1
|
Huang X, Yin T, Yu M, Zhu G, Hu X, Yu H, Zhao W, Chen J, Du J, Wu Q, Zhang W, Liu L, Du M. Decidualization-associated recruitment of cytotoxic memory CD8 +T cells to the maternal-fetal interface for immune defense. Mucosal Immunol 2025; 18:366-379. [PMID: 39675728 DOI: 10.1016/j.mucimm.2024.12.007] [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: 07/22/2024] [Revised: 12/07/2024] [Accepted: 12/10/2024] [Indexed: 12/17/2024]
Abstract
Decidual CD8+T (dCD8+T) cells are pivotal in the maintenance of the delicate balance between immune tolerance towards the fetus and immune resistance against pathogens. The endometrium and decidua represent the uterine environments before and during pregnancy, respectively, yet the composition and phenotypic alterations of uterine CD8+T cells in these tissues remain unclear. Using flow cytometry and analysis of transcriptome profiles, we demonstrated that human dCD8+T and endometrial CD8+T (eCD8+T) cells exhibited similar T cell differentiation statuses and phenotypes of tissue infiltrating or residency, compared to peripheral CD8+T (pCD8+T) cells. However, dCD8+T cells showed decreased expression of coinhibitory marker (PD-1), chemotaxis marker (CXCR3), and tissue-resident markers (CD69 and CD103), along with increased expression of granzyme B and granulysin, compared to eCD8+T cells. In vitro cytotoxicity assays further demonstrated that dCD8+T cells had greater effector functions than eCD8+T cells. Additionally, both in vitro and in vivo chemotaxis assays confirmed the recruitment of non-resident effector memory T cell subsets to the pregnant decidua, contributing to the dCD8+T cell-mediated anti-infection mechanism at the maternal-fetal interface. This work demonstrates dCD8+T cells replenished from the circulation retain their cytotoxic capacity, which may serve as an enhanced defense mechanism against infection during pregnancy.
Collapse
Affiliation(s)
- Xixi Huang
- Laboratory for Reproductive Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Tingxuan Yin
- Laboratory for Reproductive Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Min Yu
- Laboratory for Reproductive Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Guohua Zhu
- Laboratory for Reproductive Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Xianyang Hu
- Laboratory for Reproductive Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Hailin Yu
- Laboratory for Reproductive Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Weijie Zhao
- Longgang District Maternity & Child Healthcare Hospital of Shenzhen City, Longgang Maternity Child Institute of Shantou University Medical College, Shenzhen 518172, China
| | - Jiajia Chen
- Laboratory for Reproductive Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Jiangyuan Du
- Laboratory for Reproductive Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Qingyu Wu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China
| | - Wei Zhang
- Laboratory for Reproductive Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Lu Liu
- Laboratory for Reproductive Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200032, China.
| | - Meirong Du
- Laboratory for Reproductive Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200032, China; Department of Obstetrics and Gynecology, Shanghai Fourth People's Hospital, School of Medicine, Tongji University Shanghai, 200434, China.
| |
Collapse
|
2
|
Joo JS, Lee D, Hong JY. Multi-Layered Mechanisms of Immunological Tolerance at the Maternal-Fetal Interface. Immune Netw 2024; 24:e30. [PMID: 39246621 PMCID: PMC11377946 DOI: 10.4110/in.2024.24.e30] [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: 04/12/2024] [Revised: 07/04/2024] [Accepted: 07/08/2024] [Indexed: 09/10/2024] Open
Abstract
Pregnancy represents an immunological paradox where the maternal immune system must tolerate the semi-allogeneic fetus expressing paternally-derived Ags. Accumulating evidence over decades has revealed that successful pregnancy requires the active development of robust immune tolerance mechanisms. This review outlines the multi-layered processes that establish fetomaternal tolerance, including the physical barrier of the placenta, restricted chemokine-mediated leukocyte trafficking, lack of sufficient alloantigen presentation, the presence of immunosuppressive regulatory T cells and tolerogenic decidual natural killer cells, expression of immune checkpoint molecules, specific glycosylation patterns conferring immune evasion, and unique metabolic/hormonal modulations. Interestingly, many of the strategies that enable fetal tolerance parallel those employed by cancer cells to promote angiogenesis, invasion, and immune escape. As such, further elucidating the mechanistic underpinnings of fetal-maternal tolerance may reciprocally provide insights into developing novel cancer immunotherapies as well as understanding the pathogenesis of gestational complications linked to dysregulated tolerance processes.
Collapse
Affiliation(s)
- Jin Soo Joo
- Department of Systems Biology, Yonsei University, Seoul 03722, Korea
| | - Dongeun Lee
- Department of Systems Biology, Yonsei University, Seoul 03722, Korea
| | - Jun Young Hong
- Department of Systems Biology, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
3
|
Petroff MG, Nguyen SL, Ahn SH. Fetal‐placental
antigens and the maternal immune system: Reproductive immunology comes of age. Immunol Rev 2022; 308:25-39. [PMID: 35643905 PMCID: PMC9328203 DOI: 10.1111/imr.13090] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 05/07/2022] [Accepted: 05/11/2022] [Indexed: 12/20/2022]
Abstract
Reproductive physiology and immunology as scientific disciplines each have rich, largely independent histories. The physicians and philosophers of ancient Greece made remarkable observations and inferences to explain regeneration as well as illness and immunity. The scientific enlightenment of the renaissance and the technological advances of the past century have led to the explosion of knowledge that we are experiencing today. Breakthroughs in transplantation, immunology, and reproduction eventually culminated with Medawar’s discovery of acquired immunological tolerance, which helped to explain the transplantation success and failure. Medawar’s musings also keenly pointed out that the fetus apparently breaks these newly discovered rules, and with this, the field of reproductive immunology was launched. As a result of having stemmed from transplantation immunology, scientist still analogizes the fetus to a successful allograft. Although we now know of the fundamental differences between the two, this analogy remains a useful tool to understand how the fetus thrives despite its immunological disparity with the mother. Here, we review the history of reproductive immunology, and how major and minor histocompatibility antigens, blood group antigens, and tissue‐specific “self” antigens from the fetus and transplanted organs parallel and differ.
Collapse
Affiliation(s)
- Margaret G. Petroff
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine Michigan State University East Lansing Michigan USA
- Departments of Microbiology and Molecular Genetics, College of Veterinary Medicine and College of Human Medicine Michigan State University East Lansing Michigan USA
- Cell and Molecular Biology Program, College of Natural Science Michigan State University East Lansing Michigan USA
| | - Sean L. Nguyen
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine Michigan State University East Lansing Michigan USA
- Cell and Molecular Biology Program, College of Natural Science Michigan State University East Lansing Michigan USA
| | - Soo Hyun Ahn
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine Michigan State University East Lansing Michigan USA
| |
Collapse
|
4
|
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
|
5
|
Rizzuto G, Brooks JF, Tuomivaara ST, McIntyre TI, Ma S, Rideaux D, Zikherman J, Fisher SJ, Erlebacher A. Establishment of fetomaternal tolerance through glycan-mediated B cell suppression. Nature 2022; 603:497-502. [PMID: 35236989 PMCID: PMC9592526 DOI: 10.1038/s41586-022-04471-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 01/25/2022] [Indexed: 12/31/2022]
Abstract
Discrimination of self from non-self is fundamental to a wide range of immunological processes1. During pregnancy, the mother does not recognize the placenta as immunologically foreign because antigens expressed by trophoblasts, the placental cells that interface with the maternal immune system, do not activate maternal T cells2. Currently, these activation defects are thought to reflect suppression by regulatory T cells3. By contrast, mechanisms of B cell tolerance to trophoblast antigens have not been identified. Here we provide evidence that glycan-mediated B cell suppression has a key role in establishing fetomaternal tolerance in mice. B cells specific for a model trophoblast antigen are strongly suppressed through CD22-LYN inhibitory signalling, which in turn implicates the sialylated glycans of the antigen as key suppressive determinants. Moreover, B cells mediate the MHC-class-II-restricted presentation of antigens to CD4+ T cells, which leads to T cell suppression, and trophoblast-derived sialoglycoproteins are released into the maternal circulation during pregnancy in mice and humans. How protein glycosylation promotes non-immunogenic placental self-recognition may have relevance to immune-mediated pregnancy complications and to tumour immune evasion. We also anticipate that our findings will bolster efforts to harness glycan biology to control antigen-specific immune responses in autoimmune disease.
Collapse
Affiliation(s)
- G Rizzuto
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - J F Brooks
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - S T Tuomivaara
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
- Center for Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
- Sandler-Moore Mass Spectrometry Core Facility, University of California, San Francisco, CA, USA
| | - T I McIntyre
- Biomedical Sciences Program, University of California San Francisco, San Francisco, CA, USA
| | - S Ma
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, USA
| | - D Rideaux
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, USA
| | - J Zikherman
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
- Biomedical Sciences Program, University of California San Francisco, San Francisco, CA, USA
- Bakar ImmunoX Initiative, University of California San Francisco, San Francisco, CA, USA
| | - S J Fisher
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
- Center for Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
- Biomedical Sciences Program, University of California San Francisco, San Francisco, CA, USA
| | - A Erlebacher
- Center for Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA.
- Biomedical Sciences Program, University of California San Francisco, San Francisco, CA, USA.
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, USA.
- Bakar ImmunoX Initiative, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
6
|
Lewis EL, Xu R, Beltra JC, Ngiow SF, Cohen J, Telange R, Crane A, Sawinski D, Wherry EJ, Porrett PM. NFAT-dependent and -independent exhaustion circuits program maternal CD8 T cell hypofunction in pregnancy. J Exp Med 2022; 219:e20201599. [PMID: 34882194 PMCID: PMC8666877 DOI: 10.1084/jem.20201599] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 10/09/2021] [Accepted: 11/18/2021] [Indexed: 11/21/2022] Open
Abstract
Pregnancy is a common immunization event, but the molecular mechanisms and immunological consequences provoked by pregnancy remain largely unknown. We used mouse models and human transplant registry data to reveal that pregnancy induced exhausted CD8 T cells (Preg-TEX), which associated with prolonged allograft survival. Maternal CD8 T cells shared features of exhaustion with CD8 T cells from cancer and chronic infection, including transcriptional down-regulation of ribosomal proteins and up-regulation of TOX and inhibitory receptors. Similar to other models of T cell exhaustion, NFAT-dependent elements of the exhaustion program were induced by fetal antigen in pregnancy, whereas NFAT-independent elements did not require fetal antigen. Despite using conserved molecular circuitry, Preg-TEX cells differed from TEX cells in chronic viral infection with respect to magnitude and dependency of T cell hypofunction on NFAT-independent signals. Altogether, these data reveal the molecular mechanisms and clinical consequences of maternal CD8 T cell hypofunction and identify pregnancy as a previously unappreciated context in which T cell exhaustion may occur.
Collapse
Affiliation(s)
- Emma L. Lewis
- Department of Obstetrics and Gynecology, The University of Pennsylvania, Philadelphia, PA
| | - Rong Xu
- Department of Surgery, The University of Pennsylvania, Philadelphia, PA
| | - Jean-Christophe Beltra
- Department of Systems Pharmacology and Translational Therapeutics, The University of Pennsylvania, Philadelphia, PA
- Institute for Immunology, University of Pennsylvania, Philadelphia, PA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA
| | - Shin Foong Ngiow
- Department of Systems Pharmacology and Translational Therapeutics, The University of Pennsylvania, Philadelphia, PA
- Institute for Immunology, University of Pennsylvania, Philadelphia, PA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA
| | - Jordana Cohen
- Department of Medicine, The University of Pennsylvania, Philadelphia, PA
| | - Rahul Telange
- Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL
| | - Alexander Crane
- Department of Surgery, The University of Pennsylvania, Philadelphia, PA
| | - Deirdre Sawinski
- Department of Medicine, The University of Pennsylvania, Philadelphia, PA
| | - E. John Wherry
- Department of Systems Pharmacology and Translational Therapeutics, The University of Pennsylvania, Philadelphia, PA
- Institute for Immunology, University of Pennsylvania, Philadelphia, PA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA
| | - Paige M. Porrett
- Department of Surgery, The University of Pennsylvania, Philadelphia, PA
- Institute for Immunology, University of Pennsylvania, Philadelphia, PA
- Department of Surgery, The University of Alabama at Birmingham, Birmingham, AL
- Comprehensive Transplant Institute, The University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
7
|
Nellore A, Killian JT, Porrett PM. Memory B Cells in Pregnancy Sensitization. Front Immunol 2021; 12:688987. [PMID: 34276679 PMCID: PMC8278195 DOI: 10.3389/fimmu.2021.688987] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/10/2021] [Indexed: 11/13/2022] Open
Abstract
Memory B cells play an important role in immunity to pathogens as these cells are poised to rapidly differentiate into antibody-secreting cells upon antigen re-encounter. Memory B cells also develop over the course of HLA-sensitization during pregnancy and transplantation. In this review, we discuss the potential contribution of memory B cells to pregnancy sensitization as well as the impact of these cells on transplant candidacy and outcomes. We start by summarizing how B cell subsets are altered in pregnancy and discuss what is known about HLA-specific B cell responses given our current understanding of fetal antigen availability in maternal secondary lymphoid tissues. We then review the molecular mechanisms governing the generation and maintenance of memory B cells during infection - including the role of T follicular helper cells - and discuss the experimental evidence for the development of these cells during pregnancy. Finally, we discuss how memory B cells impact access to transplantation and transplant outcomes for a range of transplant recipients.
Collapse
Affiliation(s)
- Anoma Nellore
- Department of Medicine, University of Alabama at Birmingham School of Medicine, Birmingham, AL, United States
| | - John T. Killian
- Department of Surgery, University of Alabama at Birmingham School of Medicine, Birmingham, AL, United States
| | - Paige M. Porrett
- Department of Surgery, University of Alabama at Birmingham School of Medicine, Birmingham, AL, United States
| |
Collapse
|
8
|
Bai K, Li X, Zhong J, Ng EHY, Yeung WSB, Lee CL, Chiu PCN. Placenta-Derived Exosomes as a Modulator in Maternal Immune Tolerance During Pregnancy. Front Immunol 2021; 12:671093. [PMID: 34046039 PMCID: PMC8144714 DOI: 10.3389/fimmu.2021.671093] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/26/2021] [Indexed: 12/12/2022] Open
Abstract
Exosomes are a subset of extracellular vesicles with an average diameter of ~100nm. Exosomes are released by all cells through an endosome-dependent pathway and carry nucleic acids, proteins, lipids, cytokines and metabolites, mirroring the state of the originating cells. The function of exosomes has been implicated in various reproduction processes, such as embryo development, implantation, decidualization and placentation. Placenta-derived exosomes (pEXO) can be detected in the maternal blood as early as 6 weeks after conception and their levels increase with gestational age. Importantly, alternations in the molecular signatures of pEXO are observed in pregnancy-related complications. Thus, these differentially expressed molecules could be the potential biomarkers for diagnosis of the pregnancy-associated diseases. Recent studies have demonstrated that pEXO play a key role in the establishment of maternal immune tolerance, which is critical for a successful pregnancy. To gain a better understanding of the underlying mechanism, we highlighted the advanced studies of pEXO on immune cells in pregnancy.
Collapse
Affiliation(s)
- Kunfeng Bai
- Department of Obstetrics and Gynaecology, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong.,The University of Hong Kong Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Xintong Li
- Department of Obstetrics and Gynaecology, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Jiangming Zhong
- Department of Obstetrics and Gynaecology, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Ernest H Y Ng
- Department of Obstetrics and Gynaecology, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong.,The University of Hong Kong Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - William S B Yeung
- The University of Hong Kong Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Cheuk-Lun Lee
- Department of Obstetrics and Gynaecology, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong.,The University of Hong Kong Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Philip C N Chiu
- Department of Obstetrics and Gynaecology, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong.,The University of Hong Kong Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
9
|
Kinder JM, Turner LH, Stelzer IA, Miller-Handley H, Burg A, Shao TY, Pham G, Way SS. CD8 + T Cell Functional Exhaustion Overrides Pregnancy-Induced Fetal Antigen Alloimmunization. Cell Rep 2021; 31:107784. [PMID: 32579916 PMCID: PMC7383938 DOI: 10.1016/j.celrep.2020.107784] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 04/10/2020] [Accepted: 05/27/2020] [Indexed: 12/19/2022] Open
Abstract
Pregnancy necessitates physiological exposure, and often re-exposure, to foreign fetal alloantigens. The consequences after pregnancy are highly varied, with evidence of both alloimmunization and expanded tolerance phenotypes. We show that pregnancy primes the accumulation of fetal-specific maternal CD8+ T cells and their persistence as an activated memory pool after parturition. Cytolysis and the potential for robust secondary expansion occurs with antigen re-encounter in non-reproductive contexts. Comparatively, CDS+ T cell functional exhaustion associated with increased PD-1 and LAG-3 expression occurs with fetal antigen re-stimulation during subsequent pregnancy. PD-L1/LAG-3 neutralization unleashes the activation of fetal-specific CD8+ T cells, causing fetal wastage selectively during secondary but not primary pregnancy. Thus, CD8+ T cells with fetal alloantigen specificity persist in mothers after pregnancy, and protection against fetal wastage in subsequent pregnancies is maintained by their unique susceptibility to functional exhaustion. Together, distinct mechanisms whereby fetal tolerance is maintained during primary compared with subsequent pregnancies are demonstrated. Expecting mothers are immunologically aware of, but do not reject, genetically foreign tissues of the developing fetus. Comparing tolerance occuring during first and subsequent pregnancies, Kinder et al. show that activated memory CD8+ T cells primed by prior pregnancy are uniquely prone to functional exhaustion with fetal antigen re-stimulation.
Collapse
Affiliation(s)
- Jeremy M Kinder
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA.
| | - Lucien H Turner
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Ina A Stelzer
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Hilary Miller-Handley
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Ashley Burg
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Tzu-Yu Shao
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Giang Pham
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Sing Sing Way
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA.
| |
Collapse
|
10
|
Navigating the murky waters of maternal fetal tolerance: Put big data at the helm. Hum Immunol 2021; 82:340-345. [PMID: 33589281 DOI: 10.1016/j.humimm.2021.01.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 01/05/2021] [Accepted: 01/21/2021] [Indexed: 12/13/2022]
Abstract
The last 20 years have seen significant progress in our understanding of the maternal T cell response to the fetus. However, major questions still remain. In this review, we discuss the contribution of Big Data approaches to our evolving understanding of maternal fetal tolerance. This includes investigations which have employed systems immunology, high dimensional mass spectrometry, whole genome transcriptomics, as well as single-cell RNA-sequencing. These studies reveal "new" mechanisms of T cell dysfunction that are conserved across immune responses and highlight the breadth and magnitude of the systemic maternal response to the fetus during pregnancy.
Collapse
|
11
|
Huber WJ, Sauerbrun-Cutler MT, Krueger PM, Sharma S. Novel predictive and therapeutic options for better pregnancy outcome in frozen embryo transfer cycles. Am J Reprod Immunol 2020; 85:e13300. [PMID: 32654307 DOI: 10.1111/aji.13300] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 07/01/2020] [Indexed: 12/25/2022] Open
Abstract
Since 1978, in the first decades of in vitro fertilization (IVF), the use of ovarian hyperstimulation allowed for the development and transfer of multiple embryos. As IVF technology improved, the number of multiple pregnancies increased, which led to gradual reduction in the number of embryos that were transferred. Embryo freezing (vitrification) was recommended to allow subsequent transfer if the fresh cycle was unsuccessful. However, experimentation has continued to improve pregnancy outcomes. We discuss here the significance of frozen embryo transfer cycle and the impact of uterine and peripheral immunity dominated by NK cells and regulatory T cells and human chorionic gonadotropin on pregnancy outcome in this innovative mode of IVF therapy.
Collapse
Affiliation(s)
- Warren J Huber
- Department of Obstetrics and Gynecology, LSUHSC School of Medicine and The Fertility Institute, New Orleans, LA, USA
| | - May-Tal Sauerbrun-Cutler
- Department of Obstetrics and Gynecology, Alpert Medical School of Brown University, Women & Infants Hospital, Providence, RI, USA
| | - Paula M Krueger
- Department of Pediatrics, Alpert Medical School of Brown University, Women & Infants Hospital, Providence, RI, USA
| | - Surendra Sharma
- Department of Pediatrics, Alpert Medical School of Brown University, Women & Infants Hospital, Providence, RI, USA
| |
Collapse
|
12
|
Porrett PM. Biologic mechanisms and clinical consequences of pregnancy alloimmunization. Am J Transplant 2018; 18:1059-1067. [PMID: 29369525 DOI: 10.1111/ajt.14673] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 01/16/2018] [Accepted: 01/18/2018] [Indexed: 01/25/2023]
Abstract
Alloimmunization occurs during pregnancy when tissue antigens derived from the fetus and/or placenta prime maternal immune cells to divide and differentiate. For many women, the result of pregnancy alloimmunization is the formation of anti-HLA antibody that can endure for decades and preclude transplantation by limiting donor compatibility. Pregnancy alloimmunization may also generate memory B cells that can rapidly produce anti-HLA antibody after transplantation as well as pathogenic memory T cells, which pose a threat to graft survival. However, emerging data suggest that pregnancy also programs the differentiation of anergic, dysfunctional, and regulatory T cell populations, which may not mediate accelerated graft rejection. Hence, some of the immune mechanisms responsible for maternal immunologic tolerance of the fetus may persist into postpartum life and affect the response to an allograft. This review discusses these emerging data as well as the persistent knowledge gaps that affect women at multiple stages of their transplant care.
Collapse
|
13
|
Jasti S, Farahbakhsh M, Nguyen S, Petroff BK, Petroff MG. Immune response to a model shared placenta/tumor-associated antigen reduces cancer risk in parous mice. Biol Reprod 2017; 96:134-144. [PMID: 28395331 DOI: 10.1095/biolreprod.116.144907] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 11/15/2016] [Indexed: 12/21/2022] Open
Abstract
During human pregnancy, paternally inherited antigens expressed by the fetal-placental unit can elicit expansion of antigen-specific CD8+ T cells. These cells can persist for years as memory T cells, but their effects on long-term maternal health are unknown. Shared placenta/tumor-associated antigens are expressed by placenta and tumors, but are minimally expressed or absent in normal adult tissues. We hypothesized that maternal T cells elicited against these antigens can alter risk of cancers expressing the same antigen after pregnancy, and tested this in mice using chicken ovalbumin (OVA) as a surrogate shared placenta/tumor antigen. Hemizygous OVA transgenic males were bred to wild-type C57BL/6 females (H2b haplotype) such that the fetuses inherited and expressed OVA. Maternal OVA/H2Kb-specific CD8+ T cells became detectable during gestation, and persisted in some animals for up to 24 weeks. To determine whether these cells might influence growth of OVA-expressing tumors in OVA-bred females, E.G7-OVA thymoma cells were inoculated subcutaneously in OVA-bred, wild-type bred, and virgin females, and monitored for growth. OVA-bred mice had prolonged survival as compared to virgin mice and the progression of tumors was delayed in comparison to wild-type bred and virgin females. Thus, paternally inherited OVA antigen elicited a CD8+ T cell response during pregnancy that was associated with delayed growth of OVA-expressing tumors following pregnancy. These data suggest a possible role of antigen-specific T cells in protecting parous females against tumors bearing shared placenta/tumor antigens.
Collapse
Affiliation(s)
- Susmita Jasti
- Departments of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Mina Farahbakhsh
- Departments of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Sean Nguyen
- Cell and Molecular Biology Program, Michigan State University, East Lansing, Michigan, USA
| | - Brian K Petroff
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan, USA
| | - Margaret G Petroff
- Departments of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas, USA.,Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan, USA.,Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
14
|
Maternal dendrimer-based therapy for inflammation-induced preterm birth and perinatal brain injury. Sci Rep 2017; 7:6106. [PMID: 28733619 PMCID: PMC5522481 DOI: 10.1038/s41598-017-06113-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 06/08/2017] [Indexed: 01/06/2023] Open
Abstract
Preterm birth is a major risk factor for adverse neurological outcomes in ex-preterm children, including motor, cognitive, and behavioral disabilities. N-acetyl-L-cysteine therapy has been used in clinical studies; however, it requires doses that cause significant side effects. In this study, we explore the effect of low dose N-acetyl-L-cysteine therapy, delivered using a targeted, systemic, maternal, dendrimer nanoparticle (DNAC), in a mouse model of intrauterine inflammation. Our results demonstrated that intraperitoneal maternal DNAC administration significantly reduced the preterm birth rate and altered placental immune profile with decreased CD8+ T-cell infiltration. Furthermore, we demonstrated that DNAC improved neurobehavioral outcomes and reduced fetal neuroinflammation and long-term microglial activation in offspring. Our study is the first to provide evidence for the role of CD8+ T-cell in the maternal-fetal interface during inflammation and further support the efficacy of DNAC in preventing preterm birth and prematurity-related outcomes.
Collapse
|
15
|
Barton BM, Xu R, Wherry EJ, Porrett PM. Pregnancy promotes tolerance to future offspring by programming selective dysfunction in long-lived maternal T cells. J Leukoc Biol 2017; 101:975-987. [PMID: 27810945 DOI: 10.1189/jlb.1a0316-135r] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 08/31/2016] [Accepted: 10/11/2016] [Indexed: 11/24/2022] Open
Abstract
Fetal antigen available during pregnancy induces the proliferation of maternal T cells. It is unknown, however, whether these antigen-activated T cells differentiate into long-lived memory T cells that are capable of mediating rapid-recall responses to tissue antigens. To test the hypothesis that pregnancy induces an alternative fate in fetal-specific maternal T cells, we used a murine model to track longitudinally fetal-specific T cells in pregnant and postpartum animals and test the response of these cells when challenged with the same antigen during sequential pregnancy or skin transplantation. Fetal-specific CD8+ T cells were robustly primed during pregnancy but failed to acquire robust effector functions. These primed cells persisted long term in postpartum animals, frequently maintained a programmed death 1 (PD-1)+ phenotype, and failed to expand or produce cytokines robustly in response to second pregnancy or skin transplantation. However, whereas there was no impact on second pregnancy as a result of the persistence of fetal-primed memory CD8+ T cells in the mother, skin grafts bearing the same antigen were rejected more rapidly. Altogether, our data suggest that fetal antigen exposure during pregnancy induces the differentiation of long-lived maternal CD8+ T cells with context-dependent, selective effector dysfunction. This programmed effector dysfunction provides temporal and systemic restraint of maternal anti-fetal alloreactivity to promote reproductive fitness efficiently, while preserving potentially protective effector T cell responses.
Collapse
Affiliation(s)
- Brendan M Barton
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Rong Xu
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; and
| | - E John Wherry
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Paige M Porrett
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; and
| |
Collapse
|
16
|
Juiz NA, Solana ME, Acevedo GR, Benatar AF, Ramirez JC, da Costa PA, Macedo AM, Longhi SA, Schijman AG. Different genotypes of Trypanosoma cruzi produce distinctive placental environment genetic response in chronic experimental infection. PLoS Negl Trop Dis 2017; 11:e0005436. [PMID: 28273076 PMCID: PMC5358786 DOI: 10.1371/journal.pntd.0005436] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 03/20/2017] [Accepted: 02/24/2017] [Indexed: 12/15/2022] Open
Abstract
Congenital infection of Trypanosoma cruzi allows transmission of this parasite through generations. Despite the problematic that this entails, little is known about the placenta environment genetic response produced against infection. We performed functional genomics by microarray analysis in C57Bl/6J mice comparing placentas from uninfected animals and from animals infected with two different T. cruzi strains: K98, a clone of the non-lethal myotropic CA-I strain (TcI), and VD (TcVI), isolated from a human case of congenital infection. Analysis of networks by GeneMANIA of differentially expressed genes showed that “Secretory Granule” was a pathway down-regulated in both infected groups, whereas “Innate Immune Response” and “Response to Interferon-gamma” were pathways up-regulated in VD infection but not in K98. Applying another approach, the GSEA algorithm that detects small changes in predetermined gene sets, we found that metabolic processes, transcription and macromolecular transport were down-regulated in infected placentas environment and some pathways related to cascade signaling had opposite regulation: over-represented in VD and down-regulated in K98 group. We also have found a stronger tropism to the placental organ by VD strain, by detection of parasite DNA and RNA, suggesting living parasites. Our study is the first one to describe in a murine model the genetic response of placental environment to T. cruzi infection and suggests the development of a strong immune response, parasite genotype-dependent, to the detriment of cellular metabolism, which may contribute to control infection preventing the risk of congenital transmission. Congenital transmission of Trypanosoma cruzi, the causative agent of Chagas disease, remains a problem of global public health impact in endemic areas where vectorial and transfusional transmission have been controlled and in non-endemic countries due to migration movements. Little is known about how the parasite´s presence and genetic variability affect placental capacity to protect the fetus. This study explores, for the first time, the effects of placental environment infection by analyzing parasite persistence and gene expression using functional genomics and biological network analyses in mice infected by two strains of T. cruzi, with differential capacity of congenital transmission. The infection with the strain with a stronger placental tropism was associated to a higher degree of up-regulation in genes related to innate immunity and response to interferon-gamma. Our findings suggest that the placental environment exerts a strong immune response in detriment of cellular metabolism modulated by the parasite strain. These findings constitute a significant contribution to better understand the mechanisms causing congenital infection of T. cruzi.
Collapse
Affiliation(s)
- Natalia Anahí Juiz
- Grupo de Biología Molecular de la Enfermedad de Chagas (LaBMECh), Instituto de Investigaciones en Ingeniería Genética y Biología Molecular ‘‘Dr. Héctor N. Torres” (INGEBI-CONICET), Buenos Aires, Argentina
| | - María Elisa Solana
- Departamento de Microbiología, Parasitología e Inmunología, Instituto de Investigaciones en Microbiología y Parasitología Médica (IMPaM), Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Gonzalo Raúl Acevedo
- Grupo de Biología Molecular de la Enfermedad de Chagas (LaBMECh), Instituto de Investigaciones en Ingeniería Genética y Biología Molecular ‘‘Dr. Héctor N. Torres” (INGEBI-CONICET), Buenos Aires, Argentina
| | - Alejandro Francisco Benatar
- Grupo de Biología Molecular de la Enfermedad de Chagas (LaBMECh), Instituto de Investigaciones en Ingeniería Genética y Biología Molecular ‘‘Dr. Héctor N. Torres” (INGEBI-CONICET), Buenos Aires, Argentina
| | - Juan Carlos Ramirez
- Grupo de Biología Molecular de la Enfermedad de Chagas (LaBMECh), Instituto de Investigaciones en Ingeniería Genética y Biología Molecular ‘‘Dr. Héctor N. Torres” (INGEBI-CONICET), Buenos Aires, Argentina
| | - Priscilla Almeida da Costa
- Departamento de Bioquímica e Inmunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Andrea Mara Macedo
- Departamento de Bioquímica e Inmunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Silvia Andrea Longhi
- Grupo de Biología Molecular de la Enfermedad de Chagas (LaBMECh), Instituto de Investigaciones en Ingeniería Genética y Biología Molecular ‘‘Dr. Héctor N. Torres” (INGEBI-CONICET), Buenos Aires, Argentina
| | - Alejandro G. Schijman
- Grupo de Biología Molecular de la Enfermedad de Chagas (LaBMECh), Instituto de Investigaciones en Ingeniería Genética y Biología Molecular ‘‘Dr. Héctor N. Torres” (INGEBI-CONICET), Buenos Aires, Argentina
- * E-mail:
| |
Collapse
|
17
|
Tannetta D, Masliukaite I, Vatish M, Redman C, Sargent I. Update of syncytiotrophoblast derived extracellular vesicles in normal pregnancy and preeclampsia. J Reprod Immunol 2016; 119:98-106. [PMID: 27613663 DOI: 10.1016/j.jri.2016.08.008] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 08/12/2016] [Accepted: 08/23/2016] [Indexed: 11/24/2022]
Abstract
The release of extracellular vesicles (EV) by the syncytiotrophoblast (STB) may be an important mechanism by which the placenta signals to the mother. STB derived EV (STBEV) are comprised predominantly of exosomes (50-150nm) and microvesicles (100-1000nm) that contain bioactive mediators such as proteins, nucleic acids and lipids. They, along with larger syncytial nuclear aggregates are released by the STB into the maternal circulation throughout gestation in normal pregnancy where they appear to have an immunoregulatory role, inhibiting T cell and NK cell responses. In pre-eclampsia (PE) STBEV are released in significantly increased numbers and have pro-inflammatory, anti-angiogenic and procoagulant activity, implicating them in the maternal systemic inflammation, endothelial dysfunction and activation of the clotting system which typifies the disorder. Research has focused on understanding the biological significance of STBEV by measuring their size and repertoire of molecules carried and how they differ in normal pregnancy and PE, using techniques such as Nanoparticle Tracking Analysis, flow cytometry and mass spectrometry. We have also found alterations in STBEV surface glycans associated with PE. The goal is to better understand the role STBEV play in normal pregnancy and PE and whether they are potential biomarkers of placental pathology and therapeutic targets in PE.
Collapse
Affiliation(s)
- Dionne Tannetta
- Department of Food and Nutritional Sciences, PO Box 226, Whiteknights, Reading RG6 6AP, UK.
| | - Ieva Masliukaite
- Center for Reproductive Medicine, TKsO-266, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| | - Manu Vatish
- Nuffield Department of Obstetrics & Gynaecology, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK.
| | - Christopher Redman
- Nuffield Department of Obstetrics & Gynaecology, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK.
| | - Ian Sargent
- Nuffield Department of Obstetrics & Gynaecology, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU, UK.
| |
Collapse
|
18
|
Lima PDA, Zhang J, Dunk C, Lye SJ, Croy BA. Leukocyte driven-decidual angiogenesis in early pregnancy. Cell Mol Immunol 2014; 11:522-37. [PMID: 25066422 DOI: 10.1038/cmi.2014.63] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 06/21/2014] [Accepted: 06/22/2014] [Indexed: 12/15/2022] Open
Abstract
Successful pregnancy and long-term, post-natal maternal and offspring cardiac, vascular and metabolic health require key maternal cardiovascular adaptations over gestation. Within the pregnant decidualizing uterus, coordinated vascular, immunological and stromal cell changes occur. Considerable attention has been given to the roles of uterine natural killer (uNK) cells in initiating decidual spiral arterial remodeling, a process normally completed by mid-gestation in mice and in humans. However, leukocyte roles in much earlier, region specific, decidual vascular remodeling are now being defined. Interest in immune cell-promoted vascular remodeling is driven by vascular aberrations that are reported in human gestational complications such as infertility, recurrent spontaneous abortion, preeclampsia (PE) and fetal growth restriction. Appropriate maternal cardiovascular responses during pregnancy protect mothers and their children from later cardiovascular disease risk elevation. One of the earliest uterine responses to pregnancy in species with hemochorial placentation is stromal cell decidualization, which creates unique niches for angiogenesis and leukocyte recruitment. In early decidua basalis, the aspect of the implantation site that will cradle the developing placenta and provide the major blood vessels to support mature placental functions, leukocytes are greatly enriched and display specialized properties. UNK cells, the most abundant leukocyte subset in early decidua basalis, have angiogenic abilities and are essential for normal early decidual angiogenesis. The regulation of uNK cells and their roles in determining maternal and progeny cardiovascular health over pregnancy and postpartum are discussed.
Collapse
Affiliation(s)
- Patricia D A Lima
- Ottawa Hospital Research Institute, The Ottawa Hospital General Campus, Critical Care Wing, Ottawa, ON, Canada
| | - Jianhong Zhang
- Research Centre for Women's and Infants' Health, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Caroline Dunk
- Research Centre for Women's and Infants' Health, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Stephen J Lye
- 1] Research Centre for Women's and Infants' Health, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada [2] Department of Physiology and University of Toronto, Toronto, ON, Canada [3] Department of Obstetrics and Gynecology, University of Toronto, Toronto, ON, Canada
| | - B Anne Croy
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| |
Collapse
|
19
|
Xin L, Ertelt JM, Rowe JH, Jiang TT, Kinder JM, Chaturvedi V, Elahi S, Way SS. Cutting edge: committed Th1 CD4+ T cell differentiation blocks pregnancy-induced Foxp3 expression with antigen-specific fetal loss. THE JOURNAL OF IMMUNOLOGY 2014; 192:2970-4. [PMID: 24591368 DOI: 10.4049/jimmunol.1302678] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Pregnancy stimulates induced Foxp3 expression among maternal CD4(+) T cells with fetal specificity. Although sustained maternal regulatory CD4(+) T cell (Treg) expansion is essential for maintaining fetal tolerance during pregnancy, the necessity for Foxp3(+) cells with fetal specificity remains undefined. In this study, we demonstrate that mitigating Treg differentiation among maternal CD4(+) T cells with a single surrogate fetal specificity elicits Ag-specific fetal loss. Using recombinant Listeria monocytogenes to prime stably differentiated Th1 CD4(+) T cells with fetal I-A(b):2W1S55-68 specificity refractory to pregnancy-induced Foxp3 expression, we show that Ag delivery by cytoplasmic L. monocytogenes causes selective loss of 2W1S(+) offspring through CD4 cell- and IFN-γ-dependent pathways. In contrast, CD4(+) T cells primed by L. monocytogenes restricted from the cell cytoplasm are markedly more plastic for induced Foxp3 expression, with normal pregnancy outcomes. Thus, committed Th1 polarization blocks pregnancy induced Treg differentiation among maternal CD4(+) T cells with fetal specificity and triggers Ag-specific fetal loss.
Collapse
Affiliation(s)
- Lijun Xin
- Division of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229
| | | | | | | | | | | | | | | |
Collapse
|