1
|
Yu J, Yan Y, Li S, Xu Y, Parolia A, Rizvi S, Wang W, Zhai Y, Xiao R, Li X, Liao P, Zhou J, Okla K, Lin H, Lin X, Grove S, Wei S, Vatan L, Hu J, Szumilo J, Kotarski J, Freeman ZT, Skala S, Wicha M, Cho KR, Chinnaiyan AM, Schon S, Wen F, Kryczek I, Wang S, Chen L, Zou W. Progestogen-driven B7-H4 contributes to onco-fetal immune tolerance. Cell 2024; 187:4713-4732.e19. [PMID: 38968937 PMCID: PMC11344674 DOI: 10.1016/j.cell.2024.06.012] [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: 07/19/2023] [Revised: 02/09/2024] [Accepted: 06/09/2024] [Indexed: 07/07/2024]
Abstract
Immune tolerance mechanisms are shared in cancer and pregnancy. Through cross-analyzing single-cell RNA-sequencing data from multiple human cancer types and the maternal-fetal interface, we found B7-H4 (VTCN1) is an onco-fetal immune tolerance checkpoint. We showed that genetic deficiency of B7-H4 resulted in immune activation and fetal resorption in allogeneic pregnancy models. Analogously, B7-H4 contributed to MPA/DMBA-induced breast cancer progression, accompanied by CD8+ T cell exhaustion. Female hormone screening revealed that progesterone stimulated B7-H4 expression in placental and breast cancer cells. Mechanistically, progesterone receptor (PR) bound to a newly identified -58 kb enhancer, thereby mediating B7-H4 transcription via the PR-P300-BRD4 axis. PR antagonist or BRD4 degrader potentiated immunotherapy in a murine B7-H4+ breast cancer model. Thus, our work unravels a mechanistic and biological connection of a female sex hormone (progesterone) to onco-fetal immune tolerance via B7-H4 and suggests that the PR-P300-BRD4 axis is targetable for treating B7-H4+ cancer.
Collapse
Affiliation(s)
- Jiali Yu
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Yijian Yan
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Shasha Li
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Ying Xu
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Abhijit Parolia
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Syed Rizvi
- Department of Chemical Engineering, University of Michigan School of Engineering, Ann Arbor, MI, USA
| | - Weichao Wang
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Yiwen Zhai
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Rongxin Xiao
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Xiong Li
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Peng Liao
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Jiajia Zhou
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Karolina Okla
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA; Department of Oncological Gynecology and Gynecology, Medical University of Lublin, Lublin, Poland
| | - Heng Lin
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Xun Lin
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Sara Grove
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Shuang Wei
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Linda Vatan
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Jiantao Hu
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Justyna Szumilo
- Department of Clinical Pathomorphology, Medical University of Lublin, Lublin, Poland
| | - Jan Kotarski
- Department of Oncological Gynecology and Gynecology, Medical University of Lublin, Lublin, Poland
| | - Zachary T Freeman
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Stephanie Skala
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Max Wicha
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Kathleen R Cho
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Arul M Chinnaiyan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA; Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI, USA; Howard Hughes Medical Institute, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Samantha Schon
- Department of Obstetrics and Gynecology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Fei Wen
- Department of Chemical Engineering, University of Michigan School of Engineering, Ann Arbor, MI, USA
| | - Ilona Kryczek
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Shaomeng Wang
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Lieping Chen
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Weiping Zou
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA; Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA; Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, USA; Graduate Program in Cancer Biology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
2
|
Chin PY, Chan HY, Kieffer TEC, Prins JR, Russell DL, Davies MJ, Robertson SA. Clomiphene Citrate Administered in Periconception Phase Causes Fetal Loss and Developmental Impairment in Mice. Endocrinology 2024; 165:bqae047. [PMID: 38608138 PMCID: PMC11197002 DOI: 10.1210/endocr/bqae047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 04/02/2024] [Accepted: 04/10/2024] [Indexed: 04/14/2024]
Abstract
Clomiphene citrate is a common treatment for ovulation induction in subfertile women, but its use is associated with elevated risk of adverse perinatal outcomes and birth defects. To investigate the biological plausibility of a causal relationship, this study investigated the consequences in mice for fetal development and pregnancy outcome of periconception clomiphene citrate administration at doses approximating human exposures. A dose-dependent adverse effect of clomiphene citrate given twice in the 36 hours after mating was seen, with a moderate dose of 0.75 mg/kg sufficient to cause altered reproductive outcomes in 3 independent cohorts. Viable pregnancy was reduced by 30%, late gestation fetal weight was reduced by 16%, and ∼30% of fetuses exhibited delayed development and/or congenital abnormalities not seen in control dams, including defects of the lung, kidney, liver, eye, skin, limbs, and umbilicus. Clomiphene citrate also caused a 30-hour average delay in time of birth, and elevated rate of pup death in the early postnatal phase. In surviving offspring, growth trajectory tracking and body morphometry analysis at 20 weeks of age showed postweaning growth and development similar to controls. A dysregulated inflammatory response in the endometrium was observed and may contribute to the underlying pathophysiological mechanism. These results demonstrate that in utero exposure to clomiphene citrate during early pregnancy can compromise implantation and impact fetal growth and development, causing adverse perinatal outcomes. The findings raise the prospect of similar iatrogenic effects in women where clomiphene citrate may be present in the periconception phase unless its use is well-supervised.
Collapse
Affiliation(s)
- Peck Y Chin
- Robinson Research Institute and School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia
| | - Hon Yeung Chan
- Robinson Research Institute and School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia
| | - Tom E C Kieffer
- Robinson Research Institute and School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
- Amsterdam University Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Jelmer R Prins
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| | - Darryl L Russell
- Robinson Research Institute and School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia
| | - Michael J Davies
- Robinson Research Institute and School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia
| | - Sarah A Robertson
- Robinson Research Institute and School of Biomedicine, University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
3
|
Pollard JM, Hynes G, Yin D, Mandal M, Gounari F, Alegre ML, Chong AS. Pregnancy dedifferentiates memory CD8+ T cells into hypofunctional cells with exhaustion-enriched programs. JCI Insight 2024; 9:e176381. [PMID: 38771643 PMCID: PMC11383355 DOI: 10.1172/jci.insight.176381] [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: 10/03/2023] [Accepted: 05/15/2024] [Indexed: 05/23/2024] Open
Abstract
Alloreactive memory, unlike naive, CD8+ T cells resist transplantation tolerance protocols and are a critical barrier to long-term graft acceptance in the clinic. We here show that semiallogeneic pregnancy successfully reprogrammed memory fetus/graft-specific CD8+ T cells (TFGS) toward hypofunction. Female C57BL/6 mice harboring memory CD8+ T cells generated by the rejection of BALB/c skin grafts and then mated with BALB/c males achieved rates of pregnancy comparable with naive controls. Postpartum CD8+ TFGS from skin-sensitized dams upregulated expression of T cell exhaustion (TEX) markers (Tox, Eomes, PD-1, TIGIT, and Lag3). Transcriptional analysis corroborated an enrichment of canonical TEX genes in postpartum memory TFGS and revealed a downregulation of a subset of memory-associated transcripts. Strikingly, pregnancy induced extensive epigenetic modifications of exhaustion- and memory-associated genes in memory TFGS, whereas minimal epigenetic modifications were observed in naive TFGS. Finally, postpartum memory TFGS durably expressed the exhaustion-enriched phenotype, and their susceptibility to transplantation tolerance was significantly restored compared with memory TFGS. These findings advance the concept of pregnancy as an epigenetic modulator inducing hypofunction in memory CD8+ T cells that has relevance not only for pregnancy and transplantation tolerance, but also for tumor immunity and chronic infections.
Collapse
Affiliation(s)
| | - Grace Hynes
- Section of Transplantation, Department of Surgery, and
| | - Dengping Yin
- Section of Transplantation, Department of Surgery, and
| | - Malay Mandal
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Fotini Gounari
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, Illinois, USA
- Department of Immunology, Mayo Clinic, Phoenix, Arizona, USA
| | - Maria-Luisa Alegre
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Anita S Chong
- Section of Transplantation, Department of Surgery, and
| |
Collapse
|
4
|
Li QH, Zhao QY, Yang WJ, Jiang AF, Ren CE, Meng YH. Beyond Immune Balance: The Pivotal Role of Decidual Regulatory T Cells in Unexplained Recurrent Spontaneous Abortion. J Inflamm Res 2024; 17:2697-2710. [PMID: 38707955 PMCID: PMC11070170 DOI: 10.2147/jir.s459263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 04/18/2024] [Indexed: 05/07/2024] Open
Abstract
Recurrent spontaneous abortion (RSA) is defined as two or more consecutive pregnancy failures, which brings tremendous stress to women of childbearing age and seriously affects family well-being. However, the reason in about 50% of cases remains unknown and is defined as unexplained recurrent spontaneous abortion (URSA). The immunological perspective in URSA has attracted widespread attention in recent years. The embryo is regarded as a semi-allogeneic graft to the mother. A successful pregnancy requires transition to an immune environment conducive to embryo survival at the maternal-fetal interface. As an important member of regulatory immunity, regulatory T (Treg) cells play a key role in regulating immune tolerance at the maternal-fetal interface. This review will focus on the phenotypic plasticity and lineage stability of Treg cells to illustrate its relationship with URSA.
Collapse
Affiliation(s)
- Qing-Hui Li
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261021, People’s Republic of China
- Center of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China
| | - Qiu-Yan Zhao
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261021, People’s Republic of China
| | - Wei-Jing Yang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261021, People’s Republic of China
| | - Ai-Fang Jiang
- Center of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China
| | - Chun-E Ren
- Center of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China
| | - Yu-Han Meng
- Center of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China
| |
Collapse
|
5
|
Norman SJ, Fontus G, Forestier C, Hiba T, Colon Pagan S, Osondu M, Shylovich V. The Protective Effect of Abortion on Preeclampsia: An Analysis of Current Research. Cureus 2024; 16:e54131. [PMID: 38496185 PMCID: PMC10942113 DOI: 10.7759/cureus.54131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2024] [Indexed: 03/19/2024] Open
Abstract
A review of the current literature on preeclampsia (PE) confirms that this pregnancy complication remains a common cause of maternal mortality. Within the last several decades, obstetric and gynecological researchers worldwide have indicated an association between prior abortions and the development of PE. Different studies have debated whether abortion is a protective or risk factor for PE. However, the most current literature demonstrates a stronger likelihood that a positive history of abortions will offer a protective effect against PE. This association has been supported by advancements in the reproductive immunology literature, which states complex fetal and paternal pathological mechanisms help to build maternal immunological tolerance, thus protecting expectant mothers from pregnancy complications. This literature review will compare studies supporting prior abortions offering a protective effect against PE with those stating prior abortions are a risk factor for the development of PE. Additionally, this critical review will discuss the advancements and current understanding of reproductive immunology and how it pertains to this association between positive abortion history and PE.
Collapse
Affiliation(s)
- Sarah J Norman
- Medicine, American University of the Caribbean School of Medicine, Cupecoy, SXM
| | - Gena Fontus
- Medicine, American University of the Caribbean School of Medicine, Cupecoy, SXM
| | | | - Tasneem Hiba
- Medicine, American University of the Caribbean School of Medicine, Cupecoy, SXM
| | | | - Michael Osondu
- Medicine, American University of the Caribbean School of Medicine, Cupecoy, SXM
| | - Volha Shylovich
- Obstetrics and Gynecology, BronxCare Health System, New York, USA
| |
Collapse
|
6
|
Peng L, Zhao W, Yin T, Xu C, Wang G, Du M. The unique expression pattern of human leukocyte antigen in trophoblasts potentially explains the key mechanism of maternal-fetal tolerance and successful pregnancy. J Reprod Immunol 2023; 158:103980. [PMID: 37390630 DOI: 10.1016/j.jri.2023.103980] [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: 02/09/2023] [Revised: 05/15/2023] [Accepted: 06/12/2023] [Indexed: 07/02/2023]
Abstract
The success of pregnancy mainly depends on immune tolerance of the mother for the semi-allogeneic fetus. The placenta carrying paternal antigens develops in the maternal uterus without suffering immune attack, making the underlying mechanism of maternal tolerance an enduring mystery. As we all know, human leukocyte antigen (HLA) plays an important role in antigen processing and presentation, thus inducing specific immune responses. Therefore, it is reasonable to speculate that the absence of classical HLA class-I(HLA-I) and HLA class-II (HLA-II) molecules in trophoblasts may account for the maternal-fetal tolerance. Here, we review the HLA-involved interactions between trophoblast cells and decidual immune cells, which contribute to the immunotolerance in the development of normal pregnancy. We also compare the similarity between the maternal-fetal interface and tumor-immune microenvironment because the important role of HLA molecules in tumor immune invasion can provide some references to studies of maternal-fetal immune tolerance. Besides, the abnormal HLA expression is likely to be associated with unexplained miscarriage, making HLA molecules potential therapeutic targets. The advances reported by these studies may exert profound influences on other research areas, including tumor immunity, organ transplantation and autoimmune disease in the future.
Collapse
Affiliation(s)
- Lijin Peng
- The Lab of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Weijie Zhao
- The Lab of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Tingxuan Yin
- The Lab of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Chunfang Xu
- The Lab of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Guangchuan Wang
- Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Meirong Du
- The Lab of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China.
| |
Collapse
|
7
|
Zhang Y, Liu Z, Sun H. Fetal-maternal interactions during pregnancy: a 'three-in-one' perspective. Front Immunol 2023; 14:1198430. [PMID: 37350956 PMCID: PMC10282753 DOI: 10.3389/fimmu.2023.1198430] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 05/24/2023] [Indexed: 06/24/2023] Open
Abstract
A successful human pregnancy requires the maternal immune system to recognize and tolerate the semi-allogeneic fetus, allowing for appropriate trophoblasts invasion and protecting the fetus from invading pathogens. Therefore, maternal immunity is critical for the establishment and maintenance of pregnancy, especially at the maternal-fetal interface. Anatomically, the maternal-fetal interface has both maternally- and fetally- derived cells, including fetal originated trophoblasts and maternal derived immune cells and stromal cells. Besides, a commensal microbiota in the uterus was supposed to aid the unique immunity in pregnancy. The appropriate crosstalk between fetal derived and maternal originated cells and uterine microbiota are critical for normal pregnancy. Dysfunctional maternal-fetal interactions might be associated with the development of pregnancy complications. This review elaborates the latest knowledge on the interactions between trophoblasts and decidual immune cells, highlighting their critical roles in maternal-fetal tolerance and pregnancy development. We also characterize the role of commensal bacteria in promoting pregnancy progression. Furthermore, this review may provide new thought on future basic research and the development of clinical applications for pregnancy complications.
Collapse
Affiliation(s)
- Yonghong Zhang
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Zhaozhao Liu
- Reproduction Center, The Third Affiliated Hospital of ZhengZhou University, ZhengZhou, China
| | - Haixiang Sun
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
8
|
Schulz K, Trendelenburg M. C1q as a target molecule to treat human disease: What do mouse studies teach us? Front Immunol 2022; 13:958273. [PMID: 35990646 PMCID: PMC9385197 DOI: 10.3389/fimmu.2022.958273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 06/24/2022] [Indexed: 11/17/2022] Open
Abstract
The complement system is a field of growing interest for pharmacological intervention. Complement protein C1q, the pattern recognition molecule at the start of the classical pathway of the complement cascade, is a versatile molecule with additional non-canonical actions affecting numerous cellular processes. Based on observations made in patients with hereditary C1q deficiency, C1q is protective against systemic autoimmunity and bacterial infections. Accordingly, C1q deficient mice reproduce this phenotype with susceptibility to autoimmunity and infections. At the same time, beneficial effects of C1q deficiency on disease entities such as neurodegenerative diseases have also been described in murine disease models. This systematic review provides an overview of all currently available literature on the C1q knockout mouse in disease models to identify potential target diseases for treatment strategies focusing on C1q, and discusses potential side-effects when depleting and/or inhibiting C1q.
Collapse
Affiliation(s)
- Kristina Schulz
- Laboratory of Clinical Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
- Division of Internal Medicine, University Hospital Basel, Basel, Switzerland
- *Correspondence: Kristina Schulz,
| | - Marten Trendelenburg
- Laboratory of Clinical Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
- Division of Internal Medicine, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
9
|
Composition and effects of seminal plasma in the female reproductive tracts on implantation of human embryos. Biomed Pharmacother 2022; 151:113065. [PMID: 35550527 DOI: 10.1016/j.biopha.2022.113065] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/21/2022] Open
Abstract
The function of seminal plasma involves acting as a transport medium for sperm and as a means of communication between the reproductive tissues of the male and female. It is also a vital factor to prime the reproductive tracts of the female for optimal pregnancy. When the reproductive tract of the female is exposed to seminal plasma, serious alterations take place, enhancing pathogen and debris clearance observed in the uterus throughout mating. It is also capable of supporting embryo growth, promoting the receptivity of the uterus, and establishing tolerance to the semi-allogenic embryo. Moreover, seminal plasma is capable of regulating the functions of several female reproductive organs and providing an ideal condition for effective embryo implantation and pregnancy. It is believed that the health state of the offspring is affected by exposure to seminal plasma. For the treatment of infertility, assisted reproductive technologies have been extensively employed. The application of seminal plasma as a therapeutic approach to enhance the development of embryo competency and rate of implantation, receptivity of endometrium, and establishment of maternal immune tolerance in cycles of ART appears possible. Herein, current knowledge on the composition of seminal plasma and the physiological roles it possesses on various parts of the female reproductive tract are summarized. Moreover, the role of seminal plasma in the development of embryos, implantation, and the following fetal growth and survival have been reviewed in this article.
Collapse
|
10
|
Robertson SA, Moldenhauer LM, Green ES, Care AS, Hull ML. Immune determinants of endometrial receptivity: a biological perspective. Fertil Steril 2022; 117:1107-1120. [PMID: 35618356 DOI: 10.1016/j.fertnstert.2022.04.023] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/19/2022] [Accepted: 04/22/2022] [Indexed: 11/04/2022]
Abstract
Immune cells are essential for endometrial receptivity to embryo implantation and early placental development. They exert tissue-remodeling and immune regulatory roles-acting to promote epithelial attachment competence, regulate the differentiation of decidual cells, remodel the uterine vasculature, control and resolve inflammatory activation, and suppress destructive immunity to paternally inherited alloantigens. From a biological perspective, the endometrial immune response exerts a form of "quality control"-it promotes implantation success when conditions are favorable but constrains receptivity when physiological circumstances are not ideal. Women with recurrent implantation failure and recurrent miscarriage may exhibit altered numbers or disturbed function of certain uterine immune cell populations-most notably uterine natural killer cells and regulatory T cells. Preclinical and animal studies indicate that deficiencies or aberrant activation states in these cells can be causal in the pathophysiological mechanisms of infertility. Immune cells are, therefore, targets for diagnostic evaluation and therapeutic intervention. However, current diagnostic tests are overly simplistic and have limited clinical utility. To be more informative, they need to account for the full complexity and reflect the range of perturbations that can occur in uterine immune cell phenotypes and networks. Moreover, safe and effective interventions to modulate these cells are in their infancy, and personalized approaches matched to specific diagnostic criteria will be needed. Here we summarize current biological understanding and identify knowledge gaps to be resolved before the promise of therapies to target the uterine immune response can be fully realized.
Collapse
Affiliation(s)
- Sarah A Robertson
- Robinson Research Institute and School of Biomedicine, The University of Adelaide, Adelaide, South Australia, Australia.
| | - Lachlan M Moldenhauer
- Robinson Research Institute and School of Biomedicine, The University of Adelaide, Adelaide, South Australia, Australia
| | - Ella S Green
- Robinson Research Institute and School of Biomedicine, The University of Adelaide, Adelaide, South Australia, Australia
| | - Alison S Care
- Robinson Research Institute and School of Biomedicine, The University of Adelaide, Adelaide, South Australia, Australia
| | - M Louise Hull
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
11
|
Dang Y, Souchet C, Moresi F, Jeljeli M, Raquillet B, Nicco C, Chouzenoux S, Lagoutte I, Marcellin L, Batteux F, Doridot L. BCG-trained innate immunity leads to fetal growth restriction by altering immune cell profile in the mouse developing placenta. J Leukoc Biol 2021; 111:1009-1020. [PMID: 34533228 DOI: 10.1002/jlb.4a0720-458rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Trained immunity is a new concept illustrating that innate immune cells are able to undergo a long-term metabolic and epigenetic reprogramming after infection or vaccination, thus displaying either a pro- or an anti-inflammatory phenotype during a sequential unrelated challenge. Innate immune cells such as natural killer (NK) cells and macrophages constitute a large part of the decidual leukocyte population at the maternal-fetal interface, playing an important role in placental development and as such in fetal growth and development. In this study, we hypothesized that training the innate immune cells before pregnancy could have an impact on pregnancy. To test this hypothesis, we used CBA/J x DBA/2 mouse model to investigate pregnancy outcomes and leukocyte population at the maternal-fetal interface. Although we were not able to show a beneficial effect of LPS-tolerogenic training on fetal resorption, Bacillus Calmette-Guérin (BCG) training, known to prime innate immune cells to be proinflammatory, led to fetal growth restriction, without aggravating the fetal resorption rate. We also found that BCG training led to less NK cells and macrophages at the maternal-fetal interface at the early stage of placentation (E9.5), associated with a down-regulation of Ccr3 and Lif mRNA expression. This induced altered leucocyte population profile can be an explanation for the subsequent fetal growth restriction. These data suggest that preconceptional infections-induced trained immunity could influence pregnancy outcomes.
Collapse
Affiliation(s)
- Yipu Dang
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 PARIS, France, Paris, France
| | - Camille Souchet
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 PARIS, France, Paris, France
| | - Fabiana Moresi
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 PARIS, France, Paris, France
| | - Mohamed Jeljeli
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 PARIS, France, Paris, France.,Service d'immunologie Biologique, AP-HP, Hôpital Universitaire Paris Centre, F-75014 Paris, France, Paris, France
| | - Bruno Raquillet
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 PARIS, France, Paris, France
| | - Carole Nicco
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 PARIS, France, Paris, France
| | - Sandrine Chouzenoux
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 PARIS, France, Paris, France
| | - Isabelle Lagoutte
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 PARIS, France, Paris, France
| | - Louis Marcellin
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 PARIS, France, Paris, France.,Département de Gynécologie Obstétrique II et Médecine de la Reproduction, AP-HP, Hôpital Universitaire Paris Centre, F-75014 Paris, France, Paris, France
| | - Frederic Batteux
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 PARIS, France, Paris, France.,Service d'immunologie Biologique, AP-HP, Hôpital Universitaire Paris Centre, F-75014 Paris, France, Paris, France
| | - Ludivine Doridot
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 PARIS, France, Paris, France
| |
Collapse
|
12
|
Toll-like receptor-4 null mutation causes fetal loss and fetal growth restriction associated with impaired maternal immune tolerance in mice. Sci Rep 2021; 11:16569. [PMID: 34400677 PMCID: PMC8368181 DOI: 10.1038/s41598-021-95213-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 07/16/2021] [Indexed: 12/30/2022] Open
Abstract
Maternal immune adaptation to accommodate pregnancy depends on sufficient availability of regulatory T (Treg) cells to enable embryo implantation. Toll-like receptor 4 is implicated as a key upstream driver of a controlled inflammatory response, elicited by signals in male partner seminal fluid, to initiate expansion of the maternal Treg cell pool after mating. Here, we report that mice with null mutation in Tlr4 (Tlr4−/−) exhibit impaired reproductive outcomes after allogeneic mating, with reduced pregnancy rate, elevated mid-gestation fetal loss, and fetal growth restriction, compared to Tlr4+/+ wild-type controls. To investigate the effects of TLR4 deficiency on early events of maternal immune adaptation, TLR4-regulated cytokines and immune regulatory microRNAs were measured in the uterus at 8 h post-mating by qPCR, and Treg cells in uterus-draining lymph nodes were evaluated by flow cytometry on day 3.5 post-coitum. Ptgs2 encoding prostaglandin-endoperoxide synthase 2, cytokines Csf2, Il6, Lif, and Tnf, chemokines Ccl2, Cxcl1, Cxcl2, and Cxcl10, and microRNAs miR-155, miR-146a, and miR-223 were induced by mating in wild-type mice, but not, or to a lesser extent, in Tlr4−/− mice. CD4+ T cells were expanded after mating in Tlr4+/+ but not Tlr4−/− mice, with failure to expand peripheral CD25+FOXP3+ NRP1− or thymic CD25+FOXP3+ NRP1+ Treg cell populations, and fewer Treg cells expressed Ki67 proliferation marker and suppressive function marker CTLA4. We conclude that TLR4 is an essential mediator of the inflammation-like response in the pre-implantation uterus that induces generation of Treg cells to support robust pregnancy tolerance and ensure optimal fetal growth and survival.
Collapse
|
13
|
Gillis-Buck E, Miller H, Sirota M, Sanders SJ, Ntranos V, Anderson MS, Gardner JM, MacKenzie TC. Extrathymic Aire-expressing cells support maternal-fetal tolerance. Sci Immunol 2021; 6:eabf1968. [PMID: 34272228 PMCID: PMC9363019 DOI: 10.1126/sciimmunol.abf1968] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 06/17/2021] [Indexed: 12/21/2022]
Abstract
Healthy pregnancy requires tolerance to fetal alloantigens as well as syngeneic embryonic and placental antigens. Given the importance of the autoimmune regulator (Aire) gene in self-tolerance, we investigated the role of Aire-expressing cells in maternal-fetal tolerance. We report that maternal ablation of Aire-expressing (Aire +) cells during early mouse pregnancy caused intrauterine growth restriction (IUGR) in both allogeneic and syngeneic pregnancies. This phenotype is immune mediated, as IUGR was rescued in Rag1-deficient mice, and involved a memory response, demonstrated by recurrence of severe IUGR in second pregnancies. Single-cell RNA sequencing demonstrated that Aire + cell depletion in pregnancy results in expansion of activated T cells, particularly T follicular helper cells. Unexpectedly, selective ablation of either Aire-expressing medullary thymic epithelial cells or extrathymic Aire-expressing cells (eTACs) mapped the IUGR phenotype exclusively to eTACs. Thus, we report a previously undescribed mechanism for the maintenance of maternal-fetal immune homeostasis and demonstrate that eTACs protect the conceptus from immune-mediated IUGR.
Collapse
Affiliation(s)
- Eva Gillis-Buck
- Department of Surgery, University of California, San Francisco, CA, USA
| | - Haleigh Miller
- Department of Epidemiology and Biostatistics University of California, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA, USA
- Diabetes Center University of California, San Francisco, CA, USA
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA, USA
- Department of Pediatrics University of California, San Francisco, CA, USA
| | - Stephan J Sanders
- Department of Psychiatry and Behavioral Sciences, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
- Center for Maternal-Fetal Precision Medicine, University of California, San Francisco, CA, USA
| | - Vasilis Ntranos
- Department of Epidemiology and Biostatistics University of California, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA, USA
- Diabetes Center University of California, San Francisco, CA, USA
| | - Mark S Anderson
- Diabetes Center University of California, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
| | - James M Gardner
- Department of Surgery, University of California, San Francisco, CA, USA.
- Diabetes Center University of California, San Francisco, CA, USA
| | - Tippi C MacKenzie
- Department of Surgery, University of California, San Francisco, CA, USA.
- Department of Pediatrics University of California, San Francisco, CA, USA
- Center for Maternal-Fetal Precision Medicine, University of California, San Francisco, CA, USA
| |
Collapse
|
14
|
Kieffer TE, Chin PY, Green ES, Moldenhauer LM, Prins JR, Robertson SA. Prednisolone in early pregnancy inhibits regulatory T cell generation and alters fetal and placental development in mice. Mol Hum Reprod 2021; 26:340-352. [PMID: 32159777 DOI: 10.1093/molehr/gaaa019] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 02/16/2020] [Indexed: 01/01/2023] Open
Abstract
Corticosteroids have been utilised in the assisted reproduction setting with the expectation of suppressing aberrant immune activation and improving fertility in women. However, the effects of corticosteroids on fertility, and on pregnancy and offspring outcomes, are unclear. In this study, mice were administered prednisolone (1 mg/kg) or PBS daily in the pre-implantation phase, and effects on the adaptive immune response, the implantation rate, fetal development and postnatal outcomes were investigated. Prednisolone disrupted the expected expansion of CD4+ T cells in early pregnancy, inhibiting generation of both regulatory T cells (Treg cells) and effector T cells and suppressing IFNG required for T cell functional competence. Prednisolone caused an 8-20% increase in the embryo implantation rate and increased the number of viable pups per litter. In late gestation, fetal and placental weights were reduced in a litter size-dependent manner, and the canonical inverse relationship between litter size and fetal weight was lost. The duration of pregnancy was extended by ~ 0.5 day and birth weight was reduced by ~ 5% after prednisolone treatment. Viability of prednisolone-exposed offspring was comparable to controls, but body weight was altered in adulthood, particularly in male offspring. Thus, while prednisolone given in the pre-implantation phase in mice increases maternal receptivity to implantation and resource investment in fetal growth, there is a trade-off in long-term consequences for fetal development, birth weight and offspring health. These effects are associated with, and likely caused by, prednisolone suppression of the adaptive immune response at the outset of pregnancy.
Collapse
Affiliation(s)
- Tom Ec Kieffer
- Robinson Research Institute & Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia.,Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Peck Y Chin
- Robinson Research Institute & Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Ella S Green
- Robinson Research Institute & Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Lachlan M Moldenhauer
- Robinson Research Institute & Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| | - Jelmer R Prins
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Sarah A Robertson
- Robinson Research Institute & Adelaide Medical School, University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
15
|
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: 18] [Impact Index Per Article: 6.0] [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
|
16
|
Marsico TV, Caetano DP, Rodrigues R, Valente RS, Fontes PK, Mesquita FS, Andrade SCDS, Basso AC, Nogueira MFG, Sudano MJ. Transcriptional profiling of embryo cryotolerance. Mol Reprod Dev 2020; 87:1245-1259. [PMID: 33156526 DOI: 10.1002/mrd.23436] [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: 09/28/2020] [Accepted: 10/01/2020] [Indexed: 01/04/2023]
Abstract
The cryosurvival of embryos is a complex process involving dynamic and integrated morphological, functional, and molecular changes. Here, we evaluated the transcriptional profiling of bovine embryos possessing high and low cryotolerance (HC and LC, respectively) by assessing the resumption of development. Embryos were produced in vitro (N = 1137) and cryopreserved (N = 894). Blastocysts samples possessed pronounced group individualization at RNA sequencing. A total of 114 genes were differentially expressed, and 27 and 84 genes were upregulated in HC and LC, respectively. Among the over-represented biological functions, cellular growth and proliferation, cell death and survival, and organismal survival were predicted to be activated, while cellular movement and cell-to-cell signaling were predicted to be inhibited in HC embryos. Enriched canonical pathways and upstream regulators related to cellular proliferation and survival (HC), inflammatory processes, and cell death (LC) were predicted to represent two embryonic molecular profiles present during the resumption of development after cryopreservation. The marked contrast in transcriptional profiles between HC and LC strongly suggests the influence of embryonic competence after cryopreservation on its respective transcriptome and indicated that HC and LC presented two different molecular strategies to overcome cryopreservation-related stress and resume postcryopreservation development.
Collapse
Affiliation(s)
- Thamiris V Marsico
- Center for Natural and Human Sciences, Federal University of ABC, Santo André, São Paulo, Brazil
| | - Diana P Caetano
- School of Veterinary Medicine, Federal University of Pampa, Uruguaiana, Rio Grande do Sul, Brazil
| | | | - Roniele S Valente
- Center for Natural and Human Sciences, Federal University of ABC, Santo André, São Paulo, Brazil
| | - Patricia K Fontes
- Department of Pharmacology, Laboratory of Phytomedicines, Pharmacology and Biotechnology, Institute of Biosciences, University of São Paulo State (Unesp), Botucatu, São Paulo, Brazil
| | - Fernando S Mesquita
- Center for Natural and Human Sciences, Federal University of ABC, Santo André, São Paulo, Brazil
| | - Sónia C da Silva Andrade
- Department of Genetics and Evolutionary Biology, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Andréa C Basso
- In vitro Brazil - IVB, ABS Pecplan, Mogi Mirim, São Paulo, Brazil
| | - Marcelo F G Nogueira
- Department of Pharmacology, Laboratory of Phytomedicines, Pharmacology and Biotechnology, Institute of Biosciences, University of São Paulo State (Unesp), Botucatu, São Paulo, Brazil.,Department of Biological Science, School of Sciences and Languages, University of São Paulo State, Assis, São Paulo, Brazil
| | - Mateus J Sudano
- Center for Natural and Human Sciences, Federal University of ABC, Santo André, São Paulo, Brazil.,School of Veterinary Medicine, Federal University of Pampa, Uruguaiana, Rio Grande do Sul, Brazil.,Department of Genetics and Evolution, Federal University of São Carlos, São Carlos, São Paulo, Brazil
| |
Collapse
|
17
|
Abstract
Seminal fluid is often assumed to have just one function in mammalian reproduction, delivering sperm to fertilize oocytes. But seminal fluid also transmits signaling agents that interact with female reproductive tissues to facilitate conception and .pregnancy. Upon seminal fluid contact, female tissues initiate a controlled inflammatory response that affects several aspects of reproductive function to ultimately maximize the chances of a male producing healthy offspring. This effect is best characterized in mice, where the female response involves several steps. Initially, seminal fluid factors cause leukocytes to infiltrate the female reproductive tract, and to selectively target and eliminate excess sperm. Other signals stimulate ovulation, induce an altered transcriptional program in female tract tissues that modulates embryo developmental programming, and initiate immune adaptations to promote receptivity to implantation and placental development. A key result is expansion of the pool of regulatory T cells that assist implantation by suppressing inflammation, mediating tolerance to male transplantation antigens, and promoting uterine vascular adaptation and placental development. Principal signaling agents in seminal fluid include prostaglandins and transforming growth factor-β. The balance of male signals affects the nature of the female response, providing a mechanism of ‟cryptic female choiceˮ that influences female reproductive investment. Male-female seminal fluid signaling is evident in all mammalian species investigated including human, and effects of seminal fluid in invertebrates indicate evolutionarily conserved mechanisms. Understanding the female response to seminal fluid will shed new light on infertility and pregnancy disorders and is critical to defining how events at conception influence offspring health.
Collapse
Affiliation(s)
- John E Schjenken
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Sarah A Robertson
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, Australia
| |
Collapse
|
18
|
Li Y, Yu S, Huang C, Lian R, Chen C, Liu S, Li L, Diao L, Markert UR, Zeng Y. Evaluation of peripheral and uterine immune status of chronic endometritis in patients with recurrent reproductive failure. Fertil Steril 2020; 113:187-196.e1. [DOI: 10.1016/j.fertnstert.2019.09.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 08/27/2019] [Accepted: 09/03/2019] [Indexed: 01/12/2023]
|
19
|
Sharkey DJ, Glynn DJ, Schjenken JE, Tremellen KP, Robertson SA. Interferon-gamma inhibits seminal plasma induction of colony-stimulating factor 2 in mouse and human reproductive tract epithelial cells. Biol Reprod 2019; 99:514-526. [PMID: 29596569 DOI: 10.1093/biolre/ioy071] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 03/26/2018] [Indexed: 12/26/2022] Open
Abstract
Seminal fluid interacts with the female reproductive tract to initiate a permissive immune response that facilitates embryo implantation and pregnancy success. The immune-regulatory cytokine interferon-γ (IFNG), which can be elevated in seminal plasma, is associated with reduced fertility. Here, we investigated how IFNG influences the female immune response to seminal fluid. In human Ect1 cervical epithelial cells, IFNG added at physiologically relevant concentrations substantially impaired seminal plasma-induced synthesis of key cytokines colony-stimulating factor 2 (CSF2) and interleukin-6 (IL6). Seminal fluid-induced CSF2 synthesis was also suppressed in the uterus of mice in vivo, when IFNG was delivered transcervically 12 h after mating. Transforming growth factor B1 (TGFB1) is the major seminal fluid signaling factor which elicits CSF2 induction, and IFNG exhibited potent dose-dependent suppression of CSF2 synthesis induced by TGFB1 in murine uterine epithelial cells in vitro. Similarly, IFNG suppressed TGFB1-mediated CSF2 induction in Ect1 cells and human primary cervical epithelial cells; however, IL6 regulation by IFNG was independent of TGFB1. Quantitative PCR confirmed that CSF2 regulation by IFNG in Ect1 cells occurs at the gene transcription level, secondary to IFNG suppression of TGFBR2 encoding TGFB receptor 2. Conversely, TGFB1 suppressed IFNG receptor 1 and 2 genes IFNGR1 and IFNGR2. These data identify IFNG as a potent inhibitor of the TGFB-mediated seminal fluid interaction with relevant reproductive tract epithelia in mice and human. These findings raise the prospect that IFNG in the male partner's seminal fluid impairs immune adaptation for pregnancy following coitus in women.
Collapse
Affiliation(s)
- David J Sharkey
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Danielle J Glynn
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - John E Schjenken
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Kelton P Tremellen
- Repromed Pty Ltd, Dulwich, South Australia, Australia.,School of Pharmacy and Medical Sciences, University of South Australia, South Australia, Australia
| | - Sarah A Robertson
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
20
|
Schumacher A, Sharkey DJ, Robertson SA, Zenclussen AC. Immune Cells at the Fetomaternal Interface: How the Microenvironment Modulates Immune Cells To Foster Fetal Development. THE JOURNAL OF IMMUNOLOGY 2019; 201:325-334. [PMID: 29987001 DOI: 10.4049/jimmunol.1800058] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 04/06/2018] [Indexed: 12/23/2022]
Abstract
Immune cells adapt their phenotypic and functional characteristics in response to the tissue microenvironment within which they traffic and reside. The fetomaternal interface, consisting of placental trophoblasts and the maternal decidua, is a highly specialized tissue with a unique and time-limited function: to nourish and support development of the semiallogeneic fetus and protect it from inflammatory or immune-mediated injury. It is therefore important to understand how immune cells within these tissues are educated and adapt to fulfill their biological functions. This review article focuses on the local regulatory mechanisms ensuring that both innate and adaptive immune cells appropriately support the early events of implantation and placental development through direct involvement in promoting immune tolerance of fetal alloantigens, suppressing inflammation, and remodeling of maternal uterine vessels to facilitate optimal placental function and fetal growth.
Collapse
Affiliation(s)
- Anne Schumacher
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University, Magdeburg 39108, Germany; and
| | - David J Sharkey
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, South Australia 5005, Australia
| | - Sarah A Robertson
- Robinson Research Institute and Adelaide Medical School, The University of Adelaide, South Australia 5005, Australia
| | - Ana C Zenclussen
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University, Magdeburg 39108, Germany; and
| |
Collapse
|
21
|
Robertson SA, Green ES, Care AS, Moldenhauer LM, Prins JR, Hull ML, Barry SC, Dekker G. Therapeutic Potential of Regulatory T Cells in Preeclampsia-Opportunities and Challenges. Front Immunol 2019; 10:478. [PMID: 30984163 PMCID: PMC6448013 DOI: 10.3389/fimmu.2019.00478] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 02/21/2019] [Indexed: 12/26/2022] Open
Abstract
Inflammation is a central feature and is implicated as a causal factor in preeclampsia and other hypertensive disorders of pregnancy. Inflammatory mediators and leukocytes, which are elevated in peripheral blood and gestational tissues, contribute to the uterine vascular anomalies and compromised placental function that characterize particularly the severe, early onset form of disease. Regulatory T (Treg) cells are central mediators of pregnancy tolerance and direct other immune cells to counteract inflammation and promote robust placentation. Treg cells are commonly perturbed in preeclampsia, and there is evidence Treg cell insufficiency predates onset of symptoms. A causal role is implied by mouse studies showing sufficient numbers of functionally competent Treg cells must be present in the uterus from conception, to support maternal vascular adaptation and prevent later placental inflammatory pathology. Treg cells may therefore provide a tractable target for both preventative strategies and treatment interventions in preeclampsia. Steps to boost Treg cell activity require investigation and could be incorporated into pregnancy planning and preconception care. Pharmacological interventions developed to target Treg cells in autoimmune conditions warrant consideration for evaluation, utilizing rigorous clinical trial methodology, and ensuring safety is paramount. Emerging cell therapy tools involving in vitro Treg cell generation and/or expansion may in time become relevant. The success of preventative and therapeutic approaches will depend on resolving several challenges including developing informative diagnostic tests for Treg cell activity applicable before conception or during early pregnancy, selection of relevant patient subgroups, and identification of appropriate windows of gestation for intervention.
Collapse
Affiliation(s)
- Sarah A. Robertson
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Ella S. Green
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Alison S. Care
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Lachlan M. Moldenhauer
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | | | - M. Louise Hull
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- Women's and Children's Hospital, Adelaide, SA, Australia
| | - Simon C. Barry
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Gustaaf Dekker
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
22
|
Bonney EA, Johnson MR. The role of maternal T cell and macrophage activation in preterm birth: Cause or consequence? Placenta 2019; 79:53-61. [PMID: 30929747 DOI: 10.1016/j.placenta.2019.03.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 03/06/2019] [Indexed: 12/18/2022]
Abstract
The role of the immune system in term (TL) and preterm labor (PTL) is unknown. Despite the fact that globally, PTL remains the most important cause of childhood mortality. Infection, typically of the fetal membranes, termed chorioamnionitis, is the best-understood driver of PTL, but the mechanisms underpinning other causes, including idiopathic and stretch-induced PTL, are unclear, but may well involve activation of the maternal immune system. The final common pathway of placental dysfunction, fetal membrane rupture, cervical dilation and uterine contractions are highly complex processes. At term, choriodecidual rather than myometrial inflammation is thought to drive the onset of labor and similar findings are present in different types of PTL including idiopathic PTL. Although accumulated data has confirmed an association between the immune response and preterm birth, there is yet a need to understand if this response is an initiator or a consequence of tissue-level dysregulation. This review focuses on the potential role of macrophages and T cells in innate and adaptive immunity relevant to preterm birth in humans and animal models.
Collapse
Affiliation(s)
- Elizabeth A Bonney
- Department of Obstetrics, Gynecology and Reproductive Sciences University of Vermont, Larner College of Medicine, Burlington, VT, USA.
| | - Mark R Johnson
- Faculty of Medicine, Department of Surgery & Cancer, Imperial College, London, United Kingdom
| |
Collapse
|
23
|
Robertson SA, Care AS, Moldenhauer LM. Regulatory T cells in embryo implantation and the immune response to pregnancy. J Clin Invest 2018; 128:4224-4235. [PMID: 30272581 DOI: 10.1172/jci122182] [Citation(s) in RCA: 253] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
At implantation, the embryo expresses paternally derived alloantigens and evokes inflammation that can threaten reproductive success. To ensure a robust placenta and sustainable pregnancy, an active state of maternal immune tolerance mediated by CD4+ regulatory T cells (Tregs) is essential. Tregs operate to inhibit effector immunity, contain inflammation, and support maternal vascular adaptations, thereby facilitating trophoblast invasion and placental access to the maternal blood supply. Insufficient Treg numbers or inadequate functional competence are implicated in idiopathic infertility and recurrent miscarriage as well as later-onset pregnancy complications stemming from placental insufficiency, including preeclampsia and fetal growth restriction. In this Review, we summarize the mechanisms acting in the conception environment to drive the Treg response and discuss prospects for targeting the T cell compartment to alleviate immune-based reproductive disorders.
Collapse
|
24
|
Brown HM, Green ES, Tan TCY, Gonzalez MB, Rumbold AR, Hull ML, Norman RJ, Packer NH, Robertson SA, Thompson JG. Periconception onset diabetes is associated with embryopathy and fetal growth retardation, reproductive tract hyperglycosylation and impaired immune adaptation to pregnancy. Sci Rep 2018; 8:2114. [PMID: 29391475 PMCID: PMC5794861 DOI: 10.1038/s41598-018-19263-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 12/22/2017] [Indexed: 01/04/2023] Open
Abstract
Diabetes has been linked with impaired fertility but the underlying mechanisms are not well defined. Here we use a streptozotocin-induced diabetes mouse model to investigate the cellular and biochemical changes in conceptus and maternal tissues that accompany hyperglycaemia. We report that streptozotocin treatment before conception induces profound intra-cellular protein β-O-glycosylation (O-GlcNAc) in the oviduct and uterine epithelium, prominent in early pregnancy. Diabetic mice have impaired blastocyst development and reduced embryo implantation rates, and delayed mid-gestation growth and development. Peri-conception changes are accompanied by increased expression of pro-inflammatory cytokine Trail, and a trend towards increased Il1a, Tnf and Ifng in the uterus, and changes in local T-cell dynamics that skew the adaptive immune response to pregnancy, resulting in 60% fewer anti-inflammatory regulatory T-cells within the uterus-draining lymph nodes. Activation of the heat shock chaperones, a mechanism for stress deflection, was evident in the reproductive tract. Additionally, we show that the embryo exhibits elevated hyper-O-GlcNAcylation of both cytoplasmic and nuclear proteins, associated with activation of DNA damage (ɣH2AX) pathways. These results advance understanding of the impact of peri-conception diabetes, and provide a foundation for designing interventions to support healthy conception without propagation of disease legacy to offspring.
Collapse
Affiliation(s)
- Hannah M Brown
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, Australia. .,Australian Research Council (ARC) Centre for Nanoscale Biophotonics, University of Adelaide, Adelaide, Australia.
| | - Ella S Green
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Tiffany C Y Tan
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Macarena B Gonzalez
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Alice R Rumbold
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - M Louise Hull
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, Australia.,Fertility SA, Adelaide, Australia
| | - Robert J Norman
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, Australia.,Fertility SA, Adelaide, Australia
| | - Nicolle H Packer
- ARC Centre for Nanoscale Biophotonics, Department of Molecular Sciences, Macquarie University, Sydney, Australia
| | - Sarah A Robertson
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Jeremy G Thompson
- Robinson Research Institute, Adelaide Medical School, University of Adelaide, Adelaide, Australia.,Australian Research Council (ARC) Centre for Nanoscale Biophotonics, University of Adelaide, Adelaide, Australia
| |
Collapse
|