1
|
He L, Zheng S, Zhan F, Lin N. The role of necroptosis in pathological pregnancies: Mechanisms and therapeutic opportunities. J Reprod Immunol 2025; 169:104460. [PMID: 40023097 DOI: 10.1016/j.jri.2025.104460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 02/02/2025] [Accepted: 02/19/2025] [Indexed: 03/04/2025]
Abstract
Necroptosis, a distinctive form of programmed cell death differs mechanistically from apoptosis pyroptosis, and autophagy, is characterized by the activation of receptor-interacting protein kinases (RIPK1/RIPK3) and their downstream effector, mixed lineage kinase domain-like protein (MLKL). This programmed cell death pathway serves as a crucial mediator of inflammatory responses and has been implicated in the pathogenesis of diverse pathological conditions. Recent evidence has implicated dysregulated necroptosis in the pathogenesis of severe pregnancy complications, including preeclampsia (PE), fetal growth restriction (FGR), recurrent spontaneous abortion (RSA), and gestational diabetes mellitus (GDM). In these disorders, necroptosis promotes placental dysfunction through multiple interconnected mechanisms: amplification of pro-inflammatory cytokine cascades, aberrant immune activation, disruption of plasma membrane integrity, and subsequent tissue injury.These pregnancy-related pathologies consistently demonstrate elevated necroptotic signatures, correlating with adverse maternal-fetal outcomes. This comprehensive review synthesizes current understanding of the molecular mechanisms underlying necroptosis, with particular emphasis on its pivotal role in the etiopathogenesis of pregnancy-related disorders. Furthermore, we critically evaluate the therapeutic potential of targeting the necroptotic signaling axis, providing novel perspectives for developing targeted interventions to improve clinical outcomes in complicated pregnancies.
Collapse
Affiliation(s)
- Lidan He
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350004, China; College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350122, China.
| | - Shan Zheng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350004, China; College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350122, China
| | - Feng Zhan
- College of Engineering, Fujian Jiangxia University, Fuzhou 350108, China; School of Electronic Information Engineering, Taiyuan University of Science and Technology, Taiyuan, Shanxi 030024, China
| | - Na Lin
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350122, China; Medical Genetic Diagnosis and Therapy Center, Fujian Maternity and Child Health Hospital, Fuzhou 350122, China.
| |
Collapse
|
2
|
Wang Y, Guo A, Yang L, Han X, Li Q, Liu J, Han Y, Yang Y, Chao L. Immune dysregulation of decidual NK cells mediated by GRIM19 downregulation contributes to the occurrence of recurrent pregnancy loss. Mol Cell Biochem 2025; 480:3117-3131. [PMID: 39663335 DOI: 10.1007/s11010-024-05181-z] [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: 07/18/2024] [Accepted: 11/29/2024] [Indexed: 12/13/2024]
Abstract
In patients with recurrent pregnancy loss (RPL), excessive activation of decidual natural killer (dNK) cells has been widely observed, yet the precise underlying mechanisms remain to be elucidated. We collected decidual specimens from RPL patients and controls to assess GRIM19 expression, activation phenotype, cytotoxic function, inflammatory cytokine secretion, and mitochondrial homeostasis in dNK cells. Furthermore, we established a GRIM19-knockout NK-92MI cell line and a GRIM19 ± C57BL/6J mouse model to investigate the relationship between GRIM19 downregulation and dNK immune dysregulation, ultimately contributing to pregnancy loss. Decidual NK cells from RPL patients exhibited significantly lower GRIM19 expression, accompanied by abnormal hyperactivation, enhanced cytotoxicity, and abnormal mitochondrial activation. In vitro experiments confirmed that reduced GRIM19 expression significantly potentiated the cytotoxicity and pro-inflammatory cytokine secretion of NK-92MI cells, while also promoting mitochondrial homeostasis imbalance. Mouse model studies corroborated that GRIM19 downregulation triggers NK cell homeostasis imbalance, contributing to the occurrence of pregnancy loss. Downregulation of GRIM19 in dNK cells contributes to RPL through hyperactivation and disruption of mitochondrial homeostasis, emphasizing its potential as a diagnostic and therapeutic target.
Collapse
MESH Headings
- Female
- Killer Cells, Natural/immunology
- Killer Cells, Natural/pathology
- Killer Cells, Natural/metabolism
- Animals
- Humans
- Mice
- Pregnancy
- Down-Regulation
- Abortion, Habitual/immunology
- Abortion, Habitual/pathology
- Abortion, Habitual/metabolism
- Abortion, Habitual/genetics
- Decidua/immunology
- Decidua/pathology
- Decidua/metabolism
- Mice, Inbred C57BL
- Apoptosis Regulatory Proteins/immunology
- Apoptosis Regulatory Proteins/genetics
- Apoptosis Regulatory Proteins/metabolism
- Adult
- Mice, Knockout
- Mitochondria/metabolism
- Mitochondria/immunology
- NADH, NADPH Oxidoreductases
Collapse
Affiliation(s)
- Ying Wang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Qilu Hospital of Shandong University, No.44 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Anliang Guo
- Shandong University, Jinan, 250012, Shandong, China
| | - Lin Yang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Qilu Hospital of Shandong University, No.44 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Xiaojuan Han
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Qilu Hospital of Shandong University, No.44 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Qianni Li
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Qilu Hospital of Shandong University, No.44 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Jin Liu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Qilu Hospital of Shandong University, No.44 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Yilong Han
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Qilu Hospital of Shandong University, No.44 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Yang Yang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Qilu Hospital of Shandong University, No.44 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Lan Chao
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Qilu Hospital of Shandong University, No.44 Wenhua Xi Road, Jinan, 250012, Shandong, China.
| |
Collapse
|
3
|
Fan J, Zhang J, Bi X, Zhu P, Wang J, Su D, Shi W, Liu Y, Liu H, Wu X. Upregulation of OTULIN Alleviated Recurrent Pregnancy Loss by Suppressing Trophoblast Dysfunction and NF-κB Signaling Pathway. Mol Reprod Dev 2025; 92:e70029. [PMID: 40372975 DOI: 10.1002/mrd.70029] [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: 07/12/2024] [Revised: 04/13/2025] [Accepted: 04/15/2025] [Indexed: 05/17/2025]
Abstract
Recurrent pregnancy loss (RPL) is one of obstetrical diseases with no effective therapy methods. Trophoblast cell dysfunction and inflammation induce embryo implantation insufficiency, thereby resulting in RPL. OTU deubiquitinase with linear linkage specificity (OTULIN) plays a role in regulating the immune response and cell death. However, the role of OTULIN in RPL remains unclear. Spontaneous abortion mouse model and lipopolysaccharide-treated HTR-8/SVneo cells were used to investigate the role of OTULIN in RPL. OTULIN expression was downregulated in the labyrinth trophoblast of RPL mice and LPS-treated trophoblast cells. The embryonic reabsorption rate was decreased in OTULIN-overexpressed spontaneous abortion mice, accompanied with the increase in placental/fetus weight ratio. OTULIN overexpression significantly inhibited apoptosis in vivo and in vitro, as evidenced by the decrease in the activity of caspase 3. The expression of pro-inflammatory cytokines was decreased with OTULIN overexpression. Moreover, OTULIN overexpression decreased p-IκBα/IκBα and p-p65/p65 ratio. The nuclear translocation of NF-κB was suppressed via OTULIN overexpression both in vivo and in vitro. Our study suggested that OTULIN deficiency might cause inflammation and trophoblast abnormalities in RPL. The supplementation with OTULIN might alleviate the development of RPL via inhibiting NF-κB mediated inflammation response.
Collapse
Affiliation(s)
- Junmei Fan
- Reproductive Medicine Center, Shanxi Children's Hospital, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
- Reproductive Medicine Center, The affiliated Children's Hospital of Shanxi Medical University, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
| | - Junkun Zhang
- Department of Intensive Care Unit, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, China
| | - Xingyu Bi
- Reproductive Medicine Center, Shanxi Children's Hospital, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
- Reproductive Medicine Center, The affiliated Children's Hospital of Shanxi Medical University, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
| | - Pengfei Zhu
- Reproductive Medicine Center, Shanxi Children's Hospital, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
- Reproductive Medicine Center, The affiliated Children's Hospital of Shanxi Medical University, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
| | - Jinbao Wang
- Reproductive Medicine Center, Shanxi Children's Hospital, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
- Reproductive Medicine Center, The affiliated Children's Hospital of Shanxi Medical University, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
| | - Dan Su
- Reproductive Medicine Center, Shanxi Children's Hospital, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
- Reproductive Medicine Center, The affiliated Children's Hospital of Shanxi Medical University, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
| | - Wenjing Shi
- Reproductive Medicine Center, Shanxi Children's Hospital, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
- Reproductive Medicine Center, The affiliated Children's Hospital of Shanxi Medical University, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
| | - Yanling Liu
- Reproductive Medicine Center, Shanxi Children's Hospital, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
- Reproductive Medicine Center, The affiliated Children's Hospital of Shanxi Medical University, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
| | - Huiping Liu
- Reproductive Medicine Center, Shanxi Children's Hospital, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
- Reproductive Medicine Center, The affiliated Children's Hospital of Shanxi Medical University, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
| | - Xueqing Wu
- Reproductive Medicine Center, Shanxi Children's Hospital, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
- Reproductive Medicine Center, The affiliated Children's Hospital of Shanxi Medical University, Shanxi Women and Children Health Hospital, Taiyuan, Shanxi, China
| |
Collapse
|
4
|
Alhomieed MF, Al Hartany LO, Alghorab MA, Alsharif A, Kaleemullah A, Wasaya HI, Alsubaie KA, Al Jehani AN, Kayali AM, AlBasri S. The Effect of Pregnancy on Dermatological Disorders: A Systematic Review. Clin Pract 2025; 15:68. [PMID: 40310306 PMCID: PMC12026094 DOI: 10.3390/clinpract15040068] [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: 02/17/2025] [Revised: 03/02/2025] [Accepted: 03/17/2025] [Indexed: 05/02/2025] Open
Abstract
Background: Pregnancy induces hormonal, immunologic, and vascular changes that profoundly affect dermatologic health. This systematic review aimed to assess the impact of pregnancy on dermatological disorders in terms of disease incidence, severity, maternal-fetal outcomes, and optimal management strategies. Methods: A systematic search was performed in PubMed, MEDLINE, and Web of Science databases, following PRISMA guidelines. Studies evaluating pregnant women with dermatological disorders, pregnancy-related dermatoses, and pre-existing morbidities, were included. The collaboratively extracted data included patient demographics, disease severity, treatment approaches, and pregnancy outcomes. Results: A total of 8490 pregnant cases with dermatologic changes and conditions caused by pregnancy were studied. The dermatological conditions were divided into physiological changes, pregnancy-related exacerbation of pre-existing skin conditions, and pregnancy-specific dermatoses. Intrahepatic cholestasis of pregnancy and pemphigoid gestationis were associated with increased rates of adverse fetal outcomes in patients with specific dermatoses, including increased preterm birth and fetal distress rates. The atopic eruption of pregnancy and polymorphic eruption of pregnancy were highly relevant, but their effect on fetal health was minimal. The efficacy and safety of treatment modalities, including corticosteroids, antihistamines, and ursodeoxycholic acid, were variable. Conclusions: Pregnancy drastically affects dermatological health, but the nature of the impact depends on the condition. Optimal maternal and fetal outcomes rely on early diagnosis and individualized management strategies. More randomized controlled trials are required to develop standardized diagnostic and treatment guidelines to enhance the quality of dermatologic care during pregnancy.
Collapse
Affiliation(s)
- Maya Faissal Alhomieed
- Department of Medicine and Surgery, Batterjee Medical College, Jeddah 21442, Saudi Arabia; (M.F.A.); (L.O.A.H.); (M.A.A.); (A.K.); (H.I.W.); (K.A.A.); (A.N.A.J.); (A.M.K.)
| | - Lara Osama Al Hartany
- Department of Medicine and Surgery, Batterjee Medical College, Jeddah 21442, Saudi Arabia; (M.F.A.); (L.O.A.H.); (M.A.A.); (A.K.); (H.I.W.); (K.A.A.); (A.N.A.J.); (A.M.K.)
| | - Marya Aref Alghorab
- Department of Medicine and Surgery, Batterjee Medical College, Jeddah 21442, Saudi Arabia; (M.F.A.); (L.O.A.H.); (M.A.A.); (A.K.); (H.I.W.); (K.A.A.); (A.N.A.J.); (A.M.K.)
| | - Arwa Alsharif
- Department of Medicine and Surgery, Batterjee Medical College, Jeddah 21442, Saudi Arabia; (M.F.A.); (L.O.A.H.); (M.A.A.); (A.K.); (H.I.W.); (K.A.A.); (A.N.A.J.); (A.M.K.)
| | - Ahlam Kaleemullah
- Department of Medicine and Surgery, Batterjee Medical College, Jeddah 21442, Saudi Arabia; (M.F.A.); (L.O.A.H.); (M.A.A.); (A.K.); (H.I.W.); (K.A.A.); (A.N.A.J.); (A.M.K.)
| | - Hanan Ismail Wasaya
- Department of Medicine and Surgery, Batterjee Medical College, Jeddah 21442, Saudi Arabia; (M.F.A.); (L.O.A.H.); (M.A.A.); (A.K.); (H.I.W.); (K.A.A.); (A.N.A.J.); (A.M.K.)
| | - Khlood Abdulaziz Alsubaie
- Department of Medicine and Surgery, Batterjee Medical College, Jeddah 21442, Saudi Arabia; (M.F.A.); (L.O.A.H.); (M.A.A.); (A.K.); (H.I.W.); (K.A.A.); (A.N.A.J.); (A.M.K.)
| | - Ayah Nabil Al Jehani
- Department of Medicine and Surgery, Batterjee Medical College, Jeddah 21442, Saudi Arabia; (M.F.A.); (L.O.A.H.); (M.A.A.); (A.K.); (H.I.W.); (K.A.A.); (A.N.A.J.); (A.M.K.)
| | - Amal Mohamed Kayali
- Department of Medicine and Surgery, Batterjee Medical College, Jeddah 21442, Saudi Arabia; (M.F.A.); (L.O.A.H.); (M.A.A.); (A.K.); (H.I.W.); (K.A.A.); (A.N.A.J.); (A.M.K.)
| | - Samera AlBasri
- Obsetetrics and Gynecology Department, King Abdulaziz University Hospital, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| |
Collapse
|
5
|
Guo Y, Yang J, Chen H, Zhou Y, Yang Y, Wang B, Zha L, Bai D, Li W, Tang X, Fang Z, Li F, Jin L. Enhancing understanding of endometrial function in patients with PCOS: clinical and immunological insights. J Ovarian Res 2025; 18:52. [PMID: 40075432 PMCID: PMC11900192 DOI: 10.1186/s13048-025-01638-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
OBJECTIVE To evaluate the pregnancy and perinatal outcomes of different phenotypes of polycystic ovary syndrome (PCOS) patients during the frozen embryo transfer (FET) cycles. Additionally, to analyze the T cell balance in the endometrium of PCOS patients and explore its relationship with various PCOS phenotypes. DESIGN Retrospective cohort study. SETTING A single academically affiliated reproductive medicine center. PATIENTS 21,074 FET cycles were included and divided into two groups based on the diagnosis of PCOS. Patients with PCOS were further categorized into four phenotypic groups: PCOM + HA + OA, PCOM + HA, PCOM + OA, and HA + OA. Endometrial biopsies from 21 PCOS patients and 26 controls were obtained to analyze T cell subsets. METHODS Pregnancy and perinatal outcomes, as well as T cell subset abundance were compared between women with and without PCOS. Multiple logistic regression models were employed to adjust for confounding factors impacting pregnancy-related outcomes. Flow cytometry was utilized to analyze the abundance of T cell subsets. MAIN OUTCOME MEASURES Pregnancy and perinatal outcomes were assessed. T cell subsets including CD4+CD8-T cells, CD4-CD8+T cells, Th1, Th2, Th17 and Treg cells in the endometrium were determined by flow cytometry. RESULTS There was a significantly increased incidence of miscarriage, hypertensive disorders of pregnancy (HDP), preterm birth (PTB), and even fetal malformations across different phenotypes of PCOS women, especially those with the hyperandrogenic phenotype. Th1 cells decreased while Th2 cells increased significantly in the PCOS endometrium. CONCLUSIONS The unfavorable pregnancy and perinatal outcomes in FET cycles and T cell imbalance both suggest the endometrial dysfunction of PCOS patients, especially those with the hyperandrogenic phenotype.
Collapse
Affiliation(s)
- Yaxin Guo
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingfei Yang
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Chen
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yueping Zhou
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Yang
- State Key Laboratory of Material Processing and Die & Mould Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Biao Wang
- College of Life Science, Shaanxi Normal University, Xi'an, China
| | - Luyang Zha
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dijia Bai
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenxuan Li
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaojuan Tang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zishui Fang
- Department of Urology, Peking University First Hospital, Beijing, China
| | - Fei Li
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Lei Jin
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China.
- National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
6
|
Wang Y, He X, Yang C, Ding J. Global research on NK cells in miscarriage: a bibliometric study. Front Med (Lausanne) 2025; 12:1513213. [PMID: 40034381 PMCID: PMC11872723 DOI: 10.3389/fmed.2025.1513213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
Background This study aimed to assess the evolution, trends, and research hotspots of publications related to natural killer (NK) cells and miscarriage. Methods The literature on NK cells and miscarriage was retrieved from the Web of Science Core Collection. VOSviewer and CiteSpace were used to analyze the publication years, countries, institutions, journals, highly cited authors, categories, and citation bursts of keywords. Results A total of 1,275 articles were analyzed. The annual publication outputs showed steady growth, with the majority of publications in 2020 and citations in 2022. The number of publications in this field fluctuated from 1981 to 2023, with a slight downward trend observed. However, the number of citations increased steadily until 2023, followed by a minor decline. The United States contributed the highest number of publications and had the highest h-index. The American Journal of Reproductive Immunology ranked first in terms of number of publications and h-index. Reproductive biology, immunology, and obstetrics and gynecology were the most representative disciplines. Kwak-kim J, Chaouat G, and Croy BA were the top three most productive authors in the field. Keyword burst analysis demonstrated that the immune system and cytotoxicity receptors were current research hotspots. Conclusion This is the first bibliometric study to comprehensively summarize trends and advances in the study of NK cells in miscarriage. This information highlights the recent research frontiers and emerging directions and provides a reference for subsequent research in the future.
Collapse
Affiliation(s)
- Yinan Wang
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China
| | - Xiaoqin He
- Teaching Office, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chaogang Yang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, China
- Hubei Cancer Clinical Study Center, Wuhan, China
- The Clinical Medical Research Center of Peritoneal Cancer of Wuhan, Wuhan, China
| | - Jinli Ding
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China
| |
Collapse
|
7
|
Garmendia JV, De Sanctis CV, Hajdúch M, De Sanctis JB. Exploring the Immunological Aspects and Treatments of Recurrent Pregnancy Loss and Recurrent Implantation Failure. Int J Mol Sci 2025; 26:1295. [PMID: 39941063 PMCID: PMC11818386 DOI: 10.3390/ijms26031295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/26/2025] [Accepted: 01/30/2025] [Indexed: 02/16/2025] Open
Abstract
Recurrent pregnancy loss (RPL) is defined as the occurrence of two or more consecutive pregnancy losses before 24 weeks of gestation. It affects 3-5% of women who are attempting to conceive. RPL can stem from a variety of causes and is frequently associated with psychological distress and a diminished quality of life. By contrast, recurrent implantation failure (RIF) refers to the inability to achieve a successful pregnancy after three or more high-quality embryo transfers or at least two instances of egg donation. RIF shares several causative factors with RPL. The immunological underpinnings of these conditions involve alterations in uterine NK cells, reductions in M2 macrophages and myeloid-derived suppressor cells, an increased Th1/Th2 ratio, a decreased Treg/Th17 ratio, the presence of shared ≥3 HLA alleles between partners, and autoimmune disorders. Various therapeutic approaches have been employed to address these immunological concerns, achieving varying degrees of success, although some therapies remain contentious within the medical community. This review intends to explore the immunological factors implicated in RPL and RIF and to analyze the immunological treatments employed for these conditions, which may include steroids, intravenous immunoglobulins, calcineurin inhibitors, anti-TNF antibodies, intralipid infusions, granulocyte colony-stimulating factor, and lymphocyte immunotherapy.
Collapse
Affiliation(s)
- Jenny Valentina Garmendia
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 779 00 Olomouc, Czech Republic; (J.V.G.); (C.V.D.S.); (M.H.)
| | - Claudia Valentina De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 779 00 Olomouc, Czech Republic; (J.V.G.); (C.V.D.S.); (M.H.)
| | - Marián Hajdúch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 779 00 Olomouc, Czech Republic; (J.V.G.); (C.V.D.S.); (M.H.)
- Czech Advanced Technologies and Research Institute (CATRIN), Institute of Molecular and Translational Medicine, 779 00 Olomouc, Czech Republic
- Laboratory of Experimental Medicine, University Hospital Olomouc, 779 00 Olomouc, Czech Republic
| | - Juan Bautista De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, 779 00 Olomouc, Czech Republic; (J.V.G.); (C.V.D.S.); (M.H.)
- Czech Advanced Technologies and Research Institute (CATRIN), Institute of Molecular and Translational Medicine, 779 00 Olomouc, Czech Republic
| |
Collapse
|
8
|
Hautamäki H, Gissler M, Heikkinen‐Eloranta J, Tiitinen A, Peuranpää P. Pregnancy and perinatal outcomes in women with recurrent pregnancy loss-A case-control study. Acta Obstet Gynecol Scand 2025; 104:368-379. [PMID: 39711128 PMCID: PMC11782061 DOI: 10.1111/aogs.15039] [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: 09/25/2024] [Revised: 11/15/2024] [Accepted: 12/05/2024] [Indexed: 12/24/2024]
Abstract
INTRODUCTION Recurrent pregnancy loss (RPL), defined as two or more pregnancy losses, might be associated with elevated obstetrical and perinatal risks in the following pregnancies. RPL and pregnancy problems related to placental development may have similar etiological features. This study explores the incidences of pregnancy and perinatal outcomes in women with RPL. MATERIAL AND METHODS This retrospective case-control study investigated the outcomes of the next subsequent singleton pregnancy after thorough RPL examination (n = 360) in Helsinki University Hospital, Finland, in 2007-2016. Data for cases and four control women for each case, matched for age, parity, delivery month, year, and hospital (n = 1440), were retrieved from the Medical Birth Register. Primary outcomes were pregnancy and delivery complications, perinatal outcomes, and characteristics of pregnancy follow-up. Secondary outcomes were maternal and neonatal diagnoses. Associations between RPL and outcomes were estimated with risk ratios with 95% confidence intervals (CI). In sub-analyses, we compared the outcomes of secondary RPL with multipara controls and women with unexplained or explained RPL. RESULTS Women with RPL had a higher risk for gestational hypertension (3.1% vs. 1.4%, risk ratio [RR] 2.20 [Confidence interval (CI) 1.06-4.55], p = 0.03), preterm birth (8.9% vs. 5.8%, RR 1.54 [CI 1.04-2.28], p = 0.04), malpresentation of the fetus (3.3% vs. 1.5%, RR 2.18 [CI 1.09-4.37], p = 0.02), premature rupture of membranes (5.6% vs. 2.4%, RR 2.35 [CI 1.37-4.04], p = 0.002), and had more prenatal visits than controls. Mode of delivery was comparable between the study groups, although RPL women had more induced labor (28.1% vs. 22.2%, RR 1.26 [CI 1.04-1.53], p = 0.02). Mean birthweight was lower (3387 ± 680 g) in RPL women's newborns than in the control group (3482 ± 564 g, p = 0.02), and the risk of umbilical artery pH <7.10 (6.7% vs. 3.6%, RR 1.85 [CI 1.15-2.95], p = 0.03) was higher. Risk ratio for small for gestational age was higher in the secondary RPL group than in multipara controls (5.1% vs. 2.0%, RR 2.50 [CI 1.15-5.42], p = 0.02). CONCLUSIONS Women with a history of RPL seem to have higher risks in their subsequent pregnancies and should therefore be monitored carefully. These findings support the theory of placental development being the common nominator behind hypertensive pregnancy disorders and RPL.
Collapse
Affiliation(s)
- Hanna Hautamäki
- The Department of Obstetrics and GynecologyUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | - Mika Gissler
- Academic Primary Health Care Centre, Region Stockholm & Department of Molecular Medicine and SurgeryKarolinska InstitutetStockholmSweden
- Department of Data and AnalyticsFinnish Institute for Health and WelfareHelsinkiFinland
| | - Jenni Heikkinen‐Eloranta
- The Department of Obstetrics and GynecologyUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
| | | | - Pirkko Peuranpää
- The Department of Obstetrics and GynecologyUniversity of Helsinki and Helsinki University HospitalHelsinkiFinland
- University of HelsinkiHelsinkiFinland
| |
Collapse
|
9
|
Bezemer RE, Brenøe JE, Schoots MH, Feenstra ME, van Goor H, Ganzevoort W, Gordijn SJ, Prins JR. Effects of sildenafil treatment on placental immune cell subsets in early-onset fetal growth restriction. Placenta 2025; 159:62-69. [PMID: 39644752 DOI: 10.1016/j.placenta.2024.11.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/11/2024] [Accepted: 11/24/2024] [Indexed: 12/09/2024]
Abstract
INTRODUCTION Early onset fetal growth restriction is a common pregnancy complication with significant risk of perinatal mortality and morbidity. The most common etiology is placental insufficiency, reflected by several placental lesions that appear with fetal growth restriction. Placental immune cells are involved in almost all aspects of the development of the placenta and immune cell imbalances have been related to common pregnancy complications. The STRIDER trial investigated the therapeutic potential of sildenafil. No clinical improvements were observed, however, since sildenafil can have immunological effects, we aimed to investigate if sildenafil alters local placental immune cells. METHODS Placental samples from 146 patients were included from the STRIDER trial and stained with IHC for leukocytes (CD45), macrophages (CD68 and CD206), T cells (CD3 and CD8), regulatory T cells (FOXP3) and NK cells (CD56). Immune cells were quantified in the decidua basalis and villi at term using a trained detection classifier. In addition, maternal plasma cytokines were measured at inclusion. RESULTS In the sildenafil group, numbers of CD3+ T cells, CD68+ and CD206+ macrophages and CD56+ NK cell were greater in the decidua basalis compared to the control group. Correlating maternal plasma cytokines to placental immune cell subsets showed predominantly negative correlations in the placebo group, whereas most cytokines correlated positively to placental immune cells in the sildenafil group. DISCUSSION Our data demonstrates the immunomodulatory effects of sildenafil in pregnancies complicated by early onset fetal growth restriction and offers valuable insights on the use of immunomodulatory drugs in pregnancy.
Collapse
Affiliation(s)
- R E Bezemer
- Department of Gynecology and Obstetrics, University Medical Center Groningen, University of Groningen, 9713 GZ, Groningen, the Netherlands; Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9713 GZ, Groningen, the Netherlands.
| | - J E Brenøe
- Department of Gynecology and Obstetrics, University Medical Center Groningen, University of Groningen, 9713 GZ, Groningen, the Netherlands; Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9713 GZ, Groningen, the Netherlands
| | - M H Schoots
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9713 GZ, Groningen, the Netherlands
| | - M E Feenstra
- Department of Gynecology and Obstetrics, University Medical Center Groningen, University of Groningen, 9713 GZ, Groningen, the Netherlands; Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9713 GZ, Groningen, the Netherlands
| | - H van Goor
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9713 GZ, Groningen, the Netherlands
| | - W Ganzevoort
- Department of Gynecology and Obstetrics, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ, Amsterdam, the Netherlands; Amsterdam Reproduction and Development Research Institute, Amsterdam, the Netherlands
| | - S J Gordijn
- Department of Gynecology and Obstetrics, University Medical Center Groningen, University of Groningen, 9713 GZ, Groningen, the Netherlands
| | - J R Prins
- Department of Gynecology and Obstetrics, University Medical Center Groningen, University of Groningen, 9713 GZ, Groningen, the Netherlands
| |
Collapse
|
10
|
Martinez VO, Dos Santos NR, Bah HAF, Gomes EA, Costa DO, Souza MISS, de Carvalho CF, Andrade NC, Menezes-Filho JA. Impact of chronic toxoplasmosis in pregnancy: Association between maternal IgG antibodies against T. gondii and neurocognitive development effects. Neurotoxicology 2025; 106:10-16. [PMID: 39638154 DOI: 10.1016/j.neuro.2024.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/24/2024] [Accepted: 12/01/2024] [Indexed: 12/07/2024]
Abstract
Toxoplasmosis presents notable hazards in the context of pregnancy, impacting the health of the mother and the neurodevelopment of the fetus via immune reactions and possible vertical transmission. The maternal immune response from chronic Toxoplasma gondii (T. gondii) infection may negatively influence fetal neurodevelopment. This research evaluated the association between the seroprevalence of chronic T. gondii and cytomegalovirus infection in pregnant women and the neuropsychological development of their children at 12 months of age. A follow-up study evaluated women during the gestational period and their respective infants. The pregnant women were tested for the presence of antibodies to infectious agents: T. gondii, cytomegalovirus (CMV), syphilis, human immunodeficiency virus (HIV), hepatitis B and C. Detailed information about the newborns was extracted from medical records. At 12 ± 3 months of age, the infant's neurodevelopment was assessed using the Bayley-III Scales of Infant and Toddler Development by a trained specialist under the supervision of a neuropsychologist. A statistically significant association was found between maternal IgG anti-T. gondii levels and lower scores on the Bayley-III cognition scale, with a non-standardized β-coefficient of -0.078 (95 %-CI: -0.144 to -0.013), accounting for 35.1 % of the variation in this outcome. These results suggest that chronic maternal T. gondii infection, even without vertical transmission, may be associated with subtle changes in the child's cognitive development. Therefore, monitoring and early intervention are essential to identify and address possible delays in childhood neurodevelopment related to chronic maternal toxoplasmosis.
Collapse
Affiliation(s)
- Victor Otero Martinez
- Graduate Program in Pharmacy, College of Pharmacy, Federal University of Bahia, Salvador, Bahia, Brazil.
| | - Nathália Ribeiro Dos Santos
- Graduate Program in Pharmacy, College of Pharmacy, Federal University of Bahia, Salvador, Bahia, Brazil; Laboratory of Toxicology, College of Pharmacy, Federal University of Bahia, Salvador, Bahia, Brazil
| | - Homègnon Antonin Ferréol Bah
- Laboratory of Toxicology, College of Pharmacy, Federal University of Bahia, Salvador, Bahia, Brazil; Graduate Program in Public Health, Federal University of Bahia, Salvador, Bahia, Brazil
| | - Erival Amorim Gomes
- Laboratory of Toxicology, College of Pharmacy, Federal University of Bahia, Salvador, Bahia, Brazil
| | - Daisy Oliveira Costa
- Graduate Program in Pharmacy, College of Pharmacy, Federal University of Bahia, Salvador, Bahia, Brazil; Laboratory of Toxicology, College of Pharmacy, Federal University of Bahia, Salvador, Bahia, Brazil
| | | | | | | | - José Antônio Menezes-Filho
- Graduate Program in Pharmacy, College of Pharmacy, Federal University of Bahia, Salvador, Bahia, Brazil; Laboratory of Toxicology, College of Pharmacy, Federal University of Bahia, Salvador, Bahia, Brazil; Graduate Program in Public Health, Federal University of Bahia, Salvador, Bahia, Brazil
| |
Collapse
|
11
|
Yang Y, Zhao A, Wang T, Tang Q, Qi S, Shi X, Wang F, Gao Y. Identification of driving genes of recurrent miscarriage based on transcriptome sequencing and immunoinfiltration analysis. Int Immunopharmacol 2024; 143:113095. [PMID: 39395380 DOI: 10.1016/j.intimp.2024.113095] [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: 04/15/2024] [Revised: 09/01/2024] [Accepted: 09/03/2024] [Indexed: 10/14/2024]
Abstract
AIMS Recurrent miscarriage (RM) plagues 1 %-5 % women of childbearing age. Facing the limitations of clinical treatment, its pathological mechanism remains to be clarified. METHODS Decidual tissues of three induced abortions and three RM were collected for transcriptome sequencing. The pathological features of RM were identified by differential expression genes (DEGs) analysis, GSEA, GO and KEGG analysis, and a protein-protein interaction network was constructed for DEGs, and six algorithms were used to identify hub genes. In addition, the immune characteristics of RM patients were identified by CIBERSORT, and the correlation between them and hub genes was analyzed. Furthermore, in single-cell level, different cells were grouped according to the expression level of hub genes, and the expression ratio and abundance of hub genes in different cells and their regulation on cell function were explored. RESULTS Transcriptome sequencing of patients with RM showed that a large number of genes were down-regulated, which was related to fibroblast proliferation, epithelial cell migration, female pregnancy and cell chemotaxis. Fifteen hub genes were identified by constructing a protein-protein interaction network, among which DUSP1, NR4A1 and THBS1 were involved in cell migration and chemotaxis. Immune cell infiltration analysis showed that the infiltration of T cells, macrophages and NK cells was abnormal, and there was a significant correlation with hub genes. Moreover, we found that compared with the expression of DUSP1, the non-expression of DUSP1 will reduce the extracellular matrix formation of fibroblasts and the chemotaxis of macrophages. At the same time, it is worth noting that the expression ratio and abundance of hub genes are decreased in epithelial cells, fibroblasts, macrophages and NK cells. Furthermore, single-cell analysis and in vitro and in vivo experiments show that DUSP1 and NR4A1 are low-expressed in different cells of RM patients, which is accompanied by the inhibition of fibroblast proliferation and macrophage chemotaxis. Drug prediction and screening based on hub genes show that Cinobufagin and calmidazolium are expected to be candidate drugs for RM. CONCLUSION Hub genes such as DUSP1, NR4A1 and THBS1 participate in RM by regulating epithelial cell migration, fibroblast proliferation and macrophage chemotaxis, which will provide new insight for the diagnosis and targeted therapy of RM.
Collapse
Affiliation(s)
- Yijun Yang
- The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, China
| | - Ai Zhao
- The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, China
| | - Ting Wang
- The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, China
| | - Qi Tang
- The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, China
| | - Suwan Qi
- Affiliated Women's Hospital of Jiangnan University, China
| | - Xiaoling Shi
- The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, China
| | - Fei Wang
- The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, China.
| | - Yingchun Gao
- The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, China.
| |
Collapse
|
12
|
Deng H, Chen Y, Xing J, Zhang N, Xu L. Systematic low-grade chronic inflammation and intrinsic mechanisms in polycystic ovary syndrome. Front Immunol 2024; 15:1470283. [PMID: 39749338 PMCID: PMC11693511 DOI: 10.3389/fimmu.2024.1470283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 12/03/2024] [Indexed: 01/04/2025] Open
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent endocrine and metabolic disorder affecting 6-20% of women of childbearing age worldwide. Immune cell imbalance and dysregulation of inflammatory factors can lead to systematic low-grade chronic inflammation (SLCI), which plays a pivotal role in the pathogenesis of PCOS. A significant higher infiltration of immune cells such as macrophages and lymphocytes and pro-inflammatory factors IL-6 and TNF-α has been detected in PCOS organ systems, impacting not only the female reproductive system but also other organs such as the cardiovascular, intestine, liver, thyroid, brain and other organs. Obesity, insulin resistance (IR), steroid hormones imbalance and intestinal microecological imbalance, deficiencies in vitamin D and selenium, as well as hyperhomocysteinemia (HHcy) can induce systematic imbalance between pro-inflammatory and anti-inflammatory cells and molecules. The pro-inflammatory cells and cytokines also interact with obesity, steroid hormones imbalance and IR, leading to increased metabolic imbalance and reproductive-endocrine dysfunction in PCOS patients. This review aims to summarize the dysregulation of immune response in PCOS organ system and the intrinsic mechanisms affecting SLCI in PCOS to provide new insights for the systemic inflammatory treatment of PCOS in the future.
Collapse
Affiliation(s)
- Hongxia Deng
- Reproductive Endocrinology and Regulation Laboratory, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Yan Chen
- Reproductive Endocrinology and Regulation Laboratory, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jilong Xing
- Division of Renal and Endocrinology, Qin Huang Hospital, Xi’an, China
| | - Nannan Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
- National Center for Birth Defect Monitoring, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Liangzhi Xu
- Reproductive Endocrinology and Regulation Laboratory, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
13
|
Westergaard D, Lundgaard AT, Vomstein K, Fich L, Hviid KVR, Egerup P, Christiansen AMH, Nielsen JR, Lindman J, Holm PC, Hartwig TS, Jørgensen FS, Zedeler A, Kolte AM, Westh H, Jørgensen HL, la Cour Freiesleben N, Banasik K, Brunak S, Nielsen HS. Immune changes in pregnancy: associations with pre-existing conditions and obstetrical complications at the 20th gestational week-a prospective cohort study. BMC Med 2024; 22:583. [PMID: 39696496 DOI: 10.1186/s12916-024-03797-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 11/25/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Pregnancy is a complex biological process and serious complications can arise when the delicate balance between the maternal and semi-allogeneic fetal immune systems is disrupted or challenged. Gestational diabetes mellitus (GDM), pre-eclampsia, preterm birth, and low birth weight pose serious threats to maternal and fetal health. Identification of early biomarkers through an in-depth understanding of molecular mechanisms is critical for early intervention. METHODS We analyzed the associations between 47 proteins involved in inflammation, chemotaxis, angiogenesis, and immune system regulation, maternal and neonatal health outcomes, and the baseline characteristics and pre-existing conditions of the mother in a prospective cohort of 1049 pregnant women around the 20th gestational week. We used Bayesian linear regression models to examine the impact of risk factors on biomarker levels and Bayesian cause-specific parametric proportional hazards models to analyze the effect of biomarkers on maternal and neonatal outcomes. We evaluated the predictive value of baseline characteristics and 47 proteins using machine-learning models and identified the most predictive biomarkers using Shapley additive explanation scores. RESULTS Associations were identified between specific inflammatory markers and several conditions, including maternal age and pre-pregnancy body mass index, chronic diseases, complications from prior pregnancies, and COVID-19 exposure. Smoking during pregnancy affected GM-CSF and 9 other biomarkers. Distinct biomarker patterns were observed for different ethnicities. Within obstetric complications, IL-6 inversely correlated with pre-eclampsia risk, while birth weight to gestational age ratio was linked to markers including VEGF and PlGF. GDM was associated with IL-1RA, IL-17D, and eotaxin-3. Severe postpartum hemorrhage correlated with CRP, IL-13, and proteins of the IL-17 family. Predictive modeling yielded area under the receiver operating characteristic curve values of 0.708 and 0.672 for GDM and pre-eclampsia, respectively. Significant predictive biomarkers for GDM included IL-1RA and eotaxin-3, while pre-eclampsia prediction yielded the highest predictions when including MIP-1β, IL-1RA, and IL-12p70. CONCLUSIONS Our study provides novel insights into the interplay between preexisting conditions and immune dysregulation in pregnancy. These findings contribute to our understanding of the pathophysiology of obstetric complications and the identification of novel biomarkers for early intervention(s) to improve maternal and fetal health.
Collapse
Affiliation(s)
- David Westergaard
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Methods and Analysis, Statistics Denmark, Copenhagen, Denmark
- Department of Obstetrics and Gynecology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Agnete Troen Lundgaard
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kilian Vomstein
- Department of Obstetrics and Gynecology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Line Fich
- Department of Obstetrics and Gynecology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | | | - Pia Egerup
- Department of Obstetrics and Gynecology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | | | | | - Johanna Lindman
- Department of Obstetrics and Gynecology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Peter Christoffer Holm
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tanja Schlaikjær Hartwig
- Department of Obstetrics and Gynecology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Finn Stener Jørgensen
- Department of Obstetrics and Gynecology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anne Zedeler
- Department of Obstetrics and Gynecology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Astrid Marie Kolte
- Department of Obstetrics and Gynecology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Henrik Westh
- Department of Clinical Microbiology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Henrik Løvendahl Jørgensen
- Department of Clinical Biochemistry, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nina la Cour Freiesleben
- Department of Obstetrics and Gynecology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Karina Banasik
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Obstetrics and Gynecology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Henriette Svarre Nielsen
- Department of Obstetrics and Gynecology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark.
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
14
|
Sebina I, Bidgood C, Stalley F, Hartel G, Stark T, Callaway L, Amoako A, Lehner C, Dekker Nitert M, Phipps S. Pre-pregnancy obesity is associated with an altered maternal metabolome and reduced Flt3L expression in preterm birth. Sci Rep 2024; 14:30027. [PMID: 39627409 PMCID: PMC11615298 DOI: 10.1038/s41598-024-81194-4] [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: 03/14/2024] [Accepted: 11/25/2024] [Indexed: 12/06/2024] Open
Abstract
Mechanisms linking pre-pregnancy obesity to increased preterm birth risk are unclear. Here, we examined the impact of pre-pregnancy obesity on metabolites, Fms-related tyrosine kinase 3 ligand (Flt3L), and proinflammatory cytokine profiles in preterm birth. We used cytokine bead array, ELISA and Gas Chromatography-Mass Spectrometry (GC-MS) to determine cytokine and metabolite profiles in maternal and cord blood samples from 124 pregnant women in Australia, who gave birth at term (n = 86) or preterm (n = 38). Besides the expected variations in birth weight and gestational age, all demographic characteristics, including pre-pregnancy body mass index, were similar between the term and preterm birth groups. Mothers in the preterm birth group had reduced Flt3L (P = 0.002) and elevated IL-6 (P = 0.002) compared with term birthing mothers. Among mothers who gave birth preterm, those with pre-pregnancy obesity had lower Flt3L levels (P = 0.02) compared with lean mothers. Flt3L and IL-6 were similar in cord blood across both groups, but TNFα levels (P = 0.02) were reduced in preterm newborns. Metabolomic analysis revealed significant shifts in essential metabolites in women with pre-pregnancy obesity, some of which were linked to preterm births. Our findings suggest that maternal pre-pregnancy obesity alters the metabolome and reduces Flt3L expression, potentially increasing risk of preterm birth.
Collapse
Affiliation(s)
- Ismail Sebina
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia.
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia.
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, 4000, QLD, Australia.
| | - Charles Bidgood
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, 4000, QLD, Australia
| | - Felicity Stalley
- Women's and Newborn Services, Royal Brisbane and Women's Hospital, Herston, QLD, 4006, Australia
| | - Gunter Hartel
- Statistics Unit, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
- School of Public Health, The University of Queensland, Brisbane, QLD, Australia
| | - Terra Stark
- Metabolomics Australia (Queensland Node), The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Leonie Callaway
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
- Women's and Newborn Services, Royal Brisbane and Women's Hospital, Herston, QLD, 4006, Australia
| | - Akwasi Amoako
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
- Women's and Newborn Services, Royal Brisbane and Women's Hospital, Herston, QLD, 4006, Australia
| | - Christoph Lehner
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
- Women's and Newborn Services, Royal Brisbane and Women's Hospital, Herston, QLD, 4006, Australia
| | - Marloes Dekker Nitert
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Simon Phipps
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, 4000, QLD, Australia
| |
Collapse
|
15
|
Dietz S, Hebel J, Rühle J, Huff A, Eltzschig HK, Lajqi T, Poets CF, Gille C, Köstlin‐Gille N. Impact of the adenosine receptor A2BR expressed on myeloid cells on immune regulation during pregnancy. Eur J Immunol 2024; 54:e2451149. [PMID: 39460389 PMCID: PMC11628929 DOI: 10.1002/eji.202451149] [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: 03/18/2024] [Revised: 10/04/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024]
Abstract
During pregnancy, the maternal immune system must carefully balance protection against pathogens with tolerance toward the semiallogeneic fetus. Dysfunctions of the immune system can lead to severe complications such as preeclampsia, fetal growth restriction, or pregnancy loss. Adenosine plays a role in physiological processes and plasma-level increase during pregnancy. The adenosine receptor A2B (A2BR), which is expressed on both, immune and nonimmune cells, is activated by high adenosine concentrations, achieved during pregnancy. We investigated the impact of A2BR expressed on myeloid cells on immune regulation during pregnancy using a mouse model with myeloid deficiency for A2BR. We demonstrate systemic changes in myeloid and lymphoid cell populations during pregnancy in A2BR-KO (Adora2B923f/f-LysMCre) mice with increased monocytes, neutrophils, and T cells but decreased B cells as well as altered T-cell subpopulations with decreased Th1 cells and Tregs and increased Th17 cells. Lack of A2BR on myeloid cells caused an increased systemic expression of IL-6 but decreased systemic accumulation and function of MDSC and reduced numbers of uterine natural killer cells. The pregnancy outcome was only marginally affected. Our results demonstrate that A2BR on myeloid cells plays a role in immune regulation during pregnancy, but the clinical impact on pregnancy remains unclear.
Collapse
Affiliation(s)
- Stefanie Dietz
- Department of NeonatologyTuebingen University Children's HospitalTuebingenGermany
- Department of NeonatologyHeidelberg University, Medical FacultyHeidelbergGermany
| | - Janine Hebel
- Department of NeonatologyTuebingen University Children's HospitalTuebingenGermany
| | - Jessica Rühle
- Department of NeonatologyTuebingen University Children's HospitalTuebingenGermany
| | - Alisha Huff
- Department of NeonatologyTuebingen University Children's HospitalTuebingenGermany
| | | | - Trim Lajqi
- Department of NeonatologyHeidelberg University, Medical FacultyHeidelbergGermany
| | - Christian F. Poets
- Department of NeonatologyTuebingen University Children's HospitalTuebingenGermany
| | - Christian Gille
- Department of NeonatologyHeidelberg University, Medical FacultyHeidelbergGermany
| | - Natascha Köstlin‐Gille
- Department of NeonatologyTuebingen University Children's HospitalTuebingenGermany
- Department of NeonatologyHeidelberg University, Medical FacultyHeidelbergGermany
| |
Collapse
|
16
|
Jin M, Liu X, Liu X, Wu Y, Zhang Y, Zhang L, Li Z, Ye R, Li N. Association of pre-/early pregnancy high blood pressure and pregnancy outcomes: a systemic review and meta-analysis. J Matern Fetal Neonatal Med 2024; 37:2296366. [PMID: 38151254 DOI: 10.1080/14767058.2023.2296366] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 12/13/2023] [Indexed: 12/29/2023]
Abstract
BACKGROUND Maternal high blood pressure (BP) was associated with adverse pregnancy outcomes. This study aimed to synthesize evidence on the association between high BP prior to or in early pregnancy with maternal and fetal complications. METHODS We searched the cohort studies assessing the effect of high BP in the Medline, Embase, Web of Science and China National Knowledge Internet databases. A random-effects model was used to estimate the pooled odds ratios (ORs) with 95% confidence intervals (CIs). The protocol was registered in PROSPERRO (CRD 42023414945). RESULTS 23 eligible studies were identified. High BP prior to or in early pregnancy was associated with higher odds of hypertensive disorders of pregnancy (OR 2.90, 95% CI 1.91-3.89), gestational hypertension (2.56, 2.01-3.12), preeclampsia (3.20, 2.66-3.74), gestational diabetes mellitus (1.71, 1.36-2.06), preterm birth (1.66, 1.39-1.93), stillbirth (2.01, 1.45-2.58) and neonatal intensive care unit admission (1.22, 1.08-1.37). Subgroup analyses indicated that pre-hypertension could significantly increase the odds of these outcomes except for stillbirth, though the odds were lower than hypertension. CONCLUSIONS High BP prior to or in early pregnancy was associated with adverse pregnancy outcomes and this association increased with hypertension severity. The findings emphasized an urgent need for heightened surveillance for maternal BP, especially pre-hypertensive status.
Collapse
Affiliation(s)
- Ming Jin
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, China
- Institute of Reproductive and Child Health, Peking University/Key Laboratory of Reproductive Health, National Health Commission of the People's Republic of China, Beijing, China
| | - Xiaowen Liu
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, China
- Institute of Reproductive and Child Health, Peking University/Key Laboratory of Reproductive Health, National Health Commission of the People's Republic of China, Beijing, China
| | - Xiaojing Liu
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, China
- Institute of Reproductive and Child Health, Peking University/Key Laboratory of Reproductive Health, National Health Commission of the People's Republic of China, Beijing, China
| | - Yaxian Wu
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, China
- Institute of Reproductive and Child Health, Peking University/Key Laboratory of Reproductive Health, National Health Commission of the People's Republic of China, Beijing, China
| | - Yali Zhang
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, China
- Institute of Reproductive and Child Health, Peking University/Key Laboratory of Reproductive Health, National Health Commission of the People's Republic of China, Beijing, China
| | - Le Zhang
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, China
- Institute of Reproductive and Child Health, Peking University/Key Laboratory of Reproductive Health, National Health Commission of the People's Republic of China, Beijing, China
| | - Zhiwen Li
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, China
- Institute of Reproductive and Child Health, Peking University/Key Laboratory of Reproductive Health, National Health Commission of the People's Republic of China, Beijing, China
| | - Rongwei Ye
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, China
- Institute of Reproductive and Child Health, Peking University/Key Laboratory of Reproductive Health, National Health Commission of the People's Republic of China, Beijing, China
| | - Nan Li
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, China
- Institute of Reproductive and Child Health, Peking University/Key Laboratory of Reproductive Health, National Health Commission of the People's Republic of China, Beijing, China
| |
Collapse
|
17
|
High-resolution spatial multiomic census of the human placenta. Nat Med 2024; 30:3435-3436. [PMID: 39567717 DOI: 10.1038/s41591-024-03353-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
|
18
|
Ma F, Feng X, Feng S, Liu J, Li J, Mo L, Xu L, Liu Y, Wu J, Yang P, Ning Y. Impaired inducibility of immune regulatory capacity of peripheral B cells of patients with recurrent pregnancy loss. Immunol Res 2024; 72:1502-1514. [PMID: 39495420 DOI: 10.1007/s12026-024-09549-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/25/2024] [Indexed: 11/05/2024]
Abstract
The pathogenesis of recurrent pregnancy loss (RPL) is unclear. RPL may have an association with disruption of immune tolerance. The aim of this study is to characterize the inducibility of immune regulatory ability in peripheral naïve B cells of patients with RPL. In this study, blood samples were taken from patients with RPL. B220+ B cells were isolated by flow cytometry cell sorting. The gene profile of B cells was analyzed using RNA sequencing (RNAseq). The results showed that peripheral B220+ B cells of RPL patients had lower expression of IL10 and exacerbated ER stress. The induction of IL10 expression in peripheral B220+ B cells of RPL patients were impaired. High ubiquitination of c-Maf inducing protein (CMIP) was detected in RPL B cells. Exposure to thapsigargin (an ER stress agonist) decreased the amount of CMIP in B cells. The effects of ER stress on reducing CMIP quantity in B cells were mediated by the histone H2B E3 ubiquitin ligase ring finger protein 20 (RNF20). Inhibition of RNF20 or ER stress restored the inducibility of immune regulatory functions of B220+ B cells of RPL patients. In summary, peripheral B cells in patients with RPL show impaired immune regulation capacity, in which exacerbated ER stress plays a crucial role. Regulation of ER stress or inhibition of RNF20 can restore the immune regulatory capacity in the B cells.
Collapse
Affiliation(s)
- Fei Ma
- Department of Chinese Traditional Medicine, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China.
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.
| | - Xiaoyang Feng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Shiyu Feng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Jin Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Jia Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Lihua Mo
- Department of General Practice Medicine, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
- Institute of Allergy & Immunology of Shenzhen University, State Key Laboratory of Respiratory Diseases Allergy Division, Shenzhen University, Room A7-509 at Lihu Campus of Shenzhen University, 1066 Xueyuan Blvd, Shenzhen, 518055, China
| | - Lingzhi Xu
- Department of Immunology, School of Basic Medical Sciences, Weifang Medical University, Weifang, China
| | - Yulei Liu
- Department of Chinese Traditional Medicine, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Jiaman Wu
- Department of Chinese Traditional Medicine, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Pingchang Yang
- Institute of Allergy & Immunology of Shenzhen University, State Key Laboratory of Respiratory Diseases Allergy Division, Shenzhen University, Room A7-509 at Lihu Campus of Shenzhen University, 1066 Xueyuan Blvd, Shenzhen, 518055, China.
| | - Yan Ning
- Department of Chinese Traditional Medicine, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China.
| |
Collapse
|
19
|
Uța C, Tîrziu A, Zimbru EL, Zimbru RI, Georgescu M, Haidar L, Panaitescu C. Alloimmune Causes of Recurrent Pregnancy Loss: Cellular Mechanisms and Overview of Therapeutic Approaches. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1896. [PMID: 39597081 PMCID: PMC11596804 DOI: 10.3390/medicina60111896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/16/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024]
Abstract
Recurrent pregnancy loss (RPL) is a complex early pregnancy complication affecting 1-2% of couples and is often linked to immune dysfunction. Aberrations in T and B cell subpopulations, as well as natural killer (NK) cell activity, are particularly influential, with studies showing that abnormal NK cell activation and imbalances in T and B cell subtypes contribute to immune-mediated miscarriage risk. Successful pregnancy requires a tightly regulated balance between pro-inflammatory and anti-inflammatory immune responses. In the early stages, inflammation supports processes such as trophoblast invasion and spiral artery remodeling, but this must be tempered to prevent immune rejection of the fetus. In this review, we explore the underlying immune mechanisms of RPL, focusing on how dysregulated T, B, and NK cell function disrupts maternal tolerance. Specifically, we discuss the essential role of uterine NK cells in the early stages of vascular remodeling in the decidua and regulate the depth of invasion by extravillous trophoblasts. Furthermore, we focus on the delicate Treg dynamics that enable the maintenance of optimal immune homeostasis, where the balance, and not only the quantity of Tregs, is crucial for fostering maternal-fetal tolerance. Other T cell subpopulations, such as Th1, Th2, and Th17 cells, also contribute to immune imbalance, with Th1 and Th17 cells promoting inflammation and potentially harming fetal tolerance, while Th2 cells support immune tolerance. Finally, we show how changes in B cell subpopulations and their functions have been associated with adverse pregnancy outcomes. We further discuss current therapeutic strategies aimed at correcting these immune imbalances, including intravenous immunoglobulin (IVIg), glucocorticoids, and TNF-α inhibitors, examining their efficacy, challenges, and potential side effects. By highlighting both the therapeutic benefits and limitations of these interventions, we aim to offer a balanced perspective on clinical applications for women facing immune-related causes of RPL.
Collapse
Affiliation(s)
- Cristina Uța
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, “Victor Babeș” University of Medicine and Pharmacy, 2 Eftimie Murgu Square, 300041 Timisoara, Romania; (C.U.); (E.-L.Z.); (R.-I.Z.); (M.G.); (C.P.)
- Timis County Emergency Clinical Hospital “Pius Brinzeu”, 156 Liviu Rebreanu Bd., 300723 Timisoara, Romania
| | - Alexandru Tîrziu
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, “Victor Babeș” University of Medicine and Pharmacy, 2 Eftimie Murgu Square, 300041 Timisoara, Romania; (C.U.); (E.-L.Z.); (R.-I.Z.); (M.G.); (C.P.)
- Department of Functional Sciences, Physiology Discipline, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy Timișoara, 2 Eftimie Murgu Square, 300041 Timişoara, Romania
- Institute of Cardiovascular Diseases Timisoara, 13A Gheorghe Adam Street, 300310 Timisoara, Romania
| | - Elena-Larisa Zimbru
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, “Victor Babeș” University of Medicine and Pharmacy, 2 Eftimie Murgu Square, 300041 Timisoara, Romania; (C.U.); (E.-L.Z.); (R.-I.Z.); (M.G.); (C.P.)
- Department of Functional Sciences, Physiology Discipline, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy Timișoara, 2 Eftimie Murgu Square, 300041 Timişoara, Romania
- Research Center for Gene and Cellular Therapies in the Treatment of Cancer—OncoGen, Timis County Emergency Clinical Hospital “Pius Brinzeu”, 156 Liviu Rebreanu Bd., 300723 Timisoara, Romania
| | - Răzvan-Ionuț Zimbru
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, “Victor Babeș” University of Medicine and Pharmacy, 2 Eftimie Murgu Square, 300041 Timisoara, Romania; (C.U.); (E.-L.Z.); (R.-I.Z.); (M.G.); (C.P.)
- Department of Functional Sciences, Physiology Discipline, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy Timișoara, 2 Eftimie Murgu Square, 300041 Timişoara, Romania
- Research Center for Gene and Cellular Therapies in the Treatment of Cancer—OncoGen, Timis County Emergency Clinical Hospital “Pius Brinzeu”, 156 Liviu Rebreanu Bd., 300723 Timisoara, Romania
| | - Marius Georgescu
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, “Victor Babeș” University of Medicine and Pharmacy, 2 Eftimie Murgu Square, 300041 Timisoara, Romania; (C.U.); (E.-L.Z.); (R.-I.Z.); (M.G.); (C.P.)
- Department of Functional Sciences, Physiology Discipline, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy Timișoara, 2 Eftimie Murgu Square, 300041 Timişoara, Romania
| | - Laura Haidar
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, “Victor Babeș” University of Medicine and Pharmacy, 2 Eftimie Murgu Square, 300041 Timisoara, Romania; (C.U.); (E.-L.Z.); (R.-I.Z.); (M.G.); (C.P.)
- Department of Functional Sciences, Physiology Discipline, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy Timișoara, 2 Eftimie Murgu Square, 300041 Timişoara, Romania
| | - Carmen Panaitescu
- Center of Immuno-Physiology and Biotechnologies, Department of Functional Sciences, “Victor Babeș” University of Medicine and Pharmacy, 2 Eftimie Murgu Square, 300041 Timisoara, Romania; (C.U.); (E.-L.Z.); (R.-I.Z.); (M.G.); (C.P.)
- Department of Functional Sciences, Physiology Discipline, Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy Timișoara, 2 Eftimie Murgu Square, 300041 Timişoara, Romania
| |
Collapse
|
20
|
Fazaeli H, Sheikholeslami A, Ebrahimi Z, Kalhor N, Naserpour L. Comparing intra-uterine injection of mononuclear cells and platelet-rich plasma on the pregnancy rate of women with recurrent implantation failure: An RCT. Int J Reprod Biomed 2024; 22:801-810. [PMID: 39906088 PMCID: PMC11788656 DOI: 10.18502/ijrm.v22i10.17668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/17/2024] [Accepted: 08/26/2024] [Indexed: 02/06/2025] Open
Abstract
Background Recurrent implantation failure (RIF) can be explained mainly by improper crosstalk between the embryo and endometrium. The T-helper1/T-helper2 profile balance influences effective embryo implantation. Endometrial immunomodulation via intrauterine injection of activated peripheral blood mononuclear cells (PBMCs) or autologous platelet-rich plasma (PRP) is a potentially efficient treatment option. Objective This study aims to examine the biochemical and clinical pregnancies resulting from the intrauterine administering of activated PBMCs and PRP in RIF women. Materials and Methods This randomized clinical trial study was done in the Rooya Infertility Treatment Center, Qom, Iran from November 2022 to April 2024. 96 women with at least 2 RIFs were randomized into control, PBMC, and PRP groups. Briefly, 3 ml of blood sample was collected and PBMCs were isolated using Ficoll separation solution, and cultured for 72 hr. PRP was separated from 10 ml of peripheral blood through centrifugation. 2 days before embryo transfer PBMCs or PRP were transferred into the endometrial cavity. Results Except for the duration of infertility, which was higher in the PBMC group, all other baseline characteristics were not statistically different. Moreover, a significantly higher rate of biochemical pregnancy was observed in the PRP (10/32) and PBMC (12/32) groups compared to the control (3/32) (p = 0.027), while the rate of clinical pregnancy was only significantly higher in the PBMC group (10/32) than in the control group (2/32) (p = 0.038). Conclusion Neither PBMC nor PRP interventions exhibited a substantial advantage over one another regarding biochemical and clinical pregnancy rates.
Collapse
Affiliation(s)
- Hoda Fazaeli
- Department of Cell Biology and Regenerative Medicine, Academic Center for Education, Culture, and Research (ACECR), Qom Branch, Qom, Iran
| | - Azar Sheikholeslami
- Department of Cell Biology and Regenerative Medicine, Academic Center for Education, Culture, and Research (ACECR), Qom Branch, Qom, Iran
| | - Zahra Ebrahimi
- Department of Reproductive Biology, Academic Center for Education, Culture, and Research (ACECR), Qom Branch, Qom, Iran
| | - Naser Kalhor
- Department of Cell Biology and Regenerative Medicine, Academic Center for Education, Culture, and Research (ACECR), Qom Branch, Qom, Iran
| | - Leila Naserpour
- Department of Reproductive Biology, Academic Center for Education, Culture, and Research (ACECR), Qom Branch, Qom, Iran
| |
Collapse
|
21
|
Bæk O, Schaltz-Buchholzer F, Campbell A, Amenyogbe N, Campbell J, Aaby P, Benn CS, Kollmann TR. The mark of success: The role of vaccine-induced skin scar formation for BCG and smallpox vaccine-associated clinical benefits. Semin Immunopathol 2024; 46:13. [PMID: 39186134 PMCID: PMC11347488 DOI: 10.1007/s00281-024-01022-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/26/2024] [Indexed: 08/27/2024]
Abstract
Skin scar formation following Bacille Calmette-Guérin (BCG) or smallpox (Vaccinia) vaccination is an established marker of successful vaccination and 'vaccine take'. Potent pathogen-specific (tuberculosis; smallpox) and pathogen-agnostic (protection from diseases unrelated to the intentionally targeted pathogen) effects of BCG and smallpox vaccines hold significant translational potential. Yet despite their use for centuries, how scar formation occurs and how local skin-based events relate to systemic effects that allow these two vaccines to deliver powerful health promoting effects has not yet been determined. We review here what is known about the events occurring in the skin and place this knowledge in the context of the overall impact of these two vaccines on human health with a particular focus on maternal-child health.
Collapse
Affiliation(s)
- Ole Bæk
- University of Copenhagen, Copenhagen, Denmark
| | | | | | - Nelly Amenyogbe
- Telethon Kids Institute, Perth, Australia
- Dalhousie University, 5980 University Ave #5850, 4th floor Goldbloom Pavilion, Halifax, NS, B3K 6R8, Canada
- Bandim Health Project, Bissau, Guinea-Bissau
| | | | - Peter Aaby
- Bandim Health Project, Bissau, Guinea-Bissau
| | - Christine Stabell Benn
- University of Southern Denmark, Copenhagen, Denmark
- Bandim Health Project, Bissau, Guinea-Bissau
| | - Tobias R Kollmann
- Telethon Kids Institute, Perth, Australia.
- Dalhousie University, 5980 University Ave #5850, 4th floor Goldbloom Pavilion, Halifax, NS, B3K 6R8, Canada.
- Bandim Health Project, Bissau, Guinea-Bissau.
| |
Collapse
|
22
|
Zhao SJ, Hu XH, Lin XX, Zhang YJ, Wang J, Wang H, Gong GS, Mor G, Liao AH. IL-27/Blimp-1 axis regulates the differentiation and function of Tim-3+ Tregs during early pregnancy. JCI Insight 2024; 9:e179233. [PMID: 39171524 PMCID: PMC11343602 DOI: 10.1172/jci.insight.179233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 07/11/2024] [Indexed: 08/23/2024] Open
Abstract
Decidual regulatory T cells (Tregs) are essential for successful pregnancy outcome. A subset of Tregs, T cell immunoglobulin and mucin domain-containing protein 3-positive regulatory T cells (TregsTim-3+), plays a central role in the acceptance of the fetus during early stages of normal pregnancy. The molecular mechanism regulating the differentiation and function of TregsTim-3+ is unknown. Here, we investigated the role of the transcription factor B lymphocyte-induced maturation protein 1 (Blimp-1) on decidual TregTim-3+ differentiation. We demonstrated that Blimp-1 enhanced the coexpression of negative costimulatory molecules (Tim-3, T cell immunoreceptor with Ig and ITIM domains, and programmed cell death protein 1) on Tregs and improved their immunosuppressive functions, including increased IL-10 secretion, suppression of effector T cell proliferation, and promotion of macrophage polarization toward the M2 phenotype. Furthermore, we showed that IL-27 regulated the expression of Tim-3 and Blimp-1 through the STAT1 signaling pathway and that transfer of TregsBlimp-1+ into an abortion-prone mouse model effectively reduced embryo absorption rate. We postulated that abnormalities in the IL-27/Blimp-1 axis might be associated with recurrent pregnancy loss (RPL). These findings provided insights for developing more efficient immunotherapies for women with RPL.
Collapse
Affiliation(s)
- Si-Jia Zhao
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-Hui Hu
- Department of Obstetrics and Gynecology, First Clinical College Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xin-Xiu Lin
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-Jing Zhang
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Wang
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huan Wang
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guang-Shun Gong
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gil Mor
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, School of Medicine, Wayne State University, Detroit, Michigan, USA
| | - Ai-Hua Liao
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
23
|
Wekema L, Schoenmakers S, Schenkelaars N, Laskewitz A, Huurman RH, Liu L, Walters L, Harmsen HJM, Steegers-Theunissen RPM, Faas MM. Diet-Induced Obesity in Mice Affects the Maternal Gut Microbiota and Immune Response in Mid-Pregnancy. Int J Mol Sci 2024; 25:9076. [PMID: 39201761 PMCID: PMC11354285 DOI: 10.3390/ijms25169076] [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: 07/19/2024] [Revised: 08/10/2024] [Accepted: 08/17/2024] [Indexed: 09/03/2024] Open
Abstract
Maternal obesity during pregnancy is associated with adverse pregnancy outcomes. This might be due to undesired obesity-induced changes in the maternal gut microbiota and related changes in the maternal immune adaptations during pregnancy. The current study examines how obesity affects gut microbiota and immunity in pregnant obese and lean mice during mid-pregnancy (gestational day 12 (GD12)). C57BL/6 mice were fed a high-fat diet or low-fat diet from 8 weeks before mating and during pregnancy. At GD12, we analyzed the gut microbiota composition in the feces and immune responses in the intestine (Peyer's patches, mesenteric lymph nodes) and the peripheral circulation (spleen and peripheral blood). Maternal obesity reduced beneficial bacteria (e.g., Bifidobacterium and Akkermansia) and changed intestinal and peripheral immune responses (e.g., dendritic cells, Th1/Th2/Th17/Treg axis, monocytes). Numerous correlations were found between obesity-associated bacterial genera and intestinal/peripheral immune anomalies. This study shows that maternal obesity impacts the abundance of specific bacterial gut genera as compared to lean mice and deranges maternal intestinal immune responses that subsequently change peripheral maternal immune responses in mid-pregnancy. Our findings underscore the opportunities for early intervention strategies targeting maternal obesity, ideally starting in the periconceptional period, to mitigate these obesity-related pregnancy effects.
Collapse
Affiliation(s)
- Lieske Wekema
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (A.L.); (R.H.H.)
| | - Sam Schoenmakers
- Department of Obstetrics and Gynaecology, Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (S.S.); (N.S.); (R.P.M.S.-T.)
| | - Nicole Schenkelaars
- Department of Obstetrics and Gynaecology, Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (S.S.); (N.S.); (R.P.M.S.-T.)
| | - Anne Laskewitz
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (A.L.); (R.H.H.)
| | - Romy H. Huurman
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (A.L.); (R.H.H.)
| | - Lei Liu
- Department of Medical Microbiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (L.L.); (L.W.); (H.J.M.H.)
| | - Lisa Walters
- Department of Medical Microbiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (L.L.); (L.W.); (H.J.M.H.)
| | - Hermie J. M. Harmsen
- Department of Medical Microbiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (L.L.); (L.W.); (H.J.M.H.)
| | - Régine P. M. Steegers-Theunissen
- Department of Obstetrics and Gynaecology, Erasmus Medical Center, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands; (S.S.); (N.S.); (R.P.M.S.-T.)
| | - Marijke M. Faas
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (A.L.); (R.H.H.)
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
24
|
Wekema L, Schoenmakers S, Schenkelaars N, Laskewitz A, Liu L, Walters L, Harmsen HJM, Steegers-Theunissen RPM, Faas MM. Obesity and diet independently affect maternal immunity, maternal gut microbiota and pregnancy outcome in mice. Front Immunol 2024; 15:1376583. [PMID: 39072322 PMCID: PMC11272480 DOI: 10.3389/fimmu.2024.1376583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 06/24/2024] [Indexed: 07/30/2024] Open
Abstract
Introduction Maternal obesity poses risks for both mother and offspring during pregnancy, with underlying mechanisms remaining largely unexplored. Obesity is associated with microbial gut dysbiosis and low-grade inflammation, and also the diet has a major impact on these parameters. This study aimed to investigate how maternal obesity and diet contribute to changes in immune responses, exploring potential associations with gut microbiota dysbiosis and adverse pregnancy outcomes in mice. Methods Before mating, C57BL/6 mice were assigned to either a high-fat-diet (HFD) or low-fat-diet (LFD) to obtain obese (n=17) and lean (n=10) mice. To distinguish between the effects of obesity and diet, 7 obese mice were switched from the HFD to the LFD from day 7 until day 18 of pregnancy ("switch group"), which was the endpoint of the study. T helper (Th) cell subsets were studied in the spleen, mesenteric lymph nodes (MLN) and Peyer's patches (PP), while monocyte subsets and activation status were determined in maternal blood (flow cytometry). Feces were collected before and during pregnancy (day 7,14,18) for microbiota analysis (16S rRNA sequencing). Pregnancy outcome included determination of fetal and placental weight. Results Obesity increased splenic Th1 and regulatory T cells, MLN Th1 and PP Th17 cells and enhanced IFN-γ and IL-17A production by splenic Th cells upon ex vivo stimulation. Switching diet decreased splenic and PP Th2 cells and classical monocytes, increased intermediate monocytes and activation of intermediate/nonclassical monocytes. Obesity and diet independently induced changes in the gut microbiota. Various bacterial genera were increased or decreased by obesity or the diet switch. These changes correlated with the immunological changes. Fetal weight was lower in the obese than the lean group, while placental weight was lower in the switch than the obese group. Discussion This study demonstrates that obesity and diet independently impact peripheral and intestinal immune responses at the end of pregnancy. Simultaneously, both factors affect specific bacterial gut genera and lead to reduced fetal or placental weight. Our data suggest that switching diet during pregnancy to improve maternal health is not advisable and it supports pre/probiotic treatment of maternal obesity-induced gut dysbiosis to improve maternal immune responses and pregnancy outcome.
Collapse
Affiliation(s)
- Lieske Wekema
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Sam Schoenmakers
- Department of Obstetrics and Gynaecology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Nicole Schenkelaars
- Department of Obstetrics and Gynaecology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Anne Laskewitz
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Lei Liu
- Department of Medical Microbiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Lisa Walters
- Department of Medical Microbiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Hermie J. M. Harmsen
- Department of Medical Microbiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | | | - Marijke M. Faas
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
25
|
Huang J, Zhu Q, Wang B, Wang H, Xie Z, Zhu X, Zhao T, Yang Z. Antiphospholipid antibodies and the risk of adverse pregnancy outcomes in patients with systemic lupus erythematosus: a systematic review and meta-analysis. Expert Rev Clin Immunol 2024; 20:793-801. [PMID: 38445835 DOI: 10.1080/1744666x.2024.2324005] [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: 08/18/2023] [Accepted: 02/12/2024] [Indexed: 03/07/2024]
Abstract
OBJECTIVE This article aims to evaluate the magnitude of adverse pregnancy outcomes (APOs) risks associated with different antiphospholipid antibody (aPL) profiles in women with systemic lupus erythematosus (SLE). METHODS Multiple databases were investigated to identify articles that explored the relationship between aPLs and APOs in SLE patients. A random effects model was used for calculating pooled odds ratios (OR). Stata version 15.0 was utilized to conduct the meta-analysis. RESULTS There were 5234 patients involved in 30 studies. Overall aPL was linked to an increased incidence of any kind of APOs, fetal loss, and preterm birth. Any kind of APOs and preterm delivery were more common in patients with lupus anticoagulant (LA) positive. Anticardiolipin antibody (aCL) was associated with an increased risk of any kind of APOs and fetal loss. The association between aCL-IgM and fetal loss was also significant. Patients with anti-beta2-glycoprotein1 antibody (antiβ2GP1) positivity had an increased risk of fetal loss. CONCLUSIONS Both LA and aCL were risk factors of APOs in patients with SLE. Not only ACL, particularly aCL-IgM, but antiβ2GP1 were associated with an increased risk of fetal loss, while LA appeared to indicate the risk of preterm birth.PROSPERO (CRD42023388122).
Collapse
Affiliation(s)
- Jinge Huang
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qingmiao Zhu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Baizhou Wang
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Hanzheng Wang
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhijun Xie
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xingyu Zhu
- Department of Nephrology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Ting Zhao
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zi Yang
- The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
- Teaching Faculty, New Zealand College of Chinese Medicine, Greenlane, New Zealand
| |
Collapse
|
26
|
Bai S, Xu G, Mo H, Qi T, Fu S, Zhu L, Huang B, Zhang J, Chen H. Investigating into microbiota in the uterine cavity of the unexplained recurrent pregnancy loss patients in early pregnancy. Placenta 2024; 152:1-8. [PMID: 38729066 DOI: 10.1016/j.placenta.2024.05.125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/25/2024] [Accepted: 05/06/2024] [Indexed: 05/12/2024]
Abstract
INTRODUCTION The majority of unexplained recurrent pregnancy loss (URPL) cases have been attributed to immune abnormalities. Inappropriate changes in microbiota could lead to immune disorders. However, the specific role of uterine cavity microbiota in URPL remains unclear, and only a limited number of related studies are available for reference. METHODS We utilized double-lumen embryo transfer tubes to collect uterine cavity fluid samples from pregnant women in their first trimester. Subsequently, we conducted 16S rRNA sequencing to analyze the composition and abundance of the microbiota in these samples. RESULTS For this study, we enlisted 10 cases of URPL and 28 cases of induced miscarriages during early pregnancy. Microbial communities were detected in all samples of the URPL group (100 %, n = 10), whereas none were found in the control group (0 %, n = 28). Among the identified microbes, Lactobacillus and Curvibacter were the two most dominant species. The abundance of Curvibacter is correlated with the number of NK cells in peripheral blood (r = -0.759, P = 0.018). DISCUSSION This study revealed that during early pregnancy, Lactobacillus and Curvibacter were the predominant colonizers in the uterine cavity of URPL patients and were associated with URPL. Consequently, alterations in the dominant microbiota may lead to adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Shiyu Bai
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China; Center for Reproductive Genetics and Reproductive Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China.
| | - Guocai Xu
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China; Center for Reproductive Genetics and Reproductive Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China.
| | - Hanjie Mo
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China; Center for Reproductive Genetics and Reproductive Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China.
| | - Tianyuan Qi
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China; Center for Reproductive Genetics and Reproductive Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China.
| | - Shuai Fu
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China.
| | - Liqiong Zhu
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China.
| | - Bingqian Huang
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China; Center for Reproductive Genetics and Reproductive Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China.
| | - Jianping Zhang
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China.
| | - Hui Chen
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China; Department of Genetics and Cell Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China.
| |
Collapse
|
27
|
Wang J, Nuray U, Yan H, Xu Y, Fang L, Li R, Zhou X, Zhang H. Pyroptosis is involved in the immune microenvironment regulation of unexplained recurrent miscarriage. Mamm Genome 2024; 35:256-279. [PMID: 38538990 DOI: 10.1007/s00335-024-10038-3] [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: 08/22/2023] [Accepted: 03/11/2024] [Indexed: 05/29/2024]
Abstract
Unexplained recurrent miscarriage (URM) is a common pregnancy complication with few effective therapies. Moreover, little is known regarding the role of pyroptosis in the regulation of the URM immune microenvironment. To address this issue, gene expression profiles of publicly available placental datasets GSE22490 and GSE76862 were downloaded from the Gene Expression Omnibus database. Pyroptosis-related differentially expressed genes were identified and a total of 16 differentially expressed genes associated with pyroptosis were detected, among which 1 was upregulated and 15 were downregulated. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses indicated that the functionally enriched modules and pathways of these genes are closely related to immune and inflammatory responses. Four hub genes were identified: BTK, TLR8, NLRC4, and TNFSF13B. BTK, TLR8, and TNFSF13B were highly connected with immune cells, according to the correlation analysis of four hub genes and 20 different types of immune cells (p < 0.05). The four hub genes were used as research objects to construct the interaction networks. Chorionic villus tissue was used for quantitative real-time polymerase chain reaction and western blot to confirm the expression levels of hub genes, and the results showed that the expression of the four hub genes was significantly decreased in the chorionic villus tissue in the URM group. Collectively, the present study indicates that perhaps pyroptosis is essential to the diversity and complexity of the URM immune microenvironment, and provides a theoretical basis and research ideas for subsequent target gene verification and mechanism research.
Collapse
Affiliation(s)
- Jing Wang
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | | | - Hongchao Yan
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yang Xu
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Lisha Fang
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Ranran Li
- First clinical medical college of Xuzhou Medical University, Xuzhou, China
| | - Xin Zhou
- First clinical medical college of Xuzhou Medical University, Xuzhou, China
| | - Hong Zhang
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
28
|
Mills BS, Bermas BL. Pregnancy and the Autoimmune Patient. Curr Allergy Asthma Rep 2024; 24:261-267. [PMID: 38563848 DOI: 10.1007/s11882-024-01143-z] [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] [Accepted: 03/15/2024] [Indexed: 04/04/2024]
Abstract
PURPOSE OF REVIEW This article will review the current understanding of the immunologic changes that occur during pregnancy. It will discuss the impact of pregnancy on the disease activity of autoimmune or inflammatory rheumatic diseases (AIRD). Lastly, it will highlight the most recent data on pre-conception and pregnancy management practices that can improve pregnancy outcomes in autoimmune patients. RECENT FINDINGS Pregnancy is an immunologically complex and dynamic state that may affect the activity of AIRDs, with more patients having active disease during pregnancy than previously thought. Uncontrolled inflammatory diseases are associated with poor pregnancy outcomes such as preeclampsia, small for gestational age infants, and prematurity. Pre-conception counseling and early pregnancy planning discussions can help ensure optimal disease control and medication management prior to attempting conception. Adequate control of AIRDs on pregnancy-compatible medications during the pre-conception, pregnancy, and postpartum periods is required for optimal pregnancy outcomes.
Collapse
Affiliation(s)
- Brooke S Mills
- University of Texas Southwestern Medical Center, 2001 Inwood Road, Dallas, TX, 75390, USA
| | - Bonnie L Bermas
- University of Texas Southwestern Medical Center, 2001 Inwood Road, Dallas, TX, 75390, USA.
| |
Collapse
|
29
|
Fernandes KA, Lim AI. Maternal-driven immune education in offspring. Immunol Rev 2024; 323:288-302. [PMID: 38445769 DOI: 10.1111/imr.13315] [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] [Indexed: 03/07/2024]
Abstract
Maternal environmental exposures, particularly during gestation and lactation, significantly influence the immunological development and long-term immunity of offspring. Mammalian immune systems develop through crucial inputs from the environment, beginning in utero and continuing after birth. These critical developmental windows are essential for proper immune system development and, once closed, may not be reopened. This review focuses on the mechanisms by which maternal exposures, particularly to pathogens, diet, and microbiota, impact offspring immunity. Mechanisms driving maternal-offspring immune crosstalk include transfer of maternal antibodies, changes in the maternal microbiome and microbiota-derived metabolites, and transfer of immune cells and cytokines via the placenta and breastfeeding. We further discuss the role of transient maternal infections, which are common during pregnancy, in providing tissue-specific immune education to offspring. We propose a "maternal-driven immune education" hypothesis, which suggests that offspring can use maternal encounters that occur during a critical developmental window to develop optimal immune fitness against infection and inflammation.
Collapse
Affiliation(s)
| | - Ai Ing Lim
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| |
Collapse
|
30
|
Zhang L, Bi S, Gong J, Wang X, Liang J, Gu S, Su M, Wang W, Sun M, Chen J, Zheng W, Wu J, Wang Z, Liu J, Li HT, Chen D, Du L. Comparing the severity of second preeclampsia with first preeclampsia: a multicenter retrospective longitudinal cohort study. J Hypertens 2024; 42:841-847. [PMID: 38164966 DOI: 10.1097/hjh.0000000000003642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
OBJECTIVE Compare the clinical severity of second preeclampsia with the first preeclampsia. METHODS This retrospective longitudinal cohort study was conducted in three teaching hospitals in Guangzhou, where there were a total of 296 405 deliveries between 2010 and 2021. Two consecutive singleton deliveries complicated with preeclampsia were included. Clinical features, laboratory results within 1 week before delivery, and maternal and neonatal outcomes of both deliveries were collected. Univariate analyses were made using paired Wilcoxon tests and McNemar tests. Multivariable logistic regression and generalized linear models were performed to assess the association of adverse maternal and neonatal outcomes with second preeclampsia. RESULTS A total of 151 women were included in the study. The mean maternal age was 28 and 33 years for the first and second deliveries, respectively. The proportion of preventive acetylsalicylic acid use was 4.6% for the first delivery and 15.2% for the second delivery. No significant differences were observed in terms of blood pressure on admission, gestational weeks of admission and delivery, application of perinatal antihypertensive agents, rates of preterm delivery, and severe features between the two occurrences. However, the rates of heart disease, edema, and admission to the ICU were lower, and hospital stays were shorter in the second preeclampsia compared with the first preeclampsia. Sensitivity analysis conducted among women who did not use preventive acetylsalicylic acid yielded similar results. After adjusting for potential confounding variables, the occurrence of second preeclampsia was associated with significantly decreased risks of heart disease, edema, complications, and admission to the NICU, with odds ratios ranging between 0.157 and 0.336. CONCLUSION Contrary to expectations, the second preeclampsia did not exhibit worse manifestations or outcomes to the first occurrence. In fact, some clinical features and outcomes appeared to be better in the second preeclampsia.
Collapse
Affiliation(s)
- Lizi Zhang
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology; Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine; The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou
| | - Shilei Bi
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology; Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine; The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou
| | - Jingjin Gong
- Guangzhou Panyu District Maternal and Child Health Hospital
| | - Xinghe Wang
- Dongguan Maternal and Children Health Hospital
| | - Jingying Liang
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology; Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine; The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou
| | - Shifeng Gu
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology; Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine; The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou
| | - Minglian Su
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology; Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine; The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou
| | - Weiwei Wang
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology; Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine; The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou
| | - Manna Sun
- Dongguan Maternal and Children Health Hospital
| | - Jingsi Chen
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology; Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine; The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou
| | - Weitan Zheng
- Guangzhou Panyu District Maternal and Child Health Hospital
| | - Junwei Wu
- Guangzhou Panyu District Maternal and Child Health Hospital
| | - Zhijian Wang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jianmeng Liu
- Institute of Reproductive and Child Health, National Health Commission Key Laboratory of Reproductive Health, Peking University Health Science Center, Beijing
| | - Hong-Tian Li
- Institute of Reproductive and Child Health, National Health Commission Key Laboratory of Reproductive Health, Peking University Health Science Center, Beijing
| | - Dunjin Chen
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology; Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine; The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou
| | - Lili Du
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology; Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine; The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou
| |
Collapse
|
31
|
Yang K, Zeng L, Li Y, Wu L, Xiang W, Wu X, Wang G, Bao T, Huang S, Yu R, Zhang G, Liu H. Uncovering the pharmacological mechanism of Shou Tai Wan on recurrent spontaneous abortion: A integrated pharmacology strategy-based research. JOURNAL OF ETHNOPHARMACOLOGY 2024; 323:117589. [PMID: 38104875 DOI: 10.1016/j.jep.2023.117589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 12/04/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shou Tai Wan (STW), a traditional Chinese medicine formula, has been historically used for the treatment of recurrent spontaneous abortion (RSA). Despite its long-standing usage, the exact mechanism underlying the therapeutic effects of STW remains unclear in the existing literature. AIMS OF THIS STUDY To explore the Pharmacological Mechanism of STW on RSA. METHODS A network pharmacological methodology was utilized to predict the active compounds and potential targets of STW, collect the RSA targets and other human proteins of STW, and analyze the STW related networks. The animal experiments were also performed to validate the effect of STW on RSA. RESULTS The results of network analysis showed that STW may regulate PI3K/AKT, MAPK, FoxO signaling pathways and so on. Animal experiment established the RSA model with CBA/J × DBA/2 mice. It was found that STW can reduce the embryo absorption rate of RSA group (p < 0.05) and balance the expression of Th 1/Th2 type cytokines compared with the model group. After 14 days of administration, the decidual and placental tissues were taken and the CD4+ T cells were isolated, and the phosphorylation level of signaling pathway was detected by Springbio720 antibody microarray. This experiment found that STW can significantly up-regulate the phosphorylation levels of STAT3 and STAT6 proteins in the STAT signaling pathway, and down-regulating the phosphorylation level of STAT1 protein. STW also significantly up-regulated the phosphorylation levels of Raf1, A-Raf, Ask1, Mek1, Mek2, JKK1, ERK1, ERK2, c-fos, c-Jun and CREB proteins in the MAPK signaling pathway, and down-regulate the phosphorylation levels of MEK6 and IKKb proteins. Compared with the RSA group, the STW group increased the expression levels of ERK1/2 mRNA and proteins and p-ERK1/2 proteins, and there was a statistical difference (p < 0.05). This is consistent with the chip results. CONCLUSION STW may achieve therapeutic effects by interfering with the signaling pathways, biological processes and targets discovered in this study. It provides a new perspective for revealing the immunological mechanism of STW in the treatment of RSA, and also provides a theoretical basis for the clinical use of STW in the treatment of RSA.
Collapse
Affiliation(s)
- Kailin Yang
- Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Liuting Zeng
- Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Yuwei Li
- Hunan University of Science and Technology, Xiangtan, China
| | - Lingyu Wu
- Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Wang Xiang
- The First People's Hospital Changde City, Changde City, China
| | - Xiaolan Wu
- Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Guiyun Wang
- Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Tingting Bao
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5 BeiXianGe Street, Xicheng District, Beijing 100053, China
| | - Shanshan Huang
- Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Rong Yu
- Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Guomin Zhang
- Hunan University of Chinese Medicine, Changsha, Hunan Province, China.
| | - Huiping Liu
- Hunan University of Chinese Medicine, Changsha, Hunan Province, China.
| |
Collapse
|
32
|
He YB, Han L, Wang C, Fang J, Shang Y, Cai HL, Zhou Q, Zhang ZZ, Chen SL, Li JY, Liu YL. Bulk RNA-sequencing, single-cell RNA-sequencing analysis, and experimental validation reveal iron metabolism-related genes CISD2 and CYP17A1 are potential diagnostic markers for recurrent pregnancy loss. Gene 2024; 901:148168. [PMID: 38244949 DOI: 10.1016/j.gene.2024.148168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/06/2024] [Accepted: 01/13/2024] [Indexed: 01/22/2024]
Abstract
BACKGROUND Recurrent pregnancy loss (RPL) is associated with variable causes. Its etiology remains unexplained in about half of the cases, with no effective treatment available. Individuals with RPL have an irregular iron metabolism. In the present study, we identified key genes impacting iron metabolism that could be used for diagnosing and treating RPL. METHODS We obtained gene expression profiles from the Gene Expression Omnibus (GEO) database. The Molecular Signatures Database was used to identify 14 gene sets related to iron metabolism, comprising 520 iron metabolism genes. Differential analysis and a weighted gene co-expression network analysis (WGCNA) of gene expression revealed two iron metabolism-related hub genes. Reverse transcriptase-polymerase chain reaction (RT-PCR) and immunohistochemistry were used on clinical samples to confirm our results. The receiver operating characteristic (ROC) analysis and immune infiltration analysis were conducted. In addition, we analyzed the distribution of genes and performed CellChat analysis by single-cell RNA sequencing. RESULTS The expression of two hub genes, namely, CDGSH iron sulfur domain 2 (CISD2)and Cytochrome P450 family 17 subfamily A member 1 (CYP17A1), were reduced in RPL, as verified by both qPCR and immunohistochemistry. The Gene Ontology (GO) analysis revealed the genes predominantly engaged in autophagy and iron metabolism. The area under the curve (AUC) demonstrated better diagnostic performance for RPL using CISD2 and CYP17A1. The single-cell transcriptomic analysis of RPL demonstrated that CISD2 is expressed in the majority of cell subpopulations, whereas CYP17A1 is not. The cell cycle analysis revealed highly active natural killer (NK) cells that displayed the highest communications with other cells, including the strongest interaction with macrophages through the migratory inhibitory factor (MIF) pathway. CONCLUSIONS Our study suggested that CISD2 and CYP17A1 genes are involved in abnormal iron metabolism, thereby contributing to RPL. These genes could be used as potential diagnostic and therapeutic markers for RPL.
Collapse
Affiliation(s)
- Yi-Bo He
- Department of Clinical Lab, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China
| | - Lu Han
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Guiyang, Guizhou Province, China
| | - Cong Wang
- Department of Clinical Lab, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China
| | - Ju Fang
- Reproductive Center, Hainan Branch, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Sanya, Hainan Province, China
| | - Yue Shang
- Reproductive Center, Hainan Branch, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Sanya, Hainan Province, China
| | - Hua-Lei Cai
- Department of Emergency Obstetrics and Gynecology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Qun Zhou
- Obstetrics and Gynecology, First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China
| | - Zhe-Zhong Zhang
- Department of Clinical Lab, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China
| | - Shi-Liang Chen
- Department of Clinical Lab, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang Province, China.
| | - Jun-Yu Li
- Pharmacy Department, Hainan Branch, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Sanya, Hainan Province, China.
| | - Yong-Lin Liu
- Reproductive Center, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
33
|
Ali Khazaei H, Farzaneh F, Sarhadi S, Dehghan Haghighi J, Forghani F, Sheikhi V, Khazaei B, Asadollahi L. Comparison of serum levels of interleukin 33 in combination with serum levels of C-reactive protein, Immunoglobulin G, Immunoglobulin A, and Immunoglobulin M in recurrent pregnancy loss: A case-control study. Int J Reprod Biomed 2024; 22:317-322. [PMID: 39035635 PMCID: PMC11255461 DOI: 10.18502/ijrm.v22i4.16392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 06/19/2023] [Accepted: 08/12/2023] [Indexed: 07/23/2024] Open
Abstract
Background One of the critical cases of recurrent pregnancy loss is immunological factors, whereas obtaining effective prevention or treatment is necessary for cognition of reasons. Objective In this study, we tried to evaluate some immunological factors related to recurrent pregnancy loss. Materials and Methods This case-control study was conducted on 66 women at the age of 18-35 yr who were referred to the Clinic of Gynecology and Obstetrics, Ali Ibn Abi Taleb hospital, Zahedan, Iran, from August-December 2019. Interleukin 33 (IL-33) serum levels were measured using enzyme-linked immunosorbent assay. Immunoglobulin G, Immunoglobulin A, Immunoglobulin M (IgM), and C-reactive protein levels were measured by serology and hematology methods. Results The mean age of total participants was 30.8 ± 3.80 yr. The mean serum IL-33 in the case group was 318.5 ± 254.1 pg/ml and was lower than the control group (354.2 ± 259.9 pg/ml), which was not statistically significant (p = 0.52). The level of C-reactive protein in the case and control was not significantly different (p = 0.27), and Immunoglobulin A and Immunoglobulin G in the case and control were also not significantly different (p = 0.46, and p = 0.16, respectively), but there were significant differences (p = 0.003) between the level of the IgM in the case and control groups. Conclusion No statistically significant difference was observed in the IL-33 serum level, for at least 4-6 months after the last abortion in the case group and the final live birth in the control group. In contrast, serum levels of IgM were statistically significant. Finally, the need for more studies is felt according to the different results of the previous studies in this field.
Collapse
Affiliation(s)
- Hossein Ali Khazaei
- Clinical Immunology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Farahnaz Farzaneh
- Department of Obstetrics and Gynecology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Saeedeh Sarhadi
- Health Promotion Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
- Department of Community Medicine, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Javid Dehghan Haghighi
- Department of Community Medicine, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Forough Forghani
- Department of Obstetrics and Gynecology, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Vahid Sheikhi
- Department of Pediatrics, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Bahman Khazaei
- School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Lida Asadollahi
- School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|
34
|
Zhu J, Chen Y, Chen H, Sun Y, Yan L, Zhu M, Chen L, Wang Q, Zhang J. Comparison of microbial abundance and diversity in uterine and peritoneal fluid in infertile patients with or without endometriosis. BMC Womens Health 2024; 24:148. [PMID: 38424540 PMCID: PMC10903057 DOI: 10.1186/s12905-024-02985-5] [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: 09/27/2023] [Accepted: 02/19/2024] [Indexed: 03/02/2024] Open
Abstract
INTRODUCTION Endometriosis (EM) is a multifactorial disease that affects 10 - 15% of women of reproductive age. Additionally, 30-50% of women with EM suffer from infertility. The mechanism of infertility caused by EM has not yet been consistently explained. In recent years, studies have shown a link between infertility associated with EM and changes in the reproductive tract microbiota. METHODS In this study, we involved 26 EM patients (8 cases of stage I-II and 18 cases of stage III-IV) and 31 control subjects who were tubal obstruction-related infertility (TORI). The samples from peritoneal fluid (PF) and uterine fluid (UF) were collected and sequenced by 16 S rRNA amplicon. RESULTS In the comparison of microbial diversity, we found no significant differences in the microbial diversity of PF and UF between patients with stage I-II EM and those with TORI. However, there was a significant difference in microbial diversity among patients with stage III-IV EM compared to the previous two groups. Lactobacillus decreased in PF of EM compared to the control group, while it increased in UF. In PF, the abundance of Pseudomonas, Enterococcus, Dubosiella and Klebsiella was significantly higher in patients with stage III-IV compared to TORI patients. And in UF, the main differences existed between stage I-II EM compared to the other two groups. The abundance of pontibacter, aquabacterium, Rikenellaceae and so on at the genus level was significantly enriched in the EM patients with stage I-II. In the analysis based on KEGG database, EM may affect the receptivity related pathways of the endometrium by influencing changes in the uterine microbiota. CONCLUSION Our results indicated that as EM progresses, the microorganisms in UF and PF keep changing. These changes in the microbiota, as well as the resulting alternations in gene functional classification, may play an important role in the infertility associated with EM.
Collapse
Affiliation(s)
- Jue Zhu
- Department of Gynecology, Ningbo Women and Children's Hospital, #339 Liuting Road, Ningbo, Zhejiang, China
| | - Yichen Chen
- Department of Basic Research Laboratory, Ningbo Women and Children's Hospital, Ningbo, Zhejiang, China
| | - Huan Chen
- Department of Medicine, Ningbo University, Zhejiang, China
| | - Yuhui Sun
- Department of Basic Research Laboratory, Ningbo Women and Children's Hospital, Ningbo, Zhejiang, China
| | - Lifeng Yan
- Department of Gynecology, Ningbo Women and Children's Hospital, #339 Liuting Road, Ningbo, Zhejiang, China
| | - Miaohua Zhu
- Department of Gynecology, Ningbo Women and Children's Hospital, #339 Liuting Road, Ningbo, Zhejiang, China
| | - Liang Chen
- Department of Gynecology, Ningbo Women and Children's Hospital, #339 Liuting Road, Ningbo, Zhejiang, China
| | - Qiming Wang
- Department of Gynecology, Ningbo Women and Children's Hospital, #339 Liuting Road, Ningbo, Zhejiang, China
| | - Jing Zhang
- Department of Gynecology, Ningbo Women and Children's Hospital, #339 Liuting Road, Ningbo, Zhejiang, China.
| |
Collapse
|
35
|
Brummaier T, Rinchai D, Toufiq M, Karim MY, Habib T, Utzinger J, Paris DH, McGready R, Marr AK, Kino T, Terranegra A, Al Khodor S, Chaussabel D, Syed Ahamed Kabeer B. Design of a targeted blood transcriptional panel for monitoring immunological changes accompanying pregnancy. Front Immunol 2024; 15:1319949. [PMID: 38352867 PMCID: PMC10861739 DOI: 10.3389/fimmu.2024.1319949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 01/11/2024] [Indexed: 02/16/2024] Open
Abstract
Background Immunomodulatory processes exert steering functions throughout pregnancy. Detecting diversions from this physiologic immune clock may help identify pregnant women at risk for pregnancy-associated complications. We present results from a data-driven selection process to develop a targeted panel of mRNAs that may prove effective in detecting pregnancies diverting from the norm. Methods Based on a de novo dataset from a resource-constrained setting and a dataset from a resource-rich area readily available in the public domain, whole blood gene expression profiles of uneventful pregnancies were captured at multiple time points during pregnancy. BloodGen3, a fixed blood transcriptional module repertoire, was employed to analyze and visualize gene expression patterns in the two datasets. Differentially expressed genes were identified by comparing their abundance to non-pregnant postpartum controls. The selection process for a targeted gene panel considered (i) transcript abundance in whole blood; (ii) degree of correlation with the BloodGen3 module; and (iii) pregnancy biology. Results We identified 176 transcripts that were complemented with eight housekeeping genes. Changes in transcript abundance were seen in the early stages of pregnancy and similar patterns were observed in both datasets. Functional gene annotation suggested significant changes in the lymphoid, prostaglandin and inflammation-associated compartments, when compared to the postpartum controls. Conclusion The gene panel presented here holds promise for the development of predictive, targeted, transcriptional profiling assays. Such assays might become useful for monitoring of pregnant women, specifically to detect potential adverse events early. Prospective validation of this targeted assay, in-depth investigation of functional annotations of differentially expressed genes, and assessment of common pregnancy-associated complications with the aim to identify these early in pregnancy to improve pregnancy outcomes are the next steps.
Collapse
Affiliation(s)
- Tobias Brummaier
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Darawan Rinchai
- Research Department, Sidra Medicine, Doha, Qatar
- Department of Infectious Diseases, St. Jude Children Research Hospital, Memphis, TN, United States
| | | | | | - Tanwir Habib
- Research Department, Sidra Medicine, Doha, Qatar
- Bioinformatics Core, Weill Cornell Medicine-Qatar, Education City, Doha, Qatar
| | - Jürg Utzinger
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Daniel H. Paris
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Rose McGready
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | | | | | | | | | - Damien Chaussabel
- Research Department, Sidra Medicine, Doha, Qatar
- Computational Sciences Department, The Jackson Laboratory, Farmington, CT, United States
| | | |
Collapse
|
36
|
Guo D, Diao Z, Wang K, Pang C. Causal association between rheumatoid arthritis and pregnancy loss and intrauterine growth retardation: A bidirectional two-sample Mendelian randomization study. Medicine (Baltimore) 2024; 103:e36873. [PMID: 38215086 PMCID: PMC10783369 DOI: 10.1097/md.0000000000036873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 12/15/2023] [Indexed: 01/14/2024] Open
Abstract
OBJECTIVE To investigate the causal relationship between rheumatoid arthritis (RA) and pregnancy loss and intrauterine growth retardation (IUGR) using Mendelian randomization (MR). METHODS Genetic variants associated with RA (12,555 cases and 240,862 controls), miscarriage (1475 cases and 149,622 controls), and IUGR (3558 cases and 207,312 controls) were obtained from the FinnGen consortium, and supplementary data on RA (5201 cases and 457,732 controls) and miscarriage (7069 cases and 250,492 controls) were obtained from the Medical Research Council Integrated Epidemiology Unit (MRC-IEU). 47 Single nucleotide polymorphisms (SNPs) associated with RA were screened as instrumental variables (IV). The causal relationship between RA and pregnancy loss and IUGR were assessed by 5 MR methods, mainly inverse variance weighting (IVW). Sensitivity analyses were also performed to test the stability of the results. RESULTS Bidirectional MR showed that genetically predicted RA was causally associated with pregnancy loss and IUGR in forward MR analyses, and that RA significantly increased pregnancy loss [odds ratio (OR) = 1.13, 95% confidence interval (CI): 1.00-1.33, P = .03] and IUGR (OR = 1.08, 95% CI: 1.01-1.15, P = .019). In the reverse MR, there was no causal association between pregnancy loss (P = .15) and IUGR (P = .87) and RA. CONCLUSION This study found a significant genetic association between RA and pregnancy loss and IUGR. RA is considered to be a high-risk factor for adverse maternal outcomes. Pre-pregnancy prophylaxis and intra-pregnancy control of patients should be emphasized to reduce the incidence of adverse pregnancy outcomes such as pregnancy loss and IUGR.
Collapse
Affiliation(s)
- Danyang Guo
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhihao Diao
- School of Acupuncture and Tuina, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Kehua Wang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Conghui Pang
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
37
|
Liu Z, Song Y, Hu R, Geng Y, Huang Y, Li F, Ma W, Dong H, Song K, Ding J, Xu X, Wu X, Zhang M, Zhong Z. Bushen Antai recipe ameliorates immune microenvironment and maternal-fetal vascularization in STAT3-deficient abortion-prone mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116889. [PMID: 37423519 DOI: 10.1016/j.jep.2023.116889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/05/2023] [Accepted: 07/07/2023] [Indexed: 07/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Spontaneous abortion (SA) is an intricate disorder affecting women of reproductive age. Previous studies have confirmed the indispensable role of signal transducer and activator of transcription (STAT) 3 in normal pregnancy. Bushen Antai recipe (BAR) is a satisfactory formula commonly used in practice, based on the rationale of traditional Chinese medicine (TCM) for SA. AIM OF THE STUDY The current study explores the potential therapeutic effects and mechanistic insights of BAR in STAT3-deficient abortion-prone mice. MATERIALS AND METHODS A STAT3-deficient abortion-prone mouse model was developed using intraperitoneal injection of stattic from embryo day (ED) 5.5 to ED9.5 among pregnant females (C57BL/6). We separately administered BAR1 (5.7 g/kg), BAR2 (11.4 g/kg), progesterone (P4), or distilled water at 10 ml/kg/day from ED0.5 until ED10.5. The embryo resorption rate and placenta-uterus structure were observed on ED10.5. The systemic immune status was examined by analyzing the frequency of immunosuppressive myeloid-derived suppressor cells (MDSCs), the ratio of two macrophage (M) subtypes, and the protein expression of associated molecules. Morphological observation, immunohistochemistry, and western blotting were used to evaluate the vascularization conditions at the maternal-fetal interface. RESULTS BAR1, BAR2, or P4 treatment exerted remarkable effects in alleviating embryo resorption rate and disordered placental-uterus structure in STAT3-deficient abortion-prone mice. Western blotting indicated the deficiency of phosphorylated STAT3 and two prime target molecules, PR and HIF-1α, at the maternal-fetal interface under STAT3 inhibition. Simultaneously, BAR2 treatment significantly upregulated their expression levels. The systemic immune environment was disrupted, indicated by the reduced serum cytokine concentrations, MDSCs frequency, M2/M1 ratio, and the expression of immunomodulatory factors. Nonetheless, BAR2 or P4 treatment revived the immune tolerance for semi-allogenic embryos by enhancing the immune cells and factors. Besides, the western blot and immunohistochemistry results revealed that BAR2 or P4 treatment upregulated VEGFA/FGF2 and activated ERK/AKT phosphorylation. Therefore, BAR2 or P4 facilitated vascularization at the maternal-fetal interface in STAT3-deficient abortion-prone mice. CONCLUSIONS BAR sustained pregnancy by reviving the systemic immune environment and promoting angiogenesis at the maternal-fetal interface in STAT3-deficient abortion-prone mice.
Collapse
Affiliation(s)
- Zhuo Liu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yufan Song
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Runan Hu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yuli Geng
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yanjing Huang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Fan Li
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Wenwen Ma
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Haoxu Dong
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Kunkun Song
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jiahui Ding
- Department of Obstetrics and Gynecology, School of Medicine, Wayne State University, Detroit, MI, USA.
| | - Xiaohu Xu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xiao Wu
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Mingmin Zhang
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Zhiyan Zhong
- Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
38
|
Zhou W, Chen Y, Zheng Y, Bai Y, Yin J, Wu XX, Hong M, Liang L, Zhang J, Gao Y, Sun N, Li J, Zhang Y, Wu L, Jin X, Niu J. Characterizing immune variation and diagnostic indicators of preeclampsia by single-cell RNA sequencing and machine learning. Commun Biol 2024; 7:32. [PMID: 38182876 PMCID: PMC10770323 DOI: 10.1038/s42003-023-05669-2] [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: 04/02/2023] [Accepted: 12/04/2023] [Indexed: 01/07/2024] Open
Abstract
Preeclampsia is a multifactorial and heterogeneous complication of pregnancy. Here, we utilize single-cell RNA sequencing to dissect the involvement of circulating immune cells in preeclampsia. Our findings reveal downregulation of immune response in lymphocyte subsets in preeclampsia, such as reduction in natural killer cells and cytotoxic genes expression, and expansion of regulatory T cells. But the activation of naïve T cell and monocyte subsets, as well as increased MHC-II-mediated pathway in antigen-presenting cells were still observed in preeclampsia. Notably, we identified key monocyte subsets in preeclampsia, with significantly increased expression of angiogenesis pathways and pro-inflammatory S100 family genes in VCAN+ monocytes and IFN+ non-classical monocytes. Furthermore, four cell-type-specific machine-learning models have been developed to identify potential diagnostic indicators of preeclampsia. Collectively, our study demonstrates transcriptomic alternations of circulating immune cells and identifies immune components that could be involved in pathophysiology of preeclampsia.
Collapse
Affiliation(s)
- Wenwen Zhou
- BGI Research, Shenzhen, 518103, China
- College of Life Sciences, South China Agricultural University, Guangzhou, 510642, China
| | - Yixuan Chen
- Department of Obstetrics, Shenzhen Maternity & Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, 518028, China
| | - Yuhui Zheng
- BGI Research, Shenzhen, 518103, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yong Bai
- BGI Research, Shenzhen, 518103, China
| | | | - Xiao-Xia Wu
- Department of Obstetrics, Shenzhen Maternity & Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, 518028, China
| | - Mei Hong
- College of Life Sciences, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, South China Agricultural University, Guangzhou, 510642, China
| | - Langchao Liang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
- BGI Research, Qingdao, 266555, China
| | - Jing Zhang
- Department of Obstetrics, Shenzhen Maternity & Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, 518028, China
| | - Ya Gao
- BGI Research, Shenzhen, 518103, China
| | - Ning Sun
- Department of Obstetrics, Shenzhen Maternity & Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, 518028, China
| | | | - Yiwei Zhang
- Department of Obstetrics, Shenzhen Maternity & Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, 518028, China
| | - Linlin Wu
- Department of Obstetrics, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, China.
| | - Xin Jin
- BGI Research, Shenzhen, 518103, China.
- School of Medicine, South China University of Technology, Guangzhou, 510006, China.
- Shenzhen Key Laboratory of Transomics Biotechnologies, BGI-Shenzhen, Shenzhen, 518083, China.
| | - Jianmin Niu
- Department of Obstetrics, Shenzhen Maternity & Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, 518028, China.
| |
Collapse
|
39
|
Giles ML, Cole S, O’Bryan J, Krishnaswamy S, Ben-Othman R, Amenyogbe N, Davey MA, Kollmann T. The PRotective Effect of Maternal Immunisation on preTerm birth: characterising the Underlying mechanisms and Role in newborn immune function: the PREMITUR study protocol. Front Immunol 2023; 14:1212320. [PMID: 38187392 PMCID: PMC10771328 DOI: 10.3389/fimmu.2023.1212320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 12/04/2023] [Indexed: 01/09/2024] Open
Abstract
Maternal immunisation, a low cost and high efficacy intervention is recommended for its pathogen specific protection. Evidence suggests that maternal immunisation has another significant impact: reduction of preterm birth (PTB), the single greatest cause of childhood morbidity and mortality globally. Our overarching question is: how does maternal immunisation modify the immune system in pregnant women and/or their newborn to reduce adverse pregnancy outcomes and enhance the newborn infant's capacity to protect itself from infectious diseases during early childhood? To answer this question we are conducting a multi-site, prospective observational cohort study collecting maternal and infant biological samples at defined time points during pregnancy and post-partum from nulliparous women. We aim to enrol 400 women and determine the immune trajectory in pregnancy and the impact of maternal immunisation (including influenza, pertussis and/or COVID-19 vaccines) on this trajectory. The results are expected to identify areas that can be targeted for future intervention studies.
Collapse
Affiliation(s)
- Michelle L. Giles
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
- Department of Infectious Diseases, University of Melbourne, Melbourne, VIC, Australia
- Department of Obstetric Medicine and Maternal Fetal Medicine, Royal Women’s Hospital, Melbourne, VIC, Australia
| | - Stephen Cole
- Department of Obstetrics and Gynaecology, Epworth Healthcare, Melbourne, VIC, Australia
| | - Jessica O’Bryan
- Department of Infectious Diseases, Monash Health, Melbourne, VIC, Australia
| | - Sushena Krishnaswamy
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
- Department of Infectious Diseases, Monash Health, Melbourne, VIC, Australia
| | - Rym Ben-Othman
- Department of Paediatrics, Telethon Kids, Perth, WA, Australia
| | - Nelly Amenyogbe
- Department of Paediatrics, Telethon Kids, Perth, WA, Australia
| | - Mary-Ann Davey
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Tobias Kollmann
- Department of Paediatrics, Telethon Kids, Perth, WA, Australia
| |
Collapse
|
40
|
Ding H, An G. Role of the CD40-CD40L expression level pathway in the diagnosis of unexplained recurrent pregnancy loss. J OBSTET GYNAECOL 2023; 43:2280840. [PMID: 38035611 DOI: 10.1080/01443615.2023.2280840] [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: 01/09/2023] [Accepted: 11/03/2023] [Indexed: 12/02/2023]
Abstract
BACKGROUND Unexplained recurrent spontaneous pregnancy loss (URPL) lacks effective treatment and reliable early diagnosis and prediction. Immunologic dysfunction can be an underlying cause of recurrent pregnancy loss (RPL). Considering the regulatory role of CD40-CD40L in immune responses, we explored its clinical significance in URPL. METHODS The 108 women with URPL who were treated in Hebei Yanda Hospital from January 2020 to December 2022 were selected as study subjects, and another 108 healthy women who were not pregnant and matched with the age and body mass index of the study group were selected as the control group. CD40 and CD4 + CD25 + Treg cells in peripheral blood mononuclear cells (PBMCs) and CD40L in peripheral blood platelets were measured by flow cytometry. The predictive value of CD40-CD40L in URPL for the risk of RPL was determined by receiver operating characteristic (ROC) curves. The correlations of CD40-CD40L with CD4 + CD25 + Treg cells and serum pro-inflammatory factors were assessed by Pearson's analysis. RESULTS CD40 on the surface of PBMCs and CD40L on the surface of platelets were up-regulated in URPL patients. CD40 in combination with CD40L had high predictive value for the risk of RPL in URPL patients. Peripheral blood CD40-CD40L was positively linked to IL-17 and IL-23, and negatively to CD4 + CD25 + Treg cells and IL-10 in URPL patients. CONCLUSIONS The CD40-CD40L pathway expression in peripheral blood can help predict the risk of RPL in URPL patients.
Collapse
Affiliation(s)
- Hui Ding
- Department of Obstetrics and Gynecology, Hebei Yanda Hospital, Langfang City, China
| | - Guoqian An
- Department of Obstetrics and Gynecology, Hebei Yanda Hospital, Langfang City, China
| |
Collapse
|
41
|
Quan X, Lan Y, Yang X. Thyroid autoimmunity and future pregnancy outcome in women of recurrent pregnancy loss: a meta-analysis. J Assist Reprod Genet 2023; 40:2523-2537. [PMID: 37770816 PMCID: PMC10643810 DOI: 10.1007/s10815-023-02933-6] [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: 06/21/2023] [Accepted: 09/06/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND Thyroid autoimmunity (TAI) has been associated with the risk of recurrent pregnancy loss (RPL). This systematic review and meta-analysis was conducted to evaluate the influence of TAI on subsequent pregnancy outcome of women with RPL. METHODS A systematic search of Medline, Web of Science, and Embase was conducted to identify studies evaluating the influence of TAI on subsequent risk of pregnancy loss (PL) in women with RPL. Study quality was evaluated via the Newcastle-Ottawa Scale. A random-effects model was utilized to pool the results, accounting for heterogeneity. RESULTS Ten observational studies were included. Compared to women without thyroid autoantibodies, RPL women with TAI had a higher risk of PL in their subsequent pregnancy (risk ratio [RR]: 1.46. 95% confidence interval [CI]: 1.20 to 1.78, p < 0.001; I2 = 35%). Sensitivity analyses showed consistent results in studies with thyroid peroxidase antibody positivity (RR: 1.50, 95% CI: 1.23 to 1.82) and in studies with TAI assessed before pregnancy (RR: 1.28, 95% CI: 1.07 to 1.53). Subgroup analyses showed that the results were not significantly different in prospective and retrospective studies, in RPL defined as at least two or three PL, in euthyroid women and women with euthyroidism or subclinical hypothyroidism, in women with and without levothyroxine treatment, in studies reporting first-trimester or overall PL, and in studies with different quality scores (p for subgroup difference all > 0.05). CONCLUSIONS In women with RPL, positive for TAI may be related to a higher risk of PL in subsequent pregnancy.
Collapse
Affiliation(s)
- Xiaozhen Quan
- Department of Reproductive Center, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Hubei Province, No. 136, Jingzhou Street, Xiangcheng District, Xiangyang City, 441021, China
| | - Yanli Lan
- Department of Reproductive Center, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Hubei Province, No. 136, Jingzhou Street, Xiangcheng District, Xiangyang City, 441021, China
| | - Xuezhou Yang
- Department of Reproductive Center, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Hubei Province, No. 136, Jingzhou Street, Xiangcheng District, Xiangyang City, 441021, China.
| |
Collapse
|
42
|
Tang C, Hu W. The role of Th17 and Treg cells in normal pregnancy and unexplained recurrent spontaneous abortion (URSA): New insights into immune mechanisms. Placenta 2023; 142:18-26. [PMID: 37603948 DOI: 10.1016/j.placenta.2023.08.065] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 08/08/2023] [Accepted: 08/12/2023] [Indexed: 08/23/2023]
Abstract
Recurrent spontaneous abortion (RSA) has various causes, including chromosomal abnormalities, a prethrombotic state, and abnormal uterine anatomical factors. However, in about 50% of cases, the cause remains unknown and is referred to as unexplained recurrent spontaneous abortion (URSA). The fetus is protected from rejection by the maternal system, acting as an allogeneic gene, and immune tolerance serves as a crucial mechanism. The Th17/Treg cell paradigm's emergence as a new subpopulation of CD4+ T cells, interacting with one another, plays an essential role in the immune microenvironment and the body's defense system. This Th17/Treg cell model helps to explain the pathology of recurrent miscarriage that could not be accounted for by the original immune mechanism based on the Th1/Th2 model. Furthermore, the plasticity of Th17 and Treg cells holds innovative significance in autoimmunity and abortion. This paper reviews the role of Th17/Treg cellular immune response in the maintaining normal pregnancy and understanding unexplained recurrent spontaneous abortion.
Collapse
Affiliation(s)
- Cen Tang
- Kunming Medical University, Kunming, Yunnan, 650000, China
| | - Wanqin Hu
- Kunming Medical University Second Affiliated Hospital, Obstetrics Department, Kunming, Yunnan, 650000, China.
| |
Collapse
|
43
|
Zwezdaryk KJ, Kaur A. Role of immunometabolism during congenital cytomegalovirus infection. IMMUNOMETABOLISM (COBHAM, SURREY) 2023; 5:e00034. [PMID: 38037590 PMCID: PMC10683969 DOI: 10.1097/in9.0000000000000034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023]
Abstract
Cytomegalovirus (CMV) is a master manipulator of host metabolic pathways. The impact of CMV metabolic rewiring during congenital CMV on immune function is unknown. CMV infection can directly alter glycolytic and oxidative phosphorylation pathways in infected cells. Recent data suggests CMV may alter metabolism in uninfected neighboring cells. In this mini review, we discuss how CMV infection may impact immune function through metabolic pathways. We discuss how immune cells differ between maternal and decidual compartments and how altered immunometabolism may contribute to congenital infections.
Collapse
Affiliation(s)
- Kevin J. Zwezdaryk
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
- Tulane Center for Aging, Tulane University School of Medicine, New Orleans, LA, USA
- Tulane Brain Institute, Tulane University School of Medicine, New Orleans, LA, USA
| | - Amitinder Kaur
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
- Division of Immunology, Tulane National Primate Research Center, Covington, LA, USA
| |
Collapse
|
44
|
Zhu H, Zhao Z, Xu J, Chen Y, Cai J, Shi C, Zhou L, Zhu Q, Ji L. Comprehensive landscape of the T and B-cell repertoires of newly diagnosed gestational diabetes mellitus. Genomics 2023; 115:110681. [PMID: 37453476 DOI: 10.1016/j.ygeno.2023.110681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 06/03/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
This study conducted a high-throughput sequencing analysis of the T- and B- repertoires in the newly diagnosed GDM patients and evaluated the association between abnormal adaptive immunity and GDM. The unique TCR CDR3 clonotypes were mildly decreased in GDM patients, and the similarity of TCR V-J distributions was higher in the GDM group. Moreover, the usages of the V gene and V-J pair and the frequency distributions of some CDR3 amino acids (AAs) both in BCR and TCR were significantly different between groups. Moreover, the cytokines including IL-4, IL-6, IFN-γ and IL-17A were synchronously elevated in the GDM cases. Our findings provide a comprehensive view of BCR and TCR repertoires at newly diagnosed GDM patients, revealing the mild reduction in unique TCRB CDR3 sequences and slight alteration of the V gene, V-J combination and CDR3 (AA) usages of BCR and TCR. This work provides deep insight into the mechanism of maternal adaptive immunity in GDM and provides novel diagnostic biomarkers and potential immunotherapy targets for GDM.
Collapse
Affiliation(s)
- Hui Zhu
- Department of Internal Medicine, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, PR China
| | - Zhijia Zhao
- School of Public Health, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, PR China
| | - Jin Xu
- School of Public Health, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, PR China; Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, PR China
| | - Yanming Chen
- School of Public Health, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, PR China
| | - Jie Cai
- Center for Reproductive Medicine, Ningbo Women and Children's Hospital, Ningbo, Zhejiang 315211, PR China
| | - Chaoyi Shi
- Center for Reproductive Medicine, Ningbo Women and Children's Hospital, Ningbo, Zhejiang 315211, PR China
| | - Liming Zhou
- Center for Reproductive Medicine, Ningbo Women and Children's Hospital, Ningbo, Zhejiang 315211, PR China
| | - Qiong Zhu
- Department of Pediatrics, Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, PR China
| | - Lindan Ji
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, PR China; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, School of Medicine, Ningbo, Zhejiang 315211, PR China.
| |
Collapse
|
45
|
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
|
46
|
Zhang Y, Wang H, Qiu P, Jiang J, Wu X, Mei J, Sun H. Decidual macrophages derived NO downregulates PD-L1 in trophoblasts leading to decreased Treg cells in recurrent miscarriage. Front Immunol 2023; 14:1180154. [PMID: 37520550 PMCID: PMC10379637 DOI: 10.3389/fimmu.2023.1180154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 06/26/2023] [Indexed: 08/01/2023] Open
Abstract
Introduction Placental trophoblasts contribute to regulatory T (Treg) function via the programmed cell death-1 (PD-1)/PD-1 ligand 1 (PD-L1) pathway during normal pregnancy. Decreased expression of PD-L1 in trophoblasts was closely associated with Treg deficiency in the development of pregnancy failure. Thus, targeting PD-L1 might be a novel therapy to prevent pregnancy loss. However, the mechanisms for modulating the expression of PD-L1 in trophoblasts are an enigma. Methods The proportion of decidual Treg cells, and the profile of decidual macrophages (DMs) sampled from women with normal pregnancy (NP) and recurrent miscarriage (RM) were evaluated by flow cytometry. The expression of Yin and Yang 1 protein (YY1) and PD-L1 in human villous were measured by Immunohistochemistry (IHC), qRT-PCR and western blot. The determination of soluble PD-L1 (sPD-L1) in serum from NP and RM, and trophoblast conditioned media (TCM) was performed by the PD-L1 SimpleStep ELISA kit. Knockdown of YY1 was processed in the human trophoblast derived cell lines, HTR-8 and Bewo, with siYY1 transfection. Peripheral naïve CD4+ T cells were isolated from women with NP for the in vitro culture. The percentages of Treg cells differentiated from peripheral naïve CD4+ T cells were measured by flow cytometry. The interaction between YY1 and CD274 was proved by CHIP. The expression of inducible nitric oxide synthase (iNOS) in decidua was evaluated by IHC. The level of NO in serum from women with NP and RM was determined by the Griess reagent system. The effects of NO on YY1 were determined by the in vitro culture of HTR-8 cells with the NO donor, SNAP. The in vivo model comprising twelve pregnant mice and underwent different treatment. The percentages of Treg cells in murine uterus were measured by flow cytometry. Similarly, Western blot and IHC were performed to determine the expression of YY1 and PD-L1 in murine placenta. Results Decreased expression of YY1 and PD-L1 in trophoblasts and lower proportion of decidual Treg cells were observed in patients with RM. Knockdown of YY1 contributes to a lower expression of YY1 and PD-L1. Soluble PD-L1 in the supernatant from HTR-8 cells was also decreased with siYY1 administration. Lower Treg differentiation was observed in the presence of supernatant from HTR-8 cells treated with siYY1. CHIP analysis revealed that endogenous YY1 directly occupied the promoter region of the CD274 (PD-L1) gene. Accompanied with increased M1 DMs, higher NO was observed in serum sampled from patients with RM. In the presence of Reduced expression of YY1 and PD-L1 was observed in HTR-8 cells with the treatment of SNAP. Furthermore, less Treg differentiation was observed with SNAP treated TCM. Moreover, our in vivo data found that YY1 deficiency was associated with decreased PD-L1, which further resulting in less Treg differentiation and Treg deficiency at the maternal-fetal interface and increased embryo loss. Discussion Our work found the modulatory capacity of YY1 on PD-L1 in trophoblasts during early pregnancy. Furthermore, reduced YY1 was supposed resulting from higher levels of NO produced from the M1 DMs in RM.
Collapse
Affiliation(s)
| | | | | | | | | | - Jie Mei
- *Correspondence: Haixiang Sun, ; Jie Mei,
| | | |
Collapse
|
47
|
Xiao H, Lin R, Chen C, Lian R, Wu Y, Diao L, Yin T, Huang C. γδ-T cell with high toxic potential was associated with recurrent miscarriage. Am J Reprod Immunol 2023; 90:e13717. [PMID: 37382173 DOI: 10.1111/aji.13717] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/26/2023] [Accepted: 05/05/2023] [Indexed: 06/30/2023] Open
Abstract
PROBLEM RM is a common clinical disease in reproduction, affecting approximately 1%-3% of women worldwide. Previous studies have shown the role of peripheral blood γδ-T cells during physiological pregnancy. However, the relationship between the immune status of peripheral blood γδ-T cells and RM is still not well defined. METHOD OF STUDY In this study, mid-luteal peripheral blood from 51 RM patients and 40 healthy women was collected to determine the immune status of γδ-T cells. The percentage of peripheral blood γδ-T cells, and the molecules mediating their toxic potential, including cytotoxic granules (perforin, granzyme B, and granulysin) and receptors (NKG2D, CD158a, and CD158b), were detected by flow cytometry. RESULTS Compared to healthy control, an increase in the proportion of total CD3+ T cells in lymphocytes and a decrease in the ratio of γδ-T cells to CD3+ T cells were observed in patients with RM. The percentages of granzyme B+ γδ-T cells and CD158a+ γδ-T cells in total γδ-T cells or lymphocytes were significantly increased in patients with RM, compared with healthy control. Conversely, CD158b+ γδ-T cells in total γδ-T cells or lymphocytes were significantly decreased in the RM group. CONCLUSION Increased peripheral blood γδ-T cell with high toxic potential was associated with RM.
Collapse
Affiliation(s)
- Huan Xiao
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Rong Lin
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, Guangdong, China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation, Shenzhen, Guangdong, China
| | - Cong Chen
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, Guangdong, China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation, Shenzhen, Guangdong, China
| | - Ruochun Lian
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, Guangdong, China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation, Shenzhen, Guangdong, China
| | - Yulian Wu
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, Guangdong, China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation, Shenzhen, Guangdong, China
| | - Lianghui Diao
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, Guangdong, China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation, Shenzhen, Guangdong, China
| | - Tailang Yin
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Chunyu Huang
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, Guangdong, China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation, Shenzhen, Guangdong, China
| |
Collapse
|
48
|
Giles ML, Way SS, Marchant A, Aghaepour N, James T, Schaltz-Buchholzer F, Zazara D, Arck P, Kollmann TR. Maternal Vaccination to Prevent Adverse Pregnancy Outcomes: An Underutilized Molecular Immunological Intervention? J Mol Biol 2023; 435:168097. [PMID: 37080422 PMCID: PMC11533213 DOI: 10.1016/j.jmb.2023.168097] [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: 02/28/2023] [Revised: 03/27/2023] [Accepted: 04/10/2023] [Indexed: 04/22/2023]
Abstract
Adverse pregnancy outcomes including maternal mortality, stillbirth, preterm birth, intrauterine growth restriction cause millions of deaths each year. More effective interventions are urgently needed. Maternal immunization could be one such intervention protecting the mother and newborn from infection through its pathogen-specific effects. However, many adverse pregnancy outcomes are not directly linked to the infectious pathogens targeted by existing maternal vaccines but rather are linked to pathological inflammation unfolding during pregnancy. The underlying pathogenesis driving such unfavourable outcomes have only partially been elucidated but appear to relate to altered immune regulation by innate as well as adaptive immune responses, ultimately leading to aberrant maternal immune activation. Maternal immunization, like all immunization, impacts the immune system beyond pathogen-specific immunity. This raises the possibility that maternal vaccination could potentially be utilised as a pathogen-agnostic immune modulatory intervention to redirect abnormal immune trajectories towards a more favourable phenotype providing pregnancy protection. In this review we describe the epidemiological evidence surrounding this hypothesis, along with the mechanistic plausibility and present a possible path forward to accelerate addressing the urgent need of adverse pregnancy outcomes.
Collapse
Affiliation(s)
| | - Sing Sing Way
- Center for Inflammation and Tolerance; Cincinnati Children's Hospital, Cincinnati, USA
| | | | - Nima Aghaepour
- Stanford University School of Medicine, Stanford, CA, USA
| | - Tomin James
- Stanford University School of Medicine, Stanford, CA, USA
| | | | - Dimitra Zazara
- Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg, Hamburg, Germany
| | - Petra Arck
- Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg, Hamburg, Germany
| | | |
Collapse
|
49
|
Faas MM, Liu Y, Wekema L, Weis GA, van Loo-Bouwman CA, Silva Lagos L. The Effect of Antibiotics Treatment on the Maternal Immune Response and Gut Microbiome in Pregnant and Non-Pregnant Mice. Nutrients 2023; 15:2723. [PMID: 37375627 DOI: 10.3390/nu15122723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/19/2023] [Accepted: 05/19/2023] [Indexed: 06/29/2023] Open
Abstract
The gut microbiota are involved in adaptations of the maternal immune response to pregnancy. We therefore hypothesized that inducing gut dysbiosis during pregnancy alters the maternal immune response. Thus, pregnant mice received antibiotics from day 9 to day 16 to disturb the maternal gut microbiome. Feces were collected before, during and after antibiotic treatment, and microbiota were measured using 16S RNA sequencing. Mice were sacrificed at day 18 of pregnancy and intestinal (Peyer's patches (PP) and mesenteric lymph nodes (MLN)) and peripheral immune responses (blood and spleen) were measured using flow cytometry. Antibiotic treatment decreased fetal and placental weight. The bacterial count and the Shannon index were significantly decreased (Friedman, followed by Dunn's test, p < 0.05) and the bacterial genera abundance was significantly changed (Permanova, p < 0.05) following antibiotics treatment as compared with before treatment. Splenic Th1 cells and activated blood monocytes were increased, while Th2, Th17 and FoxP3/RoRgT double-positive cells in the PP and MLNs were decreased in pregnant antibiotics-treated mice as compared with untreated pregnant mice. In addition, intestinal dendritic cell subsets were affected by antibiotics. Correlation of immune cells with bacterial genera showed various correlations between immune cells in the PP, MLN and peripheral circulation (blood and spleen). We conclude the disturbed gut microbiota after antibiotics treatment disturbed the maternal immune response. This disturbed maternal immune response may affect fetal and placental weight.
Collapse
Affiliation(s)
- Marijke M Faas
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Yuanrui Liu
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Lieske Wekema
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Gisela A Weis
- Yili Innovation Center Europe, Bronland 12 E-1, 6708 WH Wageningen, The Netherlands
| | | | - Luis Silva Lagos
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| |
Collapse
|
50
|
Liang Y, Lai S, Huang L, Li Y, Zeng S, Zhang S, Chen J, Deng W, Liu Y, Liang J, Xu P, Liu M, Xiong Z, Chen D, Tu Z, Du L. JAZF1 safeguards human endometrial stromal cells survival and decidualization by repressing the transcription of G0S2. Commun Biol 2023; 6:568. [PMID: 37244968 DOI: 10.1038/s42003-023-04931-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 05/12/2023] [Indexed: 05/29/2023] Open
Abstract
Decidualization of human endometrial stromal cells (hESCs) is essential for the maintenance of pregnancy, which depends on the fine-tuned regulation of hESCs survival, and its perturbation contributes to pregnancy loss. However, the underlying mechanisms responsible for functional deficits in decidua from recurrent spontaneous abortion (RSA) patients have not been elucidated. Here, we observed that JAZF1 was significantly downregulated in stromal cells from RSA decidua. JAZF1 depletion in hESCs resulted in defective decidualization and cell death through apoptosis. Further experiments uncovered G0S2 as a important driver of hESCs apoptosis and decidualization, whose transcription was repressed by JAZF1 via interaction with G0S2 activator Purβ. Moreover, the pattern of low JAZF1, high G0S2 and excessive apoptosis in decidua were consistently observed in RSA patients. Collectively, our findings demonstrate that JAZF1 governs hESCs survival and decidualization by repressing G0S2 transcription via restricting the activity of Purβ, and highlight the clinical implications of these mechanisms in the pathology of RSA.
Collapse
Affiliation(s)
- Yingyu Liang
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, Guangdong Engineering and Technology Research Center of Maternal-Fetal Medicine, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Siying Lai
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, Guangdong Engineering and Technology Research Center of Maternal-Fetal Medicine, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Lijun Huang
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, Guangdong Engineering and Technology Research Center of Maternal-Fetal Medicine, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Yulian Li
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, Guangdong Engineering and Technology Research Center of Maternal-Fetal Medicine, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Shanshan Zeng
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, Guangdong Engineering and Technology Research Center of Maternal-Fetal Medicine, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Shuang Zhang
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, Guangdong Engineering and Technology Research Center of Maternal-Fetal Medicine, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Jingsi Chen
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, Guangdong Engineering and Technology Research Center of Maternal-Fetal Medicine, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Wenbo Deng
- Department of Obstetrics and Gynecology, Fujian Provincial Key Laboratory of Reproductive Health Research, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, 361102, China
| | - Yu Liu
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, Guangdong Engineering and Technology Research Center of Maternal-Fetal Medicine, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Jingying Liang
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, Guangdong Engineering and Technology Research Center of Maternal-Fetal Medicine, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Pei Xu
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, Guangdong Engineering and Technology Research Center of Maternal-Fetal Medicine, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Mingxing Liu
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, Guangdong Engineering and Technology Research Center of Maternal-Fetal Medicine, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Zhongtang Xiong
- Department of Pathology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Dunjin Chen
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, Guangdong Engineering and Technology Research Center of Maternal-Fetal Medicine, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| | - Zhaowei Tu
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, Guangdong Engineering and Technology Research Center of Maternal-Fetal Medicine, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| | - Lili Du
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, Guangdong Engineering and Technology Research Center of Maternal-Fetal Medicine, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| |
Collapse
|