascinated immunologists ever since Peter Medawar's observation that reproduction runs contrary to the founding tenets of immunology. During healthy pregnancy, maternal B cells interact with antigens of the foreign conceptus (placenta and fetus) yet do not elicit rejection. Instead, robust and redundant fetomaternal tolerance pathways generally prevent maternal B cells and antibodies from harming the placenta and fetus. Fetomaternal tolerance is not absolute, and unfortunately there exist several pregnancy complications that arise from breaks therein. Here, important historic and recent developments in the field of fetomaternal tolerance pertaining to maternal B cells and antibodies are reviewed. General rules from which to conceptualize humoral tolerance to the placenta and fetus are proposed. Significant but underexplored ideas are highlighted and topics for future research are suggested, findings from which are predicted to provide insight into the fundamental nature of tolerance and bolster efforts to combat immune-mediated pregnancy complications.
Collapse
Affiliation(s)
- Gabrielle Rizzuto
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine Graduate School of Medical Sciences, New York, NY, USA
- Department of Anatomic Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immuno-Oncology, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA;
| |
Collapse
|
2
|
Kulkarni HS, Tague LK, Calabrese DR, Liao F, Liu Z, Garnica L, Shankar N, Wu X, Kulkarni DH, Bernardt C, Byers D, Chen C, Huang HJ, Witt CA, Hachem RR, Kreisel D, Atkinson JP, Greenland JR, Gelman AE. Impaired complement regulation drives chronic lung allograft dysfunction after lung transplantation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.17.623951. [PMID: 39605452 PMCID: PMC11601477 DOI: 10.1101/2024.11.17.623951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
A greater understanding of chronic lung allograft dysfunction (CLAD) pathobiology, the primary cause of mortality after lung transplantation, is needed to improve outcomes. The complement system links innate to adaptive immune responses and is activated early post-lung transplantation to form the C3 convertase, a critical enzyme that cleaves the central complement component C3. We hypothesized that LTx recipients with a genetic predisposition to enhanced complement activation have worse CLAD-free survival mediated through increased adaptive alloimmunity. We interrogated a known functional C3 polymorphism (C3R102G) that increases complement activation through impaired C3 convertase inactivation in two independent LTx recipient cohorts. C3R102G, identified in at least one out of three LTx recipients, was associated with worse CLAD-free survival, particularly in the subset of recipients who developed donor-specific antibodies (DSA). In a mouse orthotopic lung transplantation model, impaired recipient complement regulation resulted in more severe obstructive airway lesions when compared to wildtype controls, despite only moderate differences in graft-infiltrating effector T cells. Impaired complement regulation promoted the intragraft accumulation of memory B cells and antibody-secreting cells, resulting in increased DSA levels. In summary, genetic predisposition to complement activation is associated with B cell activation and worse CLAD-free survival.
Collapse
Affiliation(s)
- Hrishikesh S. Kulkarni
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Medicine, David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, CA, USA
| | - Laneshia K. Tague
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Daniel R. Calabrese
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
- Medical Service, Veterans Affairs Health Care System, San Francisco, California, USA
| | - Fuyi Liao
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Zhiyi Liu
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Lorena Garnica
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Nishanth Shankar
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Xiaobo Wu
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Devesha H. Kulkarni
- Department of Medicine, David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, CA, USA
| | - Cory Bernardt
- Department of Pathology, Washington University School of Medicine, St. Louis, MO, USA
| | - Derek Byers
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Catherine Chen
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Howard J. Huang
- Department of Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Chad A. Witt
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Ramsey R. Hachem
- Department of Internal Medicine, University of Utah Spencer Fox Eccles School of Medicine, Salt Lake City, UT
| | - Daniel Kreisel
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - John P. Atkinson
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - John R. Greenland
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
- Medical Service, Veterans Affairs Health Care System, San Francisco, California, USA
| | - Andrew E. Gelman
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
3
|
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
|
4
|
Golubska M, Paukszto Ł, Kurzyńska A, Mierzejewski K, Gerwel Z, Bogacka I. PPAR beta/delta regulates the immune response mechanisms in the porcine endometrium during LPS-induced inflammation - An in vitro study. Theriogenology 2024; 226:130-140. [PMID: 38878465 DOI: 10.1016/j.theriogenology.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 06/02/2024] [Accepted: 06/10/2024] [Indexed: 07/24/2024]
Abstract
Inflammation in the reproductive tract has become a serious threat to animal fertility. Recently, the role of peroxisome proliferator-activated receptor gamma (PPARγ) in the context of reproduction and the inflammatory response has been highlighted, but the role of PPARβ/δ has not been fully elucidated. The aim of the present study was to investigate the in vitro effect of PPARβ/δ ligands (agonist: L-165,041 and antagonist: GSK 3787) on the transcriptome profile of porcine endometrium during LPS-induced inflammation in the mid-luteal and follicular phases of the oestrous cycle (days 10-12 and 18-20, respectively) using the RNA-Seq method. During the mid-luteal phase of the oestrous cycle, the current study identified 145 and 143 differentially expressed genes (DEGs) after treatment with an agonist or antagonist, respectively. During the follicular phase of the oestrous cycle, 55 and 207 DEGs were detected after treatment with an agonist or antagonist, respectively. The detected DEGs are engaged in the regulation of various processes, such as the complement and coagulation cascade, NF-κB signalling pathway, or the pathway of 15-eicosatetraenoic acid derivatives synthesis. The results of the current study indicate that PPARβ/δ ligands are involved in the control of the endometrial inflammatory response.
Collapse
Affiliation(s)
- Monika Golubska
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Łukasz Paukszto
- Department of Botany and Nature Protection, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Aleksandra Kurzyńska
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Karol Mierzejewski
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Zuzanna Gerwel
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Iwona Bogacka
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland.
| |
Collapse
|
5
|
Arachchillage DJ, Platton S, Hickey K, Chu J, Pickering M, Sommerville P, MacCallum P, Breen K. Guidelines on the investigation and management of antiphospholipid syndrome. Br J Haematol 2024; 205:855-880. [PMID: 39031476 DOI: 10.1111/bjh.19635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 06/26/2024] [Indexed: 07/22/2024]
Affiliation(s)
- Deepa J Arachchillage
- Department of Haematology, Imperial College Healthcare NHS Trust, London, UK
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Sean Platton
- The Royal London Hospital Haemophilia Centre, Barts Health NHS Trust, London, UK
| | - Kieron Hickey
- Sheffield Laboratory Medicine, Department of Coagulation, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Justin Chu
- Department of Obstetrics and Gynaecology, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - Matthew Pickering
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, UK
- Department of Rheumatology, Imperial College Healthcare NHS Trust, London, UK
| | - Peter Sommerville
- Department of Stroke Medicine, Guy's & St. Thomas' NHS Foundation Trust, London, UK
| | - Peter MacCallum
- Wolfson Institute of Population Health, Queen Mary University of London, London, UK
- Department of Clinical Haematology, Barts Health NHS Trust, London, UK
| | - Karen Breen
- Department of Haematology, Guy's & St. Thomas' NHS Foundation Trust, London, UK
| |
Collapse
|
6
|
Tsikouras P, Antsaklis P, Nikolettos K, Kotanidou S, Kritsotaki N, Bothou A, Andreou S, Nalmpanti T, Chalkia K, Spanakis V, Iatrakis G, Nikolettos N. Diagnosis, Prevention, and Management of Fetal Growth Restriction (FGR). J Pers Med 2024; 14:698. [PMID: 39063953 PMCID: PMC11278205 DOI: 10.3390/jpm14070698] [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: 06/06/2024] [Revised: 06/22/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
Fetal growth restriction (FGR), or intrauterine growth restriction (IUGR), is still the second most common cause of perinatal mortality. The factors that contribute to fetal growth restriction can be categorized into three distinct groups: placental, fetal, and maternal. The prenatal application of various diagnostic methods can, in many cases, detect the deterioration of the fetal condition in time because the nature of the above disorder is thoroughly investigated by applying a combination of biophysical and biochemical methods, which determine the state of the embryo-placenta unit and assess the possible increased risk of perinatal failure outcome and potential for many later health problems. When considering the potential for therapeutic intervention, the key question is whether it can be utilized during pregnancy. Currently, there are no known treatment interventions that effectively enhance placental function and promote fetal weight development. Nevertheless, in cases with fetuses diagnosed with fetal growth restriction, immediate termination of pregnancy may have advantages not only in terms of minimizing perinatal mortality but primarily in terms of reducing long-term morbidity during childhood and maturity.
Collapse
Affiliation(s)
- Panagiotis Tsikouras
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (K.N.); (S.K.); (N.K.); (S.A.); (T.N.); (K.C.); (V.S.); (N.N.)
| | - Panos Antsaklis
- Department of Obstetrics and Gynecology Medical School, University Hospital Alexandra, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Konstantinos Nikolettos
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (K.N.); (S.K.); (N.K.); (S.A.); (T.N.); (K.C.); (V.S.); (N.N.)
| | - Sonia Kotanidou
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (K.N.); (S.K.); (N.K.); (S.A.); (T.N.); (K.C.); (V.S.); (N.N.)
| | - Nektaria Kritsotaki
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (K.N.); (S.K.); (N.K.); (S.A.); (T.N.); (K.C.); (V.S.); (N.N.)
| | - Anastasia Bothou
- Department of Midwifery, School of Health Sciences, University of West Attica (UNIWA), 12243 Athens, Greece; (A.B.); (G.I.)
| | - Sotiris Andreou
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (K.N.); (S.K.); (N.K.); (S.A.); (T.N.); (K.C.); (V.S.); (N.N.)
| | - Theopi Nalmpanti
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (K.N.); (S.K.); (N.K.); (S.A.); (T.N.); (K.C.); (V.S.); (N.N.)
| | - Kyriaki Chalkia
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (K.N.); (S.K.); (N.K.); (S.A.); (T.N.); (K.C.); (V.S.); (N.N.)
| | - Vlasis Spanakis
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (K.N.); (S.K.); (N.K.); (S.A.); (T.N.); (K.C.); (V.S.); (N.N.)
| | - George Iatrakis
- Department of Midwifery, School of Health Sciences, University of West Attica (UNIWA), 12243 Athens, Greece; (A.B.); (G.I.)
- Department of Obstetrics and Gynecology, National and Kapodistrian University of Athens and Rea Maternity Hospital, 12462 Athens, Greece
| | - Nikolaos Nikolettos
- Department of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (K.N.); (S.K.); (N.K.); (S.A.); (T.N.); (K.C.); (V.S.); (N.N.)
| |
Collapse
|
7
|
Govender S, David M, Naicker T. Is the Complement System Dysregulated in Preeclampsia Comorbid with HIV Infection? Int J Mol Sci 2024; 25:6232. [PMID: 38892429 PMCID: PMC11172754 DOI: 10.3390/ijms25116232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 05/27/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
South Africa is the epicentre of the global HIV pandemic, with 13.9% of its population infected. Preeclampsia (PE), a hypertensive disorder of pregnancy, is often comorbid with HIV infection, leading to multi-organ dysfunction and convulsions. The exact pathophysiology of preeclampsia is triggered by an altered maternal immune response or defective development of maternal tolerance to the semi-allogenic foetus via the complement system. The complement system plays a vital role in the innate immune system, generating inflammation, mediating the clearance of microbes and injured tissue materials, and a mediator of adaptive immunity. Moreover, the complement system has a dual effect, of protecting the host against HIV infection and enhancing HIV infectivity. An upregulation of regulatory proteins has been implicated as an adaptive phenomenon in response to elevated complement-mediated cell lysis in HIV infection, further aggravated by preeclamptic complement activation. In light of the high prevalence of HIV infection and preeclampsia in South Africa, this review discusses the association of complement proteins and their role in the synergy of HIV infection and preeclampsia in South Africa. It aims to identify women at elevated risk, leading to early diagnosis and better management with targeted drug therapy, thereby improving the understanding of immunological dysregulation.
Collapse
Affiliation(s)
| | | | - Thajasvarie Naicker
- Optics and Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa; (S.G.); (M.D.)
| |
Collapse
|
8
|
Wei J, Zhang L, Xu H, Luo Q. Preterm birth, a consequence of immune deviation mediated hyperinflammation. Heliyon 2024; 10:e28483. [PMID: 38689990 PMCID: PMC11059518 DOI: 10.1016/j.heliyon.2024.e28483] [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: 12/21/2023] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 05/02/2024] Open
Abstract
Preterm birth represents a multifaceted syndrome with intricacies still present in our comprehension of its etiology. In the context of a semi-allograft, the prosperity from implantation to pregnancy to delivery hinges on the establishment of a favorable maternal-fetal immune microenvironment and a successful trilogy of immune activation, immune tolerance and then immune activation transitions. The occurrence of spontaneous preterm birth could be related to abnormalities within the immune trilogy, stemming from deviation in maternal and fetal immunity. These immune deviations, characterized by insufficient immune tolerance and early immune activation, ultimately culminated in an unsustainable pregnancy. In this review, we accentuated the role of both innate and adaptive immune reason in promoting spontaneous preterm birth, reviewed the risk of preterm birth from vaginal microbiome mediated by immune changes and the potential of vaginal microbiomes and metabolites as a new predictive marker, and discuss the changes in the role of progesterone and its interaction with immune cells in a preterm birth population. Our objective was to contribute to the growing body of knowledge in the field, shedding light on the immunologic reason of spontaneous preterm birth and effective biomarkers for early prediction, providing a roadmap for forthcoming investigations.
Collapse
Affiliation(s)
- Juan Wei
- Department of Obstetrics, Women's Hospital, of Zhejiang University School of Medicine, Hangzhou, 310006, China
- Key Laboratory of Reproductive Genetics, Ministry of Education, China
| | - LiYuan Zhang
- Department of Obstetrics, Women's Hospital, of Zhejiang University School of Medicine, Hangzhou, 310006, China
- Key Laboratory of Reproductive Genetics, Ministry of Education, China
| | - Heng Xu
- Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Qiong Luo
- Department of Obstetrics, Women's Hospital, of Zhejiang University School of Medicine, Hangzhou, 310006, China
- Key Laboratory of Reproductive Genetics, Ministry of Education, China
| |
Collapse
|
9
|
Boulanger H, Bounan S, Mahdhi A, Drouin D, Ahriz-Saksi S, Guimiot F, Rouas-Freiss N. Immunologic aspects of preeclampsia. AJOG GLOBAL REPORTS 2024; 4:100321. [PMID: 38586611 PMCID: PMC10994979 DOI: 10.1016/j.xagr.2024.100321] [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] [Indexed: 04/09/2024] Open
Abstract
Preeclampsia is a syndrome with multiple etiologies. The diagnosis can be made without proteinuria in the presence of dysfunction of at least 1 organ associated with hypertension. The common pathophysiological pathway includes endothelial cell activation, intravascular inflammation, and syncytiotrophoblast stress. There is evidence to support, among others, immunologic causes of preeclampsia. Unlike defense immunology, reproductive immunology is not based on immunologic recognition systems of self/non-self and missing-self but on immunotolerance and maternal-fetal cellular interactions. The main mechanisms of immune escape from fetal to maternal immunity at the maternal-fetal interface are a reduction in the expression of major histocompatibility complex molecules by trophoblast cells, the presence of complement regulators, increased production of indoleamine 2,3-dioxygenase, activation of regulatory T cells, and an increase in immune checkpoints. These immune protections are more similar to the immune responses observed in tumor biology than in allograft biology. The role of immune and nonimmune decidual cells is critical for the regulation of trophoblast invasion and vascular remodeling of the uterine spiral arteries. Regulatory T cells have been found to play an important role in suppressing the effectiveness of other T cells and contributing to local immunotolerance. Decidual natural killer cells have a cytokine profile that is favored by the presence of HLA-G and HLA-E and contributes to vascular remodeling. Studies on the evolution of mammals show that HLA-E, HLA-G, and HLA-C1/C2, which are expressed by trophoblasts and their cognate receptors on decidual natural killer cells, are necessary for the development of a hemochorial placenta with vascular remodeling. The activation or inhibition of decidual natural killer cells depends on the different possible combinations between killer cell immunoglobulin-like receptors, expressed by uterine natural killer cells, and the HLA-C1/C2 antigens, expressed by trophoblasts. Polarization of decidual macrophages in phenotype 2 and decidualization of stromal cells are also essential for high-quality vascular remodeling. Knowledge of the various immunologic mechanisms required for adequate vascular remodeling and their dysfunction in case of preeclampsia opens new avenues of research to identify novel biological markers or therapeutic targets to predict or prevent the onset of preeclampsia.
Collapse
Affiliation(s)
- Henri Boulanger
- Department of Nephrology and Dialysis, Clinique de l'Estrée, Stains, France (Drs Boulanger and Ahriz-Saksi)
| | - Stéphane Bounan
- Department of Obstetrics and Gynecology, Saint-Denis Hospital Center, Saint-Denis, France (Drs Bounan and Mahdhi)
| | - Amel Mahdhi
- Department of Obstetrics and Gynecology, Saint-Denis Hospital Center, Saint-Denis, France (Drs Bounan and Mahdhi)
| | - Dominique Drouin
- Department of Obstetrics and Gynecology, Clinique de l'Estrée, Stains, France (Dr Drouin)
| | - Salima Ahriz-Saksi
- Department of Nephrology and Dialysis, Clinique de l'Estrée, Stains, France (Drs Boulanger and Ahriz-Saksi)
| | - Fabien Guimiot
- Fetoplacental Unit, Robert-Debré Hospital, Assistance Publique – Hôpitaux de Paris, Paris, France (Dr Guimiot)
| | - Nathalie Rouas-Freiss
- Fundamental Research Division, CEA, Institut de biologie François Jacob, Hemato-Immunology Research Unit, Inserm UMR-S 976, Institut de Recherche Saint-Louis, Paris University, Saint-Louis Hospital, Paris, France (Dr Rouas-Freiss)
| |
Collapse
|
10
|
Huang CC, Hsueh YW, Chang CW, Hsu HC, Yang TC, Lin WC, Chang HM. Establishment of the fetal-maternal interface: developmental events in human implantation and placentation. Front Cell Dev Biol 2023; 11:1200330. [PMID: 37266451 PMCID: PMC10230101 DOI: 10.3389/fcell.2023.1200330] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/09/2023] [Indexed: 06/03/2023] Open
Abstract
Early pregnancy is a complex and well-orchestrated differentiation process that involves all the cellular elements of the fetal-maternal interface. Aberrant trophoblast-decidual interactions can lead to miscarriage and disorders that occur later in pregnancy, including preeclampsia, intrauterine fetal growth restriction, and preterm labor. A great deal of research on the regulation of implantation and placentation has been performed in a wide range of species. However, there is significant species variation regarding trophoblast differentiation as well as decidual-specific gene expression and regulation. Most of the relevant information has been obtained from studies using mouse models. A comprehensive understanding of the physiology and pathology of human implantation and placentation has only recently been obtained because of emerging advanced technologies. With the derivation of human trophoblast stem cells, 3D-organoid cultures, and single-cell analyses of differentiated cells, cell type-specific transcript profiles and functions were generated, and each exhibited a unique signature. Additionally, through integrative transcriptomic information, researchers can uncover the cellular dysfunction of embryonic and placental cells in peri-implantation embryos and the early pathological placenta. In fact, the clinical utility of fetal-maternal cellular trafficking has been applied for the noninvasive prenatal diagnosis of aneuploidies and the prediction of pregnancy complications. Furthermore, recent studies have proposed a viable path toward the development of therapeutic strategies targeting placenta-enriched molecules for placental dysfunction and diseases.
Collapse
|
11
|
Jena SR, Nayak J, Kumar S, Kar S, Samanta L. Comparative proteome profiling of seminal components reveal impaired immune cell signalling as paternal contributors in recurrent pregnancy loss patients. Am J Reprod Immunol 2023; 89:e13613. [PMID: 35998016 DOI: 10.1111/aji.13613] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 05/06/2022] [Accepted: 08/15/2022] [Indexed: 02/01/2023] Open
Abstract
PROBLEM Recurrent pregnancy loss (RPL) is usually evaluated from a women's perspective, however, recent evidence implies involvement of male factors as paternally expressed genes predominate placenta. During fertilization, prior to implantation the immune system purposefully produces early pregnancy factors with potent immunomodulatory properties for adaptation to antigenically dissimilar embryo. Therefore, it is hypothesized that paternal immunological factors play a role in RPL. METHOD OF STUDY Comparative proteome profiling (label free liquid chromatography mass spectroscopy: LC-MS/MS) of the seminal extracellular vesicles (SEVs), extracellular vesicle free seminal plasma (EVF-SP) and spermatozoa was carried out in semen of RPL patients (n = 21) and fertile donors (n = 21). This was followed by pathway and protein-protein interaction analysis, and validation of key proteins' expression (western blot). RESULTS A total of 68, 28 and 49 differentially expressed proteins in SEVs, EVF-SP and spermatozoa of RPL patients, respectively, were found to be involved in inflammatory response, immune cell signalling and apoptosis. In SEVs, underexpressed GDF-15 and overexpressed C3 imply distorted maternal immune response to paternal antigens leading to impaired decidualization. Dysregulated TGFβ signalling in EVF-SP surmises defective modulation of inflammatory response and induction of immune tolerance to seminal antigens in the female reproductive tract through generation of regulatory T cells. Retained histone variants in spermatozoa construe defective expression of early paternal genes, while underexpressed PTN may inflict defective angiogenesis resulting in expulsion of decidua. CONCLUSIONS Impaired modulation of immune response and improper placental development due to altered cytokine levels in seminal components may be the contributing paternal factors in RPL.
Collapse
Affiliation(s)
- Soumya Ranjan Jena
- Redox Biology & Proteomics Laboratory, Department of Zoology, School of Life Sciences, Ravenshaw University, College Square, Cuttack, Odisha, India.,Centre of Excellence in Environment and Public Health, Ravenshaw University, College Square, Cuttack, Odisha, India
| | - Jasmine Nayak
- Redox Biology & Proteomics Laboratory, Department of Zoology, School of Life Sciences, Ravenshaw University, College Square, Cuttack, Odisha, India.,Centre of Excellence in Environment and Public Health, Ravenshaw University, College Square, Cuttack, Odisha, India
| | - Sugandh Kumar
- School of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Sujata Kar
- Department of Obstetrics & Gynaecology, Kar Clinic and Hospital Pvt. Ltd., Bhubaneswar, India
| | - Luna Samanta
- Redox Biology & Proteomics Laboratory, Department of Zoology, School of Life Sciences, Ravenshaw University, College Square, Cuttack, Odisha, India.,Centre of Excellence in Environment and Public Health, Ravenshaw University, College Square, Cuttack, Odisha, India
| |
Collapse
|
12
|
KUNIYOSHI N, HANADA S, ANDO R, YUSTINASARI LR, KURATOMI M, KAGAWA S, IMAI H, KUSAKABE KT. Expression dynamics of Crry at the implantation sites in normal pregnancy and response against miscarriage induction. J Vet Med Sci 2023; 85:92-98. [PMID: 36450590 PMCID: PMC9887214 DOI: 10.1292/jvms.22-0286] [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: 06/21/2022] [Accepted: 11/07/2022] [Indexed: 12/02/2022] Open
Abstract
In mammals, immune tolerance against fetal tissue has been established for normal pregnancy progression. It is known that Crry regulates complemental activity to prevent injury of the mouse embryo and extra-embryonic tissue. This study aimed to examine the expression appearance and normal localization sites of Crry in the mouse placenta. Also, the emergency responses of Crry were verified at the time of experimental miscarriage induction. Moreover, we investigated an existing similar protein of Crry in animal placentas other than mice. Crry expression level showed a peak at day 8.5 of pregnancy. Trophoblast giant cells and decidual cells indicated a positive reaction to anti-Crry antibody. After treatments of interferon-γ, Crry expression was increased significantly in the survived implantation sites as compared with the controls. However, there was no significant difference in the miscarriage-initiated sites. It disclosed that Crry distributes from the early to middle periods of the placentas and involves complement regulation at the extraembryonic tissue and decidua basalis. Crry also showed an ability to respond to risk against external initiation for urgent miscarriage. Finally, we found anti-mouse Crry antibody-bound proteins in the placenta of many animals. It suggests a potency of Crry to make an environment of immune tolerance in many types of mammal placentas.
Collapse
Affiliation(s)
- Nobue KUNIYOSHI
- Laboratory of Basic Veterinary Science, The United Graduate School of Veterinary Science, Yamaguchi University, Yamaguchi, Japan
| | - Saki HANADA
- Laboratory of Veterinary Anatomy, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Reina ANDO
- Laboratory of Veterinary Anatomy, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Lita Rakhma YUSTINASARI
- Laboratory of Basic Veterinary Science, Joint Graduate School of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
- Division of Veterinary Anatomy, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Maria KURATOMI
- Laboratory of Veterinary Anatomy, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Seizaburo KAGAWA
- Laboratory of Veterinary Anatomy, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Hiroyuki IMAI
- Laboratory of Veterinary Anatomy, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
- Laboratory of Basic Veterinary Science, Joint Graduate School of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Ken Takeshi KUSAKABE
- Laboratory of Basic Veterinary Science, The United Graduate School of Veterinary Science, Yamaguchi University, Yamaguchi, Japan
- Laboratory of Veterinary Anatomy, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
- Laboratory of Basic Veterinary Science, Joint Graduate School of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| |
Collapse
|
13
|
Thurman JM, Harrison RA. The susceptibility of the kidney to alternative pathway activation-A hypothesis. Immunol Rev 2023; 313:327-338. [PMID: 36369971 DOI: 10.1111/imr.13168] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The glomerulus is often the prime target of dysregulated alternative pathway (AP) activation. In particular, AP activation is the key driver of two severe kidney diseases: atypical hemolytic uremic syndrome and C3 glomerulopathy. Both conditions are associated with a variety of predisposing molecular defects in AP regulation, such as genetic variants in complement regulators, autoantibodies targeting AP proteins, or autoantibodies that stabilize the AP convertases (C3- and C5-activating enzymes). It is noteworthy that these are systemic AP defects, yet in both diseases pathologic complement activation primarily affects the kidneys. In particular, AP activation is often limited to the glomerular capillaries. This tropism of AP-mediated inflammation for the glomerulus points to a unique interaction between AP proteins in plasma and this particular anatomic structure. In this review, we discuss the pre-clinical and clinical data linking the molecular causes of aberrant control of the AP with activation in the glomerulus, and the possible causes of this tropism. Based on these data, we propose a model for why the kidney is so uniquely and frequently targeted in patients with AP defects. Finally, we discuss possible strategies for preventing pathologic AP activation in the kidney.
Collapse
Affiliation(s)
- Joshua M Thurman
- Department of Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado, USA
| | | |
Collapse
|
14
|
Smith-Jackson K, Harrison RA. Alternative pathway activation in pregnancy, a measured amount "complements" a successful pregnancy, too much results in adverse events. Immunol Rev 2023; 313:298-319. [PMID: 36377667 PMCID: PMC10100418 DOI: 10.1111/imr.13169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
During pregnancy, the maternal host must adapt in order to enable growth of the fetus. These changes affect all organ systems and are designed both to protect the fetus and to minimize risk to the mother. One of the most prominent adaptations involves the immune system. The semi-allogenic fetoplacental unit has non-self components and must be protected against attack from the host. This requires both attenuation of adaptive immunity and protection from innate immune defense mechanisms. One of the key innate immune players is complement, and it is important that the fetoplacental unit is not identified as non-self and subjected to complement attack. Adaptation of the complement response must, however, be managed in such a way that maternal protection against infection is not compromised. As the complement system also plays a significant facilitating role in many of the stages of a normal pregnancy, it is also important that any necessary adaptation to accommodate the semi-allogenic aspects of the fetoplacental unit does not compromise this. In this review, both the physiological role of the alternative pathway of complement in facilitating a normal pregnancy, and its detrimental participation in pregnancy-specific disorders, are discussed.
Collapse
Affiliation(s)
- Kate Smith-Jackson
- Complement Therapeutics Research Group, Translational and Clinical Research Institute, Faculty of Medical Science, Newcastle University, Newcastle-upon-Tyne, UK.,The National Renal Complement Therapeutics Centre (NRCTC), Newcastle-upon-Tyne Hospitals NHS Foundation Trust, Newcastle-upon-Tyne, UK
| | | |
Collapse
|
15
|
Gibson BG, Cox TE, Marchbank KJ. Contribution of animal models to the mechanistic understanding of Alternative Pathway and Amplification Loop (AP/AL)-driven Complement-mediated Diseases. Immunol Rev 2023; 313:194-216. [PMID: 36203396 PMCID: PMC10092198 DOI: 10.1111/imr.13141] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
This review aimed to capture the key findings that animal models have provided around the role of the alternative pathway and amplification loop (AP/AL) in disease. Animal models, particularly mouse models, have been incredibly useful to define the role of complement and the alternative pathway in health and disease; for instance, the use of cobra venom factor and depletion of C3 provided the initial insight that complement was essential to generate an appropriate adaptive immune response. The development of knockout mice have further underlined the importance of the AP/AL in disease, with the FH knockout mouse paving the way for the first anti-complement drugs. The impact from the development of FB, properdin, and C3 knockout mice closely follows this in terms of mechanistic understanding in disease. Indeed, our current understanding that complement plays a role in most conditions at one level or another is rooted in many of these in vivo studies. That C3, in particular, has roles beyond the obvious in innate and adaptive immunity, normal physiology, and cellular functions, with or without other recognized AP components, we would argue, only extends the reach of this arm of the complement system. Humanized mouse models also continue to play their part. Here, we argue that the animal models developed over the last few decades have truly helped define the role of the AP/AL in disease.
Collapse
Affiliation(s)
- Beth G. Gibson
- Complement Therapeutics Research Group and Newcastle University Translational and Clinical Research InstituteFaculty of Medical ScienceNewcastle‐upon‐TyneUK
- National Renal Complement Therapeutics CentreaHUS ServiceNewcastle upon TyneUK
| | - Thomas E. Cox
- Complement Therapeutics Research Group and Newcastle University Translational and Clinical Research InstituteFaculty of Medical ScienceNewcastle‐upon‐TyneUK
- National Renal Complement Therapeutics CentreaHUS ServiceNewcastle upon TyneUK
| | - Kevin J. Marchbank
- Complement Therapeutics Research Group and Newcastle University Translational and Clinical Research InstituteFaculty of Medical ScienceNewcastle‐upon‐TyneUK
- National Renal Complement Therapeutics CentreaHUS ServiceNewcastle upon TyneUK
| |
Collapse
|
16
|
Kurokawa M, Takeshita A, Hashimoto S, Koyama M, Morimoto Y, Tachibana D. Prevention of intrauterine fetal growth restriction by administrating C1q/TNF-related protein 6, a specific inhibitor of the alternative complement pathway. J Assist Reprod Genet 2022; 39:2191-2199. [PMID: 35907048 PMCID: PMC9474761 DOI: 10.1007/s10815-022-02582-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 07/19/2022] [Indexed: 10/16/2022] Open
Abstract
PURPOSE The latest treatments do not sufficiently prevent miscarriage and fetal growth restriction (FGR) in pregnant women. Here, we assessed the effects of a human protein, CTRP6, that specifically inhibits the activation of the alternative complement pathway on miscarriage, fetal and placental development. METHODS Pregnant CBA/J mice mated with DBA/2 male mice as a model of spontaneous abortion and FGR were randomly divided into the control and CTRP6 groups. In the CTRP6 group, the mice were intravenously administered CTRP6 on days 4.5 and 6.5 post-conception (dpc). The abortion rate and fetal and placental weights on 14.5 dpc were examined. Remodeling of the spiral artery was also assessed. RESULTS The abortion rate in the CTRP6 group (13%) was reduced compared to the control group (21%), but there was no statistical difference. The placental and fetal weights in the CTRP6 group were also heavier than those in the control (P < 0.05). Moreover, the thickness of the blood vessel wall in the CTRP6 group was significantly thinner than that in the control (P < 0.05) and comparable to that in the non-abortion model (CBA/J x BALB). The ratio of the inner-per-the-outer diameter of the spiral artery increased more in the CTRP6 group than that in the control (P < 0.05). As well, the Th1/Th2 cytokine ratio was significantly reduced by CTRP6 treatment. CONCLUSIONS Taken together, the supplementation with a protein that regulates the alternative complement pathway in vivo improves FGR and promotes spiral artery remodeling in a mouse model of miscarriage and FGR.
Collapse
Affiliation(s)
- Mayu Kurokawa
- Women's Lifecare Medicine, Obstetrics and Gynecology, School of Medicine, Osaka City University, 545-8585, Osaka, Japan
- Graduate School of Medicine, Reproductive Science, Osaka City University, Osaka, 545-8585, Japan
| | - Ai Takeshita
- Graduate School of Medicine, Reproductive Science, Osaka City University, Osaka, 545-8585, Japan
- Department of Anatomy and Neurobiology, Faculty of Medicine, Kindai University, Osaka, 589-8511, Japan
| | - Shu Hashimoto
- Graduate School of Medicine, Reproductive Science, Osaka City University, Osaka, 545-8585, Japan.
| | - Masayasu Koyama
- Women's Lifecare Medicine, Obstetrics and Gynecology, School of Medicine, Osaka City University, 545-8585, Osaka, Japan
| | | | - Daisuke Tachibana
- Women's Lifecare Medicine, Obstetrics and Gynecology, School of Medicine, Osaka City University, 545-8585, Osaka, Japan
| |
Collapse
|
17
|
Castellanos Gutierrez AS, Figueras F, Morales-Prieto DM, Schleußner E, Espinosa G, Baños N. Placental damage in pregnancies with systemic lupus erythematosus: A narrative review. Front Immunol 2022; 13:941586. [PMID: 36059466 PMCID: PMC9428442 DOI: 10.3389/fimmu.2022.941586] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/12/2022] [Indexed: 11/17/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic inflammatory autoimmune disease of unknown cause, which mainly affects women of childbearing age, especially between 15 and 55 years of age. During pregnancy, SLE is associated with a high risk of perinatal morbidity and mortality. Among the most frequent complications are spontaneous abortion, fetal death, prematurity, intrauterine Fetal growth restriction (FGR), and preeclampsia (PE). The pathophysiology underlying obstetric mortality and morbidity in SLE is still under investigation, but several studies in recent years have suggested that placental dysfunction may play a crucial role. Understanding this association will contribute to developing therapeutic options and improving patient management thus reducing the occurrence of adverse pregnancy outcomes in this group of women. In this review, we will focus on the relationship between SLE and placental insufficiency leading to adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Aleida Susana Castellanos Gutierrez
- BCNatal, Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Déu), Institut Clínic de Ginecologia, Obstetrícia i Neonatologia Fetal i+D Fetal Medicine Research Center, Barcelona, Spain
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Jena, Germany
| | - Francesc Figueras
- BCNatal, Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Déu), Institut Clínic de Ginecologia, Obstetrícia i Neonatologia Fetal i+D Fetal Medicine Research Center, Barcelona, Spain
- Institut d’Investigacions Biomèdiques Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Diana M. Morales-Prieto
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Jena, Germany
- *Correspondence: Núria Baños, ; Diana M. Morales-Prieto,
| | - Ekkehard Schleußner
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Jena, Germany
| | - Gerard Espinosa
- Institut d’Investigacions Biomèdiques Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Autoimmune Diseases, Hospital Clinic, Barcelona, Spain
| | - Núria Baños
- BCNatal, Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Déu), Institut Clínic de Ginecologia, Obstetrícia i Neonatologia Fetal i+D Fetal Medicine Research Center, Barcelona, Spain
- *Correspondence: Núria Baños, ; Diana M. Morales-Prieto,
| |
Collapse
|
18
|
Rizzuto G, Erlebacher A. Trophoblast antigens, fetal blood cell antigens, and the paradox of fetomaternal tolerance. J Exp Med 2022; 219:e20211515. [PMID: 35416936 PMCID: PMC9011327 DOI: 10.1084/jem.20211515] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/15/2022] [Accepted: 03/18/2022] [Indexed: 12/16/2022] Open
Abstract
The paradox of fetomaternal tolerance has puzzled immunologists and reproductive biologists alike for almost 70 yr. Even the idea that the conceptus evokes a uniformly tolerogenic immune response in the mother is contradicted by the long-appreciated ability of pregnant women to mount robust antibody responses to paternal HLA molecules and RBC alloantigens such as Rh(D). Synthesizing these older observations with more recent work in mice, we discuss how the decision between tolerance or immunity to a given fetoplacental antigen appears to be a function of whether the antigen is trophoblast derived-and thus decorated with immunosuppressive glycans-or fetal blood cell derived.
Collapse
Affiliation(s)
- Gabrielle Rizzuto
- Department of Pathology, University of California San Francisco, San Francisco, CA
| | - Adrian Erlebacher
- Center for Reproductive Sciences, University of California San Francisco, San Francisco, CA
- Biomedical Sciences Program, University of California San Francisco, San Francisco, CA
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA
- Bakar ImmunoX Initiative, University of California San Francisco, San Francisco, CA
| |
Collapse
|
19
|
Zhu XC, Liu L, Dai WZ, Ma T. Crry silencing alleviates Alzheimer's disease injury by regulating neuroinflammatory cytokines and the complement system. Neural Regen Res 2022; 17:1841-1849. [PMID: 35017447 PMCID: PMC8820699 DOI: 10.4103/1673-5374.332160] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Complement component (3b/4b) receptor 1 (CR1) expression is positively related to the abundance of phosphorylated microtubule-associated protein tau (tau), and CR1 expression is associated with susceptibility to Alzheimer’s disease. However, the exact role of CR1 in tau protein-associated neurodegenerative diseases is unknown. In this study, we show that the mouse Cr1-related protein Y (Crry) gene, Crry, is localized to microglia. We also found that Crry protein expression in the hippocampus and cortex was significantly elevated in P301S mice (a mouse model widely used for investigating tau pathology) compared with that in wild-type mice. Tau protein phosphorylation (at serine 202, threonine 205, threonine 231, and serine 262) and expression of the major tau kinases glycogen synthase kinase-3 beta and cyclin-dependent-like kinase 5 were greater in P301S mice than in wild-type mice. Crry silencing by lentivirus-transfected short hairpin RNA led to greatly reduced tau phosphorylation and glycogen synthase kinase-3 beta and cyclin-dependent-like kinase 5 activity. Crry silencing reduced neuronal apoptosis and rescued cognitive impairment of P301S mice. Crry silencing also reduced the levels of the neuroinflammatory factors interleukin-1 beta, tumor necrosis factor alpha, and interleukin-6 and the complement components complement 3 and complement component 3b. Our results suggest that Crry silencing in the P301S mouse model reduces tau protein phosphorylation by reducing the levels of neuroinflammation and complement components, thereby improving cognitive function.
Collapse
Affiliation(s)
- Xi-Chen Zhu
- Department of Neurology, Wuxi No. 2 People's Hospital of Nanjing Medical University; Department of Neurology, Wuxi No. 2 People's Hospital, Affiliated Wuxi Clinical College of Nantong University, Wuxi, Jiangsu Province, China
| | - Lu Liu
- Department of Neurology, Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu Province, China
| | - Wen-Zhuo Dai
- Department of Neurology, Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu Province, China
| | - Tao Ma
- Department of Neurology, Wuxi No. 2 People's Hospital of Nanjing Medical University; Department of Neurology, Wuxi No. 2 People's Hospital, Affiliated Wuxi Clinical College of Nantong University, Wuxi, Jiangsu Province, China
| |
Collapse
|
20
|
Preterm Labor, a Syndrome Attributed to the Combination of External and Internal Factors. MATERNAL-FETAL MEDICINE 2022. [DOI: 10.1097/fm9.0000000000000136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
21
|
Mesenchymal stem cell therapy attenuates complement C3 deposition and improves the delicate equilibrium between angiogenic and anti-angiogenic factors in abortion-prone mice. Mol Immunol 2021; 141:246-256. [PMID: 34875452 DOI: 10.1016/j.molimm.2021.11.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 10/15/2021] [Accepted: 11/10/2021] [Indexed: 12/12/2022]
Abstract
Immunological disorders are one of the main causes of recurrent spontaneous abortions (RSA). A rapidly expanding body of evidence indicates that excessive activation of the complement system is critically involved in the development of miscarriages. In the CBA/J × DBA/2 murine model of recurrent miscarriage, exaggerated and unrestrained complement activation is reported to be the underlying cause of angiogenic factor imbalance and persistent inflammation. We have previously shown that mesenchymal stem cell (MSC) therapy can significantly reduce the abortion rate in abortion-prone mice through regulating the feto-maternal immune response. In the present study, we hypothesized that MSCs might improve the balance of angiogenic factors at the feto-maternal unit of CBA/J × DBA/2 mice by restraining complement activation and deposition. To explore this hypothesis, autologous adipose tissue-derived mesenchymal stem cells (AD-MSCs) were administered intra-peritoneally to abortion-prone mice on the 4.5th day of gestation. Control mice received PBS as vehicle. On day 13.5 of pregnancy, deposition of the complement component C3 and expression levels of Crry, CFD (adipsin), VEGF, PlGF and FLT-1 were measured at the feto-maternal interface by immunohistochemistry and real-time PCR, respectively. Decidual cells were also cultured in RPMI 1640 medium for 48 h and VEGF and sFLT-1 protein levels were quantified in supernatants using enzyme-linked immunosorbent assay (ELISA). Our results indicated that MSC therapy significantly reduced C3 deposition and adipsin transcription in the fetal-maternal interface of abortion-prone mice. Furthermore, administration of MSCs robustly upregulated the mRNA expression levels of Crry, VEGF, PlGF and FLT-1 in the placenta and decidua of CBA/J × DBA/2 mice. Consistently, the in vitro results demonstrated that decidual cells obtained from MSC-treated dams produced increased concentrations of VEGF in culture supernatants, with concomitant decreased levels of sFLT-1 protein. Here, we show for the first time that adoptive transfer of MSCs rectifies the disturbed balance of angiogenic factors observed at the feto-maternal unit of CBA/J × DBA/2 mice, in part at least, through inhibiting excessive complement activation and promoting the production of angiogenic factors. Collectively, these alterations seem to play a pivotal role in reducing the abortion rate and improving the intrauterine condition for the benefit of the fetus.
Collapse
|
22
|
Lu J, Wang X, Zhu Y, Ma L, Zheng S, Hu Z, Chen X. Clinical and Immunological Factors Associated with Postpartum Hepatic Flares in Immune-Tolerant Pregnant Women with Hepatitis B Virus Infection Treated with Telbivudine. Gut Liver 2021; 15:887-894. [PMID: 34446613 PMCID: PMC8593496 DOI: 10.5009/gnl21001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 01/15/2023] Open
Abstract
Background/Aims To investigate postpartum hepatic flares and associated factors in highly viremic pregnant patients in the immune tolerance phase who adopted telbivudine (LdT) treatment in the last trimester to reduce vertical transmission of hepatitis B virus. Methods Hepatitis B e antigen (HBeAg)-positive, highly viremic pregnant women were recruited for this prospective study. Treatment with LdT was started from 28 weeks of gestation. Virological and biochemical markers were examined before LdT treatment, antepartum and postpartum. Serial blood samples at the same time were collected to detect cytokines and cortisol (COR). Results Fifty-six of 153 patients (36.6%) had postpartum hepatic flares, defined as a 2-fold increase in alanine aminotransferase 6 weeks after delivery. Age and the antepartum alanine aminotransferase and postpartum HBeAg levels were independent influencing factors of postpartum hepatic flares. Cytokines showed no regularity during or after pregnancy. Compared with the patients with no postpartum flares, the patients with flares had lower baseline interferon γ and COR levels (p=0.022 and p=0.028) and higher postpartum interferon γ levels (p=0.026). Conclusions A high proportion of highly viremic and immune-tolerant pregnant patients treated with LdT in the last trimester had postpartum hepatic flares, which implied that these patients entered the immune clearance phase after delivery. Thus, this may create an appropriate opportunity for re-antiviral therapy.
Collapse
Affiliation(s)
- Junfeng Lu
- First Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Xiaoxiao Wang
- First Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yunxia Zhu
- Department of Obstetrics and Gynecology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Lina Ma
- First Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Sujun Zheng
- First Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Zhongjie Hu
- First Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Xinyue Chen
- First Department of Liver Disease, Beijing Youan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
23
|
Suah AN, Tran DKV, Khiew SH, Andrade MS, Pollard JM, Jain D, Young JS, Yin D, Chalasani G, Alegre ML, Chong AS. Pregnancy-induced humoral sensitization overrides T cell tolerance to fetus-matched allografts in mice. J Clin Invest 2021; 131:140715. [PMID: 33393512 PMCID: PMC7773355 DOI: 10.1172/jci140715] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 10/02/2020] [Indexed: 12/19/2022] Open
Abstract
Immunological tolerance to semiallogeneic fetuses is necessary to achieving successful first pregnancy and permitting subsequent pregnancies with the same father. Paradoxically, pregnancy is an important cause of sensitization, resulting in the accelerated rejection of offspring-matched allografts. The underlying basis for divergent outcomes following reencounter of the same alloantigens on transplanted organs versus fetuses in postpartum females is incompletely understood. Using a mouse model that allows concurrent tracking of endogenous fetus-specific T and B cell responses in a single recipient, we show that semiallogeneic pregnancies simultaneously induce fetus-specific T cell tolerance and humoral sensitization. Pregnancy-induced antibodies, but not B cells, impeded transplantation tolerance elicited by costimulation blockade to offspring-matched cardiac grafts. Remarkably, in B cell-deficient mice, allogeneic pregnancy enabled the spontaneous acceptance of fetus-matched allografts. The presence of pregnancy-sensitized B cells that cannot secrete antibodies at the time of heart transplantation was sufficient to precipitate rejection and override pregnancy-established T cell tolerance. Thus, while induction of memory B cells and alloantibodies by pregnancies establishes formidable barriers to transplant success for multigravid women, our observations raise the possibility that humoral desensitization will not only improve transplantation outcomes, but also reveal an unexpected propensity of multiparous recipients to achieve tolerance to offspring-matched allografts.
Collapse
Affiliation(s)
- Ashley N Suah
- Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | - Dong-Kha V Tran
- Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | - Stella Hw Khiew
- Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | - Michael S Andrade
- Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | - Jared M Pollard
- Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | - Dharmendra Jain
- Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | - James S Young
- Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | - Dengping Yin
- Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | - Geetha Chalasani
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Anita S Chong
- Department of Surgery, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
24
|
Yousefzadeh Y, Soltani-Zangbar MS, Hemmatzadeh M, Shomali N, Mahmoodpoor A, Ahmadian Heris J, Yousefi M. Fetomaternal Immune Tolerance: Crucial Mechanisms of Tolerance for Successful Pregnancy in Humans. Immunol Invest 2021; 51:1108-1125. [PMID: 33830854 DOI: 10.1080/08820139.2021.1909061] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
For many years, the question of how the maternal immune system tolerates the foreign fetus has remained unanswered, and numerous studies have considerably attempted to elucidate underlying mechanisms for fetomaternal tolerance. This review aimed at discussing various significant mechanisms in fetomaternal compatibility. At the fetomaternal interface, in addition to having efficient control against infections, innate and adaptive maternal immune systems selectively prevent fetal rejection. In general, understanding the complex mechanisms of fetomaternal tolerance is critical for immunologic tolerance induction and spontaneous abortion prevention in high-risk populations. Different cells and molecules, such as regulatory T-cells, dendritic cells, decidua cells, IDO, Class I HLA molecules, TGF-β, and IL-10, induce maternal immune tolerance in the fetus in numerous ways. The findings on fetomaternal immune tolerance have remained controversial and require further research.
Collapse
Affiliation(s)
- Yousef Yousefzadeh
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Committee Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Sadegh Soltani-Zangbar
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Hemmatzadeh
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Navid Shomali
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ata Mahmoodpoor
- Department of Anesthesiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Ahmadian Heris
- Department of Allergy and Clinical Immunology, Pediatric Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
25
|
Brown RJP, Tegtmeyer B, Sheldon J, Khera T, Anggakusuma, Todt D, Vieyres G, Weller R, Joecks S, Zhang Y, Sake S, Bankwitz D, Welsch K, Ginkel C, Engelmann M, Gerold G, Steinmann E, Yuan Q, Ott M, Vondran FWR, Krey T, Ströh LJ, Miskey C, Ivics Z, Herder V, Baumgärtner W, Lauber C, Seifert M, Tarr AW, McClure CP, Randall G, Baktash Y, Ploss A, Thi VLD, Michailidis E, Saeed M, Verhoye L, Meuleman P, Goedecke N, Wirth D, Rice CM, Pietschmann T. Liver-expressed Cd302 and Cr1l limit hepatitis C virus cross-species transmission to mice. SCIENCE ADVANCES 2020; 6:eabd3233. [PMID: 33148654 PMCID: PMC7673688 DOI: 10.1126/sciadv.abd3233] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 09/21/2020] [Indexed: 12/06/2023]
Abstract
Hepatitis C virus (HCV) has no animal reservoir, infecting only humans. To investigate species barrier determinants limiting infection of rodents, murine liver complementary DNA library screening was performed, identifying transmembrane proteins Cd302 and Cr1l as potent restrictors of HCV propagation. Combined ectopic expression in human hepatoma cells impeded HCV uptake and cooperatively mediated transcriptional dysregulation of a noncanonical program of immunity genes. Murine hepatocyte expression of both factors was constitutive and not interferon inducible, while differences in liver expression and the ability to restrict HCV were observed between the murine orthologs and their human counterparts. Genetic ablation of endogenous Cd302 expression in human HCV entry factor transgenic mice increased hepatocyte permissiveness for an adapted HCV strain and dysregulated expression of metabolic process and host defense genes. These findings highlight human-mouse differences in liver-intrinsic antiviral immunity and facilitate the development of next-generation murine models for preclinical testing of HCV vaccine candidates.
Collapse
Affiliation(s)
- Richard J P Brown
- Division of Veterinary Medicine, Paul Ehrlich Institute, 63225 Langen, Germany.
- Institute for Experimental Virology, Centre for Experimental and Clinical Infection Research, Twincore, Feodor-Lynen-Strasse 7, 30625 Hannover, Germany
| | - Birthe Tegtmeyer
- Institute for Experimental Virology, Centre for Experimental and Clinical Infection Research, Twincore, Feodor-Lynen-Strasse 7, 30625 Hannover, Germany
| | - Julie Sheldon
- Institute for Experimental Virology, Centre for Experimental and Clinical Infection Research, Twincore, Feodor-Lynen-Strasse 7, 30625 Hannover, Germany
| | - Tanvi Khera
- Institute for Experimental Virology, Centre for Experimental and Clinical Infection Research, Twincore, Feodor-Lynen-Strasse 7, 30625 Hannover, Germany
- Department of Gastroenterology and Hepatology, Faculty of Medicine, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Anggakusuma
- Institute for Experimental Virology, Centre for Experimental and Clinical Infection Research, Twincore, Feodor-Lynen-Strasse 7, 30625 Hannover, Germany
- Department of Research and Development, uniQure Biopharma, BV, Amsterdam, Netherlands
| | - Daniel Todt
- Institute for Experimental Virology, Centre for Experimental and Clinical Infection Research, Twincore, Feodor-Lynen-Strasse 7, 30625 Hannover, Germany
- Ruhr University Bochum, Faculty of Medicine, Department for Molecular and Medical Virology, Bochum, Germany
- European Virus Bioinformatics Center (EVBC), 07743 Jena, Germany
| | - Gabrielle Vieyres
- Institute for Experimental Virology, Centre for Experimental and Clinical Infection Research, Twincore, Feodor-Lynen-Strasse 7, 30625 Hannover, Germany
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Romy Weller
- Institute for Experimental Virology, Centre for Experimental and Clinical Infection Research, Twincore, Feodor-Lynen-Strasse 7, 30625 Hannover, Germany
| | - Sebastian Joecks
- Institute for Experimental Virology, Centre for Experimental and Clinical Infection Research, Twincore, Feodor-Lynen-Strasse 7, 30625 Hannover, Germany
| | - Yudi Zhang
- Institute for Experimental Virology, Centre for Experimental and Clinical Infection Research, Twincore, Feodor-Lynen-Strasse 7, 30625 Hannover, Germany
| | - Svenja Sake
- Institute for Experimental Virology, Centre for Experimental and Clinical Infection Research, Twincore, Feodor-Lynen-Strasse 7, 30625 Hannover, Germany
| | - Dorothea Bankwitz
- Institute for Experimental Virology, Centre for Experimental and Clinical Infection Research, Twincore, Feodor-Lynen-Strasse 7, 30625 Hannover, Germany
| | - Kathrin Welsch
- Institute for Experimental Virology, Centre for Experimental and Clinical Infection Research, Twincore, Feodor-Lynen-Strasse 7, 30625 Hannover, Germany
| | - Corinne Ginkel
- Institute for Experimental Virology, Centre for Experimental and Clinical Infection Research, Twincore, Feodor-Lynen-Strasse 7, 30625 Hannover, Germany
| | - Michael Engelmann
- Institute for Experimental Virology, Centre for Experimental and Clinical Infection Research, Twincore, Feodor-Lynen-Strasse 7, 30625 Hannover, Germany
- Ruhr University Bochum, Faculty of Medicine, Department for Molecular and Medical Virology, Bochum, Germany
| | - Gisa Gerold
- Department of Physiological Chemistry, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hannover, Germany
- Department of Clinical Microbiology, Virology and Wallenberg Center for Molecular Medicine (WCMM), Umeå University, 901 85 Umeå, Sweden
| | - Eike Steinmann
- Institute for Experimental Virology, Centre for Experimental and Clinical Infection Research, Twincore, Feodor-Lynen-Strasse 7, 30625 Hannover, Germany
- Ruhr University Bochum, Faculty of Medicine, Department for Molecular and Medical Virology, Bochum, Germany
| | - Qinggong Yuan
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany
- Twincore Centre for Experimental and Clinical Infection Research, Feodor-Lynen-Strasse 7, 30625 Hannover, Germany
| | - Michael Ott
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, 30625 Hannover, Germany
- Twincore Centre for Experimental and Clinical Infection Research, Feodor-Lynen-Strasse 7, 30625 Hannover, Germany
| | - Florian W R Vondran
- Department of General, Visceral, and Transplant Surgery, Hannover Medical School, 30625 Hannover, Germany
- German Centre for Infection Research (DZIF), Hannover-Braunschweig Site, Braunschweig, Germany
| | - Thomas Krey
- German Centre for Infection Research (DZIF), Hannover-Braunschweig Site, Braunschweig, Germany
- Institute of Virology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
- Center of Structural and Cell Biology in Medicine, Institute of Biochemistry, University of Luebeck, Luebeck, Germany
- Centre for Structural Systems Biology (CSSB), Hamburg, Germany
| | - Luisa J Ströh
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Csaba Miskey
- Division of Medical Biotechnology, Paul Ehrlich Institute, 63225 Langen, Germany
| | - Zoltán Ivics
- Division of Medical Biotechnology, Paul Ehrlich Institute, 63225 Langen, Germany
| | - Vanessa Herder
- Department of Pathology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Chris Lauber
- Institute for Experimental Virology, Centre for Experimental and Clinical Infection Research, Twincore, Feodor-Lynen-Strasse 7, 30625 Hannover, Germany
- Institute for Medical Informatics and Biometry, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Michael Seifert
- Institute for Medical Informatics and Biometry, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Alexander W Tarr
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
- School of Life Sciences and NIHR Nottingham BRC, University of Nottingham, Nottingham, UK
| | - C Patrick McClure
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
- School of Life Sciences and NIHR Nottingham BRC, University of Nottingham, Nottingham, UK
| | - Glenn Randall
- Department of Microbiology, The University of Chicago, Chicago, IL 60439, USA
| | - Yasmine Baktash
- Instituto de Biología Integrativa de Sistemas (I2SysBio), Parc Científic de Barcelona, Carrer del Catedràtic Agustín Escardino 9, 46980 Paterna, Valencia, Spain
| | - Alexander Ploss
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Viet Loan Dao Thi
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
- Schaller Research Group at Department of Infectious Diseases, Molecular Virology, Heidelberg University Hospital, Cluster of Excellence CellNetworks, Heidelberg, Germany
| | - Eleftherios Michailidis
- Schaller Research Group at Department of Infectious Diseases, Molecular Virology, Heidelberg University Hospital, Cluster of Excellence CellNetworks, Heidelberg, Germany
| | - Mohsan Saeed
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
- Department of Biochemistry, Boston University School of Medicine, National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA 02118, USA
| | - Lieven Verhoye
- Laboratory of Liver Infectious Diseases, Ghent University, Ghent, Belgium
| | - Philip Meuleman
- Laboratory of Liver Infectious Diseases, Ghent University, Ghent, Belgium
| | - Natascha Goedecke
- Helmholtz Centre for Infection Research, Division Model Systems for Infection and Immunity, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Dagmar Wirth
- Helmholtz Centre for Infection Research, Division Model Systems for Infection and Immunity, Inhoffenstraße 7, 38124 Braunschweig, Germany
- Department of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Charles M Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Thomas Pietschmann
- Institute for Experimental Virology, Centre for Experimental and Clinical Infection Research, Twincore, Feodor-Lynen-Strasse 7, 30625 Hannover, Germany.
- German Centre for Infection Research (DZIF), Hannover-Braunschweig Site, Braunschweig, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| |
Collapse
|
26
|
Ayers CD, Carlson KS. Spontaneous Pregnancy in the Setting of Primary Ovarian Insufficiency and Breastfeeding: Does Immunosuppression Play a Role? AMERICAN JOURNAL OF CASE REPORTS 2020; 21:e926980. [PMID: 33127872 PMCID: PMC7643410 DOI: 10.12659/ajcr.926980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Patient: Female, 34-year-old Final Diagnosis: Pregnancy • premature ovarian insufficiency Symptoms: Amenorrhea • pregnancy Medication: — Clinical Procedure: — Specialty: Obstetrics and Gynecology
Collapse
Affiliation(s)
- Caleb D Ayers
- College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Karen S Carlson
- Department of Obstetrics and Gynecology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
27
|
Banda NK, Tomlinson S, Scheinman RI, Ho N, Ramirez JR, Mehta G, Wang G, Vu VP, Simberg D, Kulik L, Holers VM. C2 IgM Natural Antibody Enhances Inflammation and Its Use in the Recombinant Single Chain Antibody-Fused Complement Inhibitor C2-Crry to Target Therapeutics to Joints Attenuates Arthritis in Mice. Front Immunol 2020; 11:575154. [PMID: 33178202 PMCID: PMC7596757 DOI: 10.3389/fimmu.2020.575154] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 09/08/2020] [Indexed: 01/12/2023] Open
Abstract
Natural IgM antibodies (NAbs) have been shown to recognize injury-associated neoepitopes and to initiate pathogenic complement activation. The NAb termed C2 binds to a subset of phospholipids displayed on injured cells, and its role(s) in arthritis, as well as the potential therapeutic benefit of a C2 NAb-derived ScFv-containing protein fused to a complement inhibitor, complement receptor-related y (Crry), on joint inflammation are unknown. Our first objective was to functionally test mAb C2 binding to apoptotic cells from the joint and also evaluate its inflammation enhancing capacity in collagen antibody-induced arthritis (CAIA). The second objective was to generate and test the complement inhibitory capacity of C2-Crry fusion protein in the collagen-induced arthritis (CIA) model. The third objective was to demonstrate in vivo targeting of C2-Crry to damaged joints in mice with arthritis. The effect of C2-NAb on CAIA in C57BL/6 mice was examined by inducing a suboptimal disease. The inhibitory effect of C2-Crry in DBA/1J mice with CIA was determined by injecting 2x per week with a single dose of 0.250 mg/mouse. Clinical disease activity (CDA) was examined, and knee joints were fixed for analysis of histopathology, C3 deposition, and macrophage infiltration. In mice with suboptimal CAIA, at day 10 there was a significant (p < 0.017) 74% increase in the CDA in mice treated with C2 NAb, compared to mice treated with F632 control NAb. In mice with CIA, at day 35 there was a significant 39% (p < 0.042) decrease in the CDA in mice treated with C2-Crry. Total scores for histopathology were also 50% decreased (p < 0.0005) in CIA mice treated with C2-Crry. C3 deposition was significantly decreased in the synovium (44%; p < 0.026) and on the surface of cartilage (42%; p < 0.008) in mice treated with C2-Crry compared with PBS treated CIA mice. Furthermore, C2-Crry specifically bound to apoptotic fibroblast-like synoviocytes in vitro, and also localized in the knee joints of arthritic mice as analyzed by in vivo imaging. In summary, NAb C2 enhanced arthritis-related injury, and targeted delivery of C2-Crry to inflamed joints demonstrated disease modifying activity in a mouse model of human inflammatory arthritis.
Collapse
Affiliation(s)
- Nirmal K Banda
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Robert I Scheinman
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Nhu Ho
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States.,Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Joseline Ramos Ramirez
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Gaurav Mehta
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Guankui Wang
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Vivian Pham Vu
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Dmitri Simberg
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Liudmila Kulik
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - V Michael Holers
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
28
|
Jackson HM, Foley KE, O'Rourke R, Stearns TM, Fathalla D, Morgan BP, Howell GR. A novel mouse model expressing human forms for complement receptors CR1 and CR2. BMC Genet 2020; 21:101. [PMID: 32907542 PMCID: PMC7487969 DOI: 10.1186/s12863-020-00893-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/21/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The complement cascade is increasingly implicated in development of a variety of diseases with strong immune contributions such as Alzheimer's disease and Systemic Lupus Erythematosus. Mouse models have been used to determine function of central components of the complement cascade such as C1q and C3. However, species differences in their gene structures mean that mice do not adequately replicate human complement regulators, including CR1 and CR2. Genetic variation in CR1 and CR2 have been implicated in modifying disease states but the mechanisms are not known. RESULTS To decipher the roles of human CR1 and CR2 in health and disease, we engineered C57BL/6J (B6) mice to replace endogenous murine Cr2 with human complement receptors, CR1 and CR2 (B6.CR2CR1). CR1 has an array of allotypes in human populations and using traditional recombination methods (Flp-frt and Cre-loxP) two of the most common alleles (referred to here as CR1long and CR1short) can be replicated within this mouse model, along with a CR1 knockout allele (CR1KO). Transcriptional profiling of spleens and brains identified genes and pathways differentially expressed between mice homozygous for either CR1long, CR1short or CR1KO. Gene set enrichment analysis predicts hematopoietic cell number and cell infiltration are modulated by CR1long, but not CR1short or CR1KO. CONCLUSION The B6.CR2CR1 mouse model provides a novel tool for determining the relationship between human-relevant CR1 alleles and disease.
Collapse
Affiliation(s)
- Harriet M Jackson
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
- Dementia Research Institute Cardiff and Systems Immunity Research Institute, School of Medicine, Cardiff University, Cardiff, Wales, UK
| | - Kate E Foley
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| | - Rita O'Rourke
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA
| | | | - Dina Fathalla
- Dementia Research Institute Cardiff and Systems Immunity Research Institute, School of Medicine, Cardiff University, Cardiff, Wales, UK
| | - B Paul Morgan
- Dementia Research Institute Cardiff and Systems Immunity Research Institute, School of Medicine, Cardiff University, Cardiff, Wales, UK
| | - Gareth R Howell
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, USA.
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA.
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, USA.
| |
Collapse
|
29
|
Girardi G, Lingo JJ, Fleming SD, Regal JF. Essential Role of Complement in Pregnancy: From Implantation to Parturition and Beyond. Front Immunol 2020; 11:1681. [PMID: 32849586 PMCID: PMC7411130 DOI: 10.3389/fimmu.2020.01681] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 06/23/2020] [Indexed: 12/12/2022] Open
Abstract
The complement cascade was identified over 100 years ago, yet investigation of its role in pregnancy remains an area of intense research. Complement inhibitors at the maternal-fetal interface prevent inappropriate complement activation to protect the fetus. However, this versatile proteolytic cascade also favorably influences numerous stages of pregnancy, including implantation, fetal development, and labor. Inappropriate complement activation in pregnancy can have adverse lifelong sequelae for both mother and child. This review summarizes the current understanding of complement activation during all stages of pregnancy. In addition, consequences of complement dysregulation during adverse pregnancy outcomes from miscarriage, preeclampsia, and pre-term birth are examined. Finally, future research directions into complement activation during pregnancy are considered.
Collapse
Affiliation(s)
- Guillermina Girardi
- Department of Basic Medical Sciences, College of Medicine, Member of QU Health, Qatar University, Doha, Qatar
| | - Joshua J Lingo
- Division of Biology, Kansas State University, Manhattan, KS, United States
| | - Sherry D Fleming
- Division of Biology, Kansas State University, Manhattan, KS, United States
| | - Jean F Regal
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, MN, United States
| |
Collapse
|
30
|
Guo Q, Li J, Zhong L, Cai H, Wang H, Yan H, Xia G, Kang Y, Wang C. Prostaglandin-E2 deficiency during late pregnancy and the associated increase in interleukin-1β derived from periaortic lymph nodes lead to abortion. Mol Hum Reprod 2020; 25:825-837. [PMID: 31633177 DOI: 10.1093/molehr/gaz058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 09/17/2019] [Indexed: 01/07/2023] Open
Abstract
Prostaglandin E2 (PGE2) is a hormone with many physiological functions. During pregnancy, it is generally believed that there is a high level of PGE2 at the final stage of pregnancy, which induces the contraction of uterine smooth muscle and promotes the occurrence of childbirth. However, we find that high PGE2 levels are present throughout late pregnancy in mice, not just during childbirth, and that PGE2 deficiency induced by indomethacin during late pregnancy causes damage to the placental labyrinth and eventually leads to abortion. Interestingly, the damage is closely related to inflammation, which involves the role of inflammatory factors produced by the periaortic lymph nodes (PLNs) near the uterus. Further, through RNA sequencing, we reveal that PLNs produce a large amount of interleukin-1β (IL-1β) when exposed to PGE2 deficiency, which causes damage to the placental labyrinth, probably via destroying the extracellular matrix. Finally, events leading to abortion following indomethacin administration are effectively prevented by supplementing PGE2 or by PLN removal. These results suggest that high levels of PGE2 during late pregnancy protect fetuses from inflammatory damage related to IL-1β. This work suggests a new role of PGE2 during late pregnancy and may provide potential therapeutic strategies for pathological pregnancy.
Collapse
Affiliation(s)
- Qirui Guo
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Junhong Li
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Liang Zhong
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Han Cai
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Huarong Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Hao Yan
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Guoliang Xia
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.,Key Lab of Ministry of Education for Protection and Utilization of Special Biological Resources in Western China, School of Life Science, Ningxia University, Yinchuan 750021, China
| | - Youmin Kang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Chao Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| |
Collapse
|
31
|
Abstract
Pregnancy is a natural process that poses an immunological challenge because non-self fetus must be accepted. During the pregnancy period, the fetus as 'allograft' inherits maternal and also paternal antigens. For successful and term pregnancy, the fetus is tolerated and nurtured enjoying immune privileges that minimize the risk of being rejected by maternal immune system. Multiple mechanisms contribute to tolerate the semi-allogeneic fetus. Here, we summarize the recent progresses on how the maternal immune system actively collaborates to maintain the immune balance and maternal-fetal tolerance.
Collapse
Affiliation(s)
- Xiaopeng Li
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.,Guangdong Engineering and Technology Research Center of Maternal-Fetal Medicine, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.,CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Jiayi Zhou
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.,Guangdong Engineering and Technology Research Center of Maternal-Fetal Medicine, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Min Fang
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.,CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,International College, University of the Chinese Academy of Sciences, Beijing, China
| | - Bolan Yu
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.,Guangdong Engineering and Technology Research Center of Maternal-Fetal Medicine, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
32
|
Timmermans SAMEG, Werion A, Spaanderman MEA, Reutelingsperger CP, Damoiseaux JGMC, Morelle J, van Paassen P. The natural course of pregnancies in women with primary atypical haemolytic uraemic syndrome and asymptomatic relatives. Br J Haematol 2020; 190:442-449. [PMID: 32342491 PMCID: PMC7496636 DOI: 10.1111/bjh.16626] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/06/2020] [Accepted: 03/09/2020] [Indexed: 11/28/2022]
Abstract
Pregnancy has been linked to various microangiopathies, including primary atypical haemolytic uraemic syndrome (aHUS). Complement dysregulation, often linked to rare variants in complement genes, is key for primary aHUS to manifest and may play a role in pregnancy complications of the mother and fetus. The burden of such complications is unknown, making counselling of women with primary aHUS and asymptomatic relatives difficult. We analyzed the maternal and fetal outcomes of 39 pregnancies from 17 women with primary aHUS and two asymptomatic relatives. Seven out of 39 pregnancies were complicated by pregnancy‐associated aHUS. Five out of 32 pregnancies not linked to pregnancy‐associated aHUS were complicated by pre‐eclampsia or HELLP. Rare genetic variants were identified in 10 women (asymptomatic relatives, n = 2) who had a total of 14 pregnancies, including 10 uncomplicated pregnancies. Thirty‐five out of 39 pregnancies resulted in live birth. Eight out of 19 women had progressed to end‐stage kidney disease, with an incidence of 2·95 (95% confidence interval, 1·37–5·61) per 100 person‐years after the first pregnancy. Thus, we emphasized the frequency of successful pregnancies in women with primary aHUS and asymptomatic relatives. Pregnancies should be monitored closely. Rare genetic variants cannot predict the risk of a given pregnancy.
Collapse
Affiliation(s)
- Sjoerd A M E G Timmermans
- Department of Nephrology and Clinical Immunology, Maastricht University Medical Center, Maastricht, the Netherlands.,Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Alexis Werion
- Division of Nephrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Marc E A Spaanderman
- Department of Obstetrics and Gynecology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Chris P Reutelingsperger
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Jan G M C Damoiseaux
- Department of Central Diagnostic Laboratory, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Johann Morelle
- Division of Nephrology, Cliniques Universitaires Saint-Luc, Brussels, Belgium.,Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium
| | - Pieter van Paassen
- Department of Nephrology and Clinical Immunology, Maastricht University Medical Center, Maastricht, the Netherlands.,Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
33
|
Hansen VL, Miller RD. Evidence for regulation of the complement system during pregnancy being ancient and conserved in mammals. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 104:103562. [PMID: 31785265 PMCID: PMC6937380 DOI: 10.1016/j.dci.2019.103562] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 11/26/2019] [Accepted: 11/26/2019] [Indexed: 06/10/2023]
Abstract
Here we demonstrate that regulation of the Complement (C') components of the immune system is an ancient and conserved feature of mammalian pregnancy. Transcript levels were reduced for complement components C3 and C4 throughout pregnancy in a marsupial, Monodelphis domestica. Downstream C' component transcripts were significantly less abundant relative to non-pregnant controls at the start of pregnancy but increased during late pregnancy, in some cases peaking close to parturition. These results are consistent with observations in human pregnancy that deposition of C5 through C9 on fetal membranes is associated with labor and parturition. Complement regulators CD46 and CD59 are present at the fetomaternal interface during M. domestica pregnancy as well, implying regulation of C' effector mechanisms is necessary for maintenance of normal marsupial pregnancy. Collectively these results support regulating the complement system may have contributed to the transition from oviparity to viviparity in mammals over 165 million years ago.
Collapse
Affiliation(s)
- Victoria L Hansen
- Center for Evolutionary and Theoretical Immunology, Department of Biology, University of New Mexico, Albuquerque, NM, USA
| | - Robert D Miller
- Center for Evolutionary and Theoretical Immunology, Department of Biology, University of New Mexico, Albuquerque, NM, USA.
| |
Collapse
|
34
|
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
|
35
|
Galindo-Sevilla N, Reyes-Arroyo F, Mancilla-Ramírez J. The role of complement in preterm birth and prematurity. J Perinat Med 2019; 47:793-803. [PMID: 31494635 DOI: 10.1515/jpm-2019-0175] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 08/14/2019] [Indexed: 12/17/2022]
Abstract
Complications of preterm birth (PTB) are the global leading cause of death in children younger than 5 years of age. Almost 15 million children are born prematurely in the world each year. Increasing evidence suggests that labor and delivery have many hallmarks of an inflammatory reaction, where complement activation has an active participation. As one of the most important components of inflammation, the role of complement during labor and PTB is becoming an attractive research target. The complement components C1q and C5b-9 are deposited on fetal membranes and release inflammatory mediators that contribute to uterine contractions, cervical ripening, cell chemotaxis, metalloproteinases production, membrane awaking and rupture, and it participates as a co-adjuvant in the onset and progress of labor. This article reviews a basic description of the complement system, its role in preterm birth and current concepts regarding its contribution in novel therapy strategies and new biomarkers.
Collapse
Affiliation(s)
- Norma Galindo-Sevilla
- Departamento de Infectología e Inmunología, Instituto Nacional de Perinatología, Secretaría de Salud, Mexico City 11000, Mexico
| | - Frida Reyes-Arroyo
- Departamento de Infectología e Inmunología, Instituto Nacional de Perinatología, Secretaría de Salud, Mexico City 11000, Mexico
- Servicio Social en Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Javier Mancilla-Ramírez
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico
- Hospital de la Mujer, Secretaría de Salud, Salvador Díaz Mirón esq, Plan de San Luis S/N, Casco de Santo Tomás, Miguel Hidalgo, Mexico City 11340, Mexico
| |
Collapse
|
36
|
Zhang Y, Jin S. Mitigating placental injuries through up-regulating DAF in experimental APS mice: new mechanism of progesterone. Clin Exp Immunol 2019; 197:376-386. [PMID: 31091357 PMCID: PMC6693963 DOI: 10.1111/cei.13313] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2019] [Indexed: 01/06/2023] Open
Abstract
Anti-phospholipid syndrome (APS) is characterized by recurrent pathological pregnancy, arterial or venous thrombosis in the presence of anti-phospholipid antibody (aPL). Complement activation is recognized as an intermediate link leading to placental thrombosis and placental inflammation in APS model mice. Decay accelerating factor (DAF, CD55), MAC-inhibitory protein (MAC-IP, CD59) and membrane co-factor protein (MCP, CD46) are important complement inhibitory proteins (CIPs) highly expressed in normal placenta to curb excessive complement activation and its mediated injuries. Anti-β2 glycoprotein I (anti-β2GPI) antibody is an important aPL. We found that placental DAF and CD46 decreased in β2GPI passively immunized APS model mice, accompanied by C3 deposition, neutrophil infiltration and increased proinflammatory cytokine levels detected in its placenta. Progesterone supplement can up-regulate DAF but not CD46 expression, curb C3 activation and decrease proinflammatory cytokines levels to reduce fetal loss frequency. Progesterone receptor antagonist (mifepristone) or knock-down DAF with specific siRNA, above the protective effects of progesterone, were significantly weakened. Another sex hormone, oestrogen, has no significant effect on placental DAF and C3 contents and fetal loss frequency in the APS mice model. This may be an important mechanism by which progesterone induces maternal-fetal immune tolerance. At the same time, it may provide evidence for the use of progesterone in APS abortion patients.
Collapse
Affiliation(s)
- Y. Zhang
- Department of Traditional Chinese MedicineMaternal and Child Health Hospital of Hubei ProvinceHubeiChina
| | - S. Jin
- Department of Traditional Chinese MedicineMaternal and Child Health Hospital of Hubei ProvinceHubeiChina
- First Clinical Medical CollegeHubei University of Chinese MedicineHubeiChina
| |
Collapse
|
37
|
Ojha H, Ghosh P, Singh Panwar H, Shende R, Gondane A, Mande SC, Sahu A. Spatially conserved motifs in complement control protein domains determine functionality in regulators of complement activation-family proteins. Commun Biol 2019; 2:290. [PMID: 31396570 PMCID: PMC6683126 DOI: 10.1038/s42003-019-0529-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 07/03/2019] [Indexed: 12/14/2022] Open
Abstract
Regulation of complement activation in the host cells is mediated primarily by the regulators of complement activation (RCA) family proteins that are formed by tandemly repeating complement control protein (CCP) domains. Functional annotation of these proteins, however, is challenging as contiguous CCP domains are found in proteins with varied functions. Here, by employing an in silico approach, we identify five motifs which are conserved spatially in a specific order in the regulatory CCP domains of known RCA proteins. We report that the presence of these motifs in a specific pattern is sufficient to annotate regulatory domains in RCA proteins. We show that incorporation of the lost motif in the fourth long-homologous repeat (LHR-D) in complement receptor 1 regains its regulatory activity. Additionally, the motif pattern also helped annotate human polydom as a complement regulator. Thus, we propose that the motifs identified here are the determinants of functionality in RCA proteins.
Collapse
Affiliation(s)
- Hina Ojha
- Complement Biology Laboratory, National Centre for Cell Science, S. P. Pune University campus, Pune, 411007 India
| | - Payel Ghosh
- Bioinformatics Centre, S. P. Pune University, Pune, 411007 India
| | - Hemendra Singh Panwar
- Complement Biology Laboratory, National Centre for Cell Science, S. P. Pune University campus, Pune, 411007 India
| | - Rajashri Shende
- Complement Biology Laboratory, National Centre for Cell Science, S. P. Pune University campus, Pune, 411007 India
| | | | - Shekhar C. Mande
- Structural Biology Laboratory, National Centre for Cell Science, S. P. Pune University campus, Pune, 411007 India
- Present Address: Council of Scientific and Industrial Research (CSIR), Anusandhan Bhawan, 2 Rafi Marg, New Delhi, 110001 India
| | - Arvind Sahu
- Complement Biology Laboratory, National Centre for Cell Science, S. P. Pune University campus, Pune, 411007 India
| |
Collapse
|
38
|
Ueda M, Sato Y, Horie A, Tani H, Miyazaki Y, Okunomiya A, Matsumoto H, Hamanishi J, Kondoh E, Mandai M. Endovascular trophoblast expresses CD59 to evade complement-dependent cytotoxicity. Mol Cell Endocrinol 2019; 490:57-67. [PMID: 30981734 DOI: 10.1016/j.mce.2019.04.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 03/10/2019] [Accepted: 04/07/2019] [Indexed: 11/27/2022]
Abstract
In the human placenta, extravillous trophoblasts (EVTs) invade maternal decidual tissues (interstitial trophoblasts) and maternal spiral arteries (endovascular trophoblasts). Although endovascular trophoblasts are directly exposed to maternal blood containing complement components, they are not eliminated by complement-dependent cytotoxicity (CDC). In this study, we investigated the expression and possible function of CD59, one of the membrane-bound complement regulators, in EVTs. Immunohistochemistry of early embryo implantation sites revealed that CD59 was hardly expressed on interstitial trophoblasts, whereas it was intensely expressed on endovascular trophoblasts. Using the human EVT-like cell line Swan71, we established CD59-silencing Swan71 cells (Sw_CD59sh) and non-silencing control Swan71 cells (Sw_CTRsh). In vitro cell apoptosis assay showed that Sw_CD59sh cells were significantly more susceptible to CDC as compared to Sw_CTRsh. Our results suggest that CD59 confers some protection against maternal complement attack to the endovascular trophoblasts.
Collapse
Affiliation(s)
- Masashi Ueda
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Yukiyasu Sato
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan; Department of Obstetrics and Gynecology, Takamatsu Red Cross Hospital, Takamatsu, 760-0017, Japan.
| | - Akihito Horie
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Hirohiko Tani
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Yumiko Miyazaki
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Fukui, Fukui, 910-1193, Japan
| | - Asuka Okunomiya
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Hisanori Matsumoto
- Department of Obstetrics and Gynecology, National Hospital Organization Osaka National Hospital, Osaka, 540-0006, Japan
| | - Junzo Hamanishi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Eiji Kondoh
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| | - Masaki Mandai
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, 606-8507, Japan
| |
Collapse
|
39
|
Chaouat G. A unified (but in fact not fully testable) model of preeclampsia triggering. J Reprod Immunol 2019; 132:49-53. [PMID: 30947086 DOI: 10.1016/j.jri.2019.03.002] [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: 03/04/2019] [Revised: 03/13/2019] [Accepted: 03/19/2019] [Indexed: 10/27/2022]
Abstract
In this summary of my presentation in the last Reunion workshop I discuss a few assertions on preeclampsia, then turn on a (not fully testable) model where an embryonic defect in expression of embryo/ placental regulatory proteins results in complement activation, itself responsible for a down regulation of the T regs activity, resulting in a very early lack of complete down regulation of the preimplantation decidual inflammation, causing in the post implantation stage a low grade but chronic inflammatory state.
Collapse
Affiliation(s)
- Gerard Chaouat
- U976 INSERM Hopital Saint Louis Pavillon Bazin, 75010 Paris France.
| |
Collapse
|
40
|
Banach P, Dereziński P, Matuszewska E, Matysiak J, Bochyński H, Kokot ZJ, Nowak-Markwitz E. MALDI-TOF-MS Analysis in the Identification of Urine Proteomic Patterns of Gestational Trophoblastic Disease. Metabolites 2019; 9:metabo9020030. [PMID: 30744112 PMCID: PMC6409522 DOI: 10.3390/metabo9020030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/02/2019] [Accepted: 02/03/2019] [Indexed: 12/20/2022] Open
Abstract
Gestational trophoblastic disease (GTD) is a group of highly aggressive, rare tumors. Human chorionic gonadotropin is a common biomarker used in the diagnosis and monitoring of GTD. To improve our knowledge of the pathology of GTD, we performed protein-peptide profiling on the urine of patients affected with gestational trophoblastic neoplasm (GTN). We analyzed urine samples from patients diagnosed with GTN (n = 26) and from healthy pregnant and non-pregnant controls (n = 17) using matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF-MS). Ions were examined in a linear mode over a m/z range of 1000–10,000. All GTN urine samples were analyzed before and after treatment and compared with those of the controls. The statistical analyses included multivariate classification algorithms as well as ROC curves. Urine sample analyses revealed there were significant differences in the composition of the ions between the evaluated groups. Comparing the pre-treatment and group with the pregnant controls, we identified two discriminatory proteins: hemoglobin subunit α (m/z = 1951.81) and complement C4A (m/z = 1895.43). Then, comparing urine samples from the post-treatment cases with those from the non-pregnant controls, we identified the peptides uromodulin fragments (m/z = 1682.34 and 1913.54) and complement C4A (m/z = 1895.43).
Collapse
Affiliation(s)
- Paulina Banach
- Gynecologic Oncology Department, Poznan University of Medical Sciences, Polna 33, 60-535 Poznan, Poland.
| | - Paweł Dereziński
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, Grunwaldzka 6, 60-780 Poznan, Poland.
| | - Eliza Matuszewska
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, Grunwaldzka 6, 60-780 Poznan, Poland.
| | - Jan Matysiak
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, Grunwaldzka 6, 60-780 Poznan, Poland.
| | - Hubert Bochyński
- Gynecologic Oncology Department, Poznan University of Medical Sciences, Polna 33, 60-535 Poznan, Poland.
| | - Zenon J Kokot
- Department of Inorganic and Analytical Chemistry, Poznan University of Medical Sciences, Grunwaldzka 6, 60-780 Poznan, Poland.
| | - Ewa Nowak-Markwitz
- Gynecologic Oncology Department, Poznan University of Medical Sciences, Polna 33, 60-535 Poznan, Poland.
| |
Collapse
|
41
|
Smith-Jackson K, Yang Y, Denton H, Pappworth IY, Cooke K, Barlow PN, Atkinson JP, Liszewski MK, Pickering MC, Kavanagh D, Cook HT, Marchbank KJ. Hyperfunctional complement C3 promotes C5-dependent atypical hemolytic uremic syndrome in mice. J Clin Invest 2019; 129:1061-1075. [PMID: 30714990 PMCID: PMC6391106 DOI: 10.1172/jci99296] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 12/18/2018] [Indexed: 12/13/2022] Open
Abstract
Atypical hemolytic uremic syndrome (aHUS) is frequently associated in humans with loss-of-function mutations in complement-regulating proteins or gain-of-function mutations in complement-activating proteins. Thus, aHUS provides an archetypal complement-mediated disease with which to model new therapeutic strategies and treatments. Herein, we show that, when transferred to mice, an aHUS-associated gain-of-function change (D1115N) to the complement-activation protein C3 results in aHUS. Homozygous C3 p.D1115N (C3KI) mice developed spontaneous chronic thrombotic microangiopathy together with hematuria, thrombocytopenia, elevated creatinine, and evidence of hemolysis. Mice with active disease had reduced plasma C3 with C3 fragment and C9 deposition within the kidney. Therapeutic blockade or genetic deletion of C5, a protein downstream of C3 in the complement cascade, protected homozygous C3KI mice from thrombotic microangiopathy and aHUS. Thus, our data provide in vivo modeling evidence that gain-of-function changes in complement C3 drive aHUS. They also show that long-term C5 deficiency is not accompanied by development of other renal complications (such as C3 glomerulopathy) despite sustained dysregulation of C3. Our results suggest that this preclinical model will allow testing of novel complement inhibitors with the aim of developing precisely targeted therapeutics that could have application in many complement-mediated diseases.
Collapse
Affiliation(s)
- Kate Smith-Jackson
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.,The National Renal Complement Therapeutics Centre (NRCTC), Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Yi Yang
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Harriet Denton
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Isabel Y Pappworth
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Katie Cooke
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Paul N Barlow
- Department of Chemistry, University of Edinburgh, Edinburgh, United Kingdom
| | - John P Atkinson
- Division of Rheumatology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - M Kathryn Liszewski
- Division of Rheumatology, Washington University in St. Louis, St. Louis, Missouri, USA
| | | | - David Kavanagh
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.,The National Renal Complement Therapeutics Centre (NRCTC), Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - H Terence Cook
- Department of Medicine, Imperial College London, London, United Kingdom
| | - Kevin J Marchbank
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.,The National Renal Complement Therapeutics Centre (NRCTC), Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
42
|
Zhang T, Chen X, Wang CC, Li TC, Kwak-Kim J. Intrauterine infusion of human chorionic gonadotropin before embryo transfer in IVF/ET cycle: The critical review. Am J Reprod Immunol 2019; 81:e13077. [PMID: 30589989 DOI: 10.1111/aji.13077] [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: 09/19/2018] [Revised: 12/07/2018] [Accepted: 12/10/2018] [Indexed: 12/16/2022] Open
Abstract
Intrauterine infusion of human chorionic gonadotropin (IUI-hCG) has been proposed to improve the outcome of in vitro fertilization-embryo transfer (IVF-ET), since it plays a critical role in synchronizing endometrial and fetal development. As the early mediator from embryo, hCG promotes the decidualization, angiogenesis, maternal immune tolerance, and trophoblast invasion, favoring successful implantation of embryo. Although multiple clinical trials have been conducted to verify the efficacy of IUI-hCG on IVF-ET outcome in recent years, the findings remained controversial. The difference in study design and population might be the cause to the different consequences after administration of hCG. More importantly, the endometrial receptivity, which might affect the efficacy of IUI-hCG, has not been assessed in women receiving this intervention. Selecting the right population suitable for IUI-hCG based on known etiology would be crucial in enhancing its efficacy and minimize any possible complications. Investigation of optimal indications for IUI-hCG should be highlighted in the future.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR.,Shenzhen Youshare Biotechnology Co. Ltd, Shenzhen, China
| | - Xiaoyan Chen
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR
| | - Chi-Chiu Wang
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR.,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong City, Hong Kong.,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong City, Hong Kong
| | - Tin Chiu Li
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR
| | - Joanne Kwak-Kim
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois.,Reproductive Medicine, Department of Obstetrics and Gynecology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, Vernon Hills, Illinois
| |
Collapse
|
43
|
Ander SE, Diamond MS, Coyne CB. Immune responses at the maternal-fetal interface. Sci Immunol 2019; 4:eaat6114. [PMID: 30635356 PMCID: PMC6744611 DOI: 10.1126/sciimmunol.aat6114] [Citation(s) in RCA: 378] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 11/29/2018] [Indexed: 12/13/2022]
Abstract
Pregnancy poses an immunological challenge because a genetically distinct (nonself) fetus must be supported within the pregnant female for the required gestational period. Placentation, or the establishment of the fetally derived placenta, is a common strategy used by eutherian mammals to protect the fetus and promote its growth. However, the substantial morphological differences of the placental architecture among species suggest that the process of placentation results from convergent evolution. Although there are considerable similarities in placental function across placental mammals, there are important differences that arise owing to species-specific immunological (and other biological) constraints. This Review focuses on the immunological similarities and differences that occur at the maternal-fetal interface in the context of human and mouse pregnancies. We discuss how the decidua and placenta of these different species form key immunological barriers that sustain maternal tolerance yet generate innate immune responses that prevent microbial infections.
Collapse
Affiliation(s)
- Stephanie E Ander
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
- Center for Microbial Pathogenesis, University of Pittsburgh Medical Center (UPMC) Children's Hospital of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carolyn B Coyne
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.
- Center for Microbial Pathogenesis, University of Pittsburgh Medical Center (UPMC) Children's Hospital of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
- R. K. Mellon Pediatric Research Institute, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15219, USA
| |
Collapse
|
44
|
Cismaru CA, Pop L, Berindan-Neagoe I. Incognito: Are Microchimeric Fetal Stem Cells that Cross Placental Barrier Real Emissaries of Peace? Stem Cell Rev Rep 2018; 14:632-641. [PMID: 29948753 DOI: 10.1007/s12015-018-9834-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Chimerism occurs naturaly throughout gestation and can also occur as a consequence of transfusion and transplantation therapy. It consists of the acquisition and long-term persistence of a genetically distinct population of allogenic cells inside another organism. Previous reports have suggested that feto-maternal microchimerism could exert a beneficial effect on the treatment of hematological and solid tumors in patients treated by PBSCT. In this review we report the mechanism of transplacental fetal stem cell trafficking during pregnancy and the effect of their long-term persistence on autoimmunity, GVHD, PBSCT, cancer and stem cell treatment.
Collapse
Affiliation(s)
- Cosmin Andrei Cismaru
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, 23 Gh. Marinescu street, 400337, Cluj-Napoca, Romania.
| | - Laura Pop
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, 23 Gh. Marinescu street, 400337, Cluj-Napoca, Romania
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, 23 Gh. Marinescu street, 400337, Cluj-Napoca, Romania
| |
Collapse
|
45
|
Abeln M, Albers I, Peters-Bernard U, Flächsig-Schulz K, Kats E, Kispert A, Tomlinson S, Gerardy-Schahn R, Münster-Kühnel A, Weinhold B. Sialic acid is a critical fetal defense against maternal complement attack. J Clin Invest 2018; 129:422-436. [PMID: 30382946 DOI: 10.1172/jci99945] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 10/30/2018] [Indexed: 02/06/2023] Open
Abstract
The negatively charged sugar sialic acid (Sia) occupies the outermost position in the bulk of cell surface glycans. Lack of sialylated glycans due to genetic ablation of the Sia-activating enzyme CMP-sialic acid synthase (CMAS) resulted in embryonic lethality around day 9.5 post coitum (E9.5) in mice. Developmental failure was caused by complement activation on trophoblasts in Cmas-/- implants and was accompanied by infiltration of maternal neutrophils at the fetal-maternal interface, intrauterine growth restriction, impaired placental development, and a thickened Reichert's membrane. This phenotype, which shared features with complement receptor 1-related protein Y (Crry) depletion, was rescued in E8.5 Cmas-/- mice upon injection of cobra venom factor, resulting in exhaustion of the maternal complement component C3. Here we show that Sia is dispensable for early development of the embryo proper but pivotal for fetal-maternal immune homeostasis during pregnancy, i.e., for protecting the allograft implant against attack by the maternal innate immune system. Finally, embryos devoid of cell surface sialylation suffered from malnutrition due to inadequate placentation as a secondary effect.
Collapse
Affiliation(s)
| | | | | | | | | | - Andreas Kispert
- Institut for Molecular Biology, Hannover Medical School, Hannover, Germany
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Children's Research Institute, Medical University of South Carolina, Charleston, South Carolina, USA
| | | | | | | |
Collapse
|
46
|
Lokki AI, Heikkinen-Eloranta JK, Laivuori H. The Immunogenetic Conundrum of Preeclampsia. Front Immunol 2018; 9:2630. [PMID: 30483272 PMCID: PMC6243043 DOI: 10.3389/fimmu.2018.02630] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 10/25/2018] [Indexed: 12/12/2022] Open
Abstract
Pregnancy is an immunological challenge to the mother. The fetal tissues including the placenta must be protected from activation of the maternal immune system. On the other hand, the placental tissue sheds into the maternal circulation and must be adequately identified and phagocytized by the maternal immune system. During a healthy pregnancy, numerous immunosuppressive processes take place that allow the allograft fetus to thrive under exposure to humoral and cellular components of the maternal immune system. Breakdown of immune tolerance may result in sterile inflammation and cause adverse pregnancy outcomes such as preeclampsia, a vascular disease of the pregnancy with unpredictable course and symptoms from several organs. Immunological incompatibility between mother and fetus is strongly indicated in preeclampsia. Recently, genetic factors linking immunological pathways to predisposition to preeclampsia have been identified. In this mini-review genetic variation in immunological factors are discussed in the context of preeclampsia. Specifically, we explore immunogenetic and immunomodulary mechanisms contributing to loss of tolerance, inflammation, and autoimmunity in preeclampsia.
Collapse
Affiliation(s)
- A Inkeri Lokki
- Research Programs Unit, Immunobiology Research Program, University of Helsinki, Helsinki, Finland.,Bacteriology and Immunology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | | | - Hannele Laivuori
- Medical and Clinical genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland.,Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland.,Department of Obstetrics and Gynecology, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
47
|
Deshmukh H, Way SS. Immunological Basis for Recurrent Fetal Loss and Pregnancy Complications. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2018; 14:185-210. [PMID: 30183507 DOI: 10.1146/annurev-pathmechdis-012418-012743] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Pregnancy stimulates an elaborate assortment of dynamic changes, allowing intimate approximation of genetically discordant maternal and fetal tissues. Although the cellular and molecular details about how this works remain largely undefined, important clues arise from evaluating how a prior pregnancy influences the outcome of a future pregnancy. The risk of complications is consistently increased when complications occurred in a prior pregnancy. Reciprocally, a prior successful pregnancy protects against complications in a future pregnancy. Here, we summarize immunological perturbations associated with fetal loss, with particular focus on how both harmful and protective adaptations may persist in mothers. Immunological aberrancy as a root cause of pregnancy complications is also considered, given their shared overlapping risk factors and the sustained requirement for averting maternal-fetal conflict throughout pregnancy. Understanding pregnancy-induced immunological changes may expose not only new therapeutic strategies for improving pregnancy outcomes but also new facets of how immune tolerance works that may be applicable to other physiological and pathological contexts.
Collapse
Affiliation(s)
- Hitesh Deshmukh
- Division of Pulmonary Biology, Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267, USA
| | - Sing Sing Way
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267, USA.,Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229, USA;
| |
Collapse
|
48
|
Mohlin FC, Gros P, Mercier E, Gris JCR, Blom AM. Analysis of C3 Gene Variants in Patients With Idiopathic Recurrent Spontaneous Pregnancy Loss. Front Immunol 2018; 9:1813. [PMID: 30131807 PMCID: PMC6090058 DOI: 10.3389/fimmu.2018.01813] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 07/23/2018] [Indexed: 12/18/2022] Open
Abstract
Miscarriage is the most common complication of pregnancy. Approximately 1% of couples trying to conceive will experience recurrent miscarriages, defined as three or more consecutive pregnancy losses and many of these cases remain idiopathic. Complement is implicated both in the physiology and pathology of pregnancy. Therefore, we hypothesized that alterations in the C3 gene could potentially predispose to this disorder. We performed full Sanger sequencing of all exons of C3, in 192 childless women, with at least two miscarriages and without any known risk factors. All exons carrying non-synonymous alterations found in the patients were then sequenced in a control group of 192 women. None of the identified alterations were significantly associated with the disorder. Thirteen identified non-synonymous alterations (R102G, K155Q, L302P, P314L, Y325H, V326A, S327P, V330I, K633R, R735W, R1591G, G1606D, and S1619R) were expressed recombinantly, upon which C3 expression and secretion were determined. The L302P and S327P were not secreted from the cells, likely due to misfolding and intracellular degradation. Y325H, V326A, V3301I, R1591G, and G1606D yielded approximately half C3 concentration in the cell media compared with wild type (WT). We analyzed the hemolytic activity of the secreted C3 variants by reconstituting C3-depleted serum. In this assay, R1591G had impaired hemolytic activity while majority of remaining mutants instead had increased activity. R1591G also yielded more factor B activation in solution compared with WT. R1591G and G1606D showed impaired degradation by factor I, irrespectively if factor H, CD46, or C4b-binding protein were used as cofactors. These two C3 mutants showed impaired binding of the cofactors and/or factor I. Taken together, several alterations in C3 were identified and some of these affected the secretion and/or the function of the protein, which might contribute to the disorder but the degree of association must be evaluated in larger cohorts.
Collapse
Affiliation(s)
- Frida C. Mohlin
- Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Piet Gros
- Crystal and Structural Chemistry, Bijvoet Center for Biomolecular Research, Department of Chemistry, Utrecht University, Utrecht, Netherlands
| | - Eric Mercier
- Laboratory of Hematology, University Hospital, Nimes, France
| | | | - Anna M. Blom
- Department of Translational Medicine, Lund University, Malmö, Sweden
| |
Collapse
|
49
|
Zhao X, Jiang Y, Wang L, Li Z, Li Q, Feng X. Advances in Understanding the Immune Imbalance between T-Lymphocyte Subsets and NK Cells in Recurrent Spontaneous Abortion. Geburtshilfe Frauenheilkd 2018; 78:677-683. [PMID: 30258242 PMCID: PMC6150770 DOI: 10.1055/a-0634-1813] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 04/16/2018] [Accepted: 05/24/2018] [Indexed: 01/07/2023] Open
Abstract
Recurrent spontaneous abortion is a global problem, and unexplained recurrent abortion triggered by immunological factors is an important focus of current research. Helper T lymphocytes (Th cells) and regulatory T lymphocytes (Treg cells) are central in human immune regulation and play a complex role in pregnancy. Natural killer cells (NK cells) exist in the endometrium and cooperate with T lymphocytes to create immune tolerance at the maternal-fetal interface, which is essential for successful pregnancy. This review has analyzed studies on Th17 cell, Treg cell and NK cell dysfunction and cellular imbalances which may contribute to unexplained recurrent spontaneous abortion to suggest a possible direction for future immunotherapies.
Collapse
Affiliation(s)
- Xiaoxuan Zhao
- Heilongjiang University of Chinese Medicine, Harbin 150040, Heilongjiang, China
| | - Yuepeng Jiang
- Heilongjiang University of Chinese Medicine, Harbin 150040, Heilongjiang, China
| | - Lin Wang
- Heilongjiang University of Chinese Medicine, Harbin 150040, Heilongjiang, China
| | - Zhihao Li
- Heilongjiang University of Chinese Medicine, Harbin 150040, Heilongjiang, China
| | - Qiang Li
- Heilongjiang University of Chinese Medicine, Harbin 150040, Heilongjiang, China
| | - Xiaoling Feng
- First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin 150040, Heilongjiang, China
| |
Collapse
|
50
|
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
|