1
|
Yu T, Wang G, Xu X, Yan J. Identification and validation of key biomarkers associated with immune and oxidative stress for preeclampsia by WGCNA and machine learning. Front Genet 2025; 16:1500061. [PMID: 40151199 PMCID: PMC11949101 DOI: 10.3389/fgene.2025.1500061] [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: 10/10/2024] [Accepted: 02/19/2025] [Indexed: 03/29/2025] Open
Abstract
Background Preeclampsia (PE), a major obstetric disorder marked by dysfunction in both placental and maternal vascular systems, continues to pose critical challenges in global maternal healthcare. This multisystem pregnancy complication contributes significantly to adverse perinatal outcomes and remains a leading cause of pregnancy-related morbidity worldwide. However, the available treatment options at present remain restricted. Our investigation employs an integrative bioinformatics approach to elucidate critical molecular signatures linked to the interplay between immunological dysregulation and oxidative stress mechanisms in PE pathogenesis. Methods In this study, we sourced the dataset from the GEO database with the aim of pinpointing differentially expressed genes (DEGs) between PE samples and control samples. Genes associated with oxidative stress were procured from the Genecards database. Next, we employed a comprehensive approach. This involved integrating WGCNA, GO and KEGG pathway analyses, constructing PPI networks, applying machine learning algorithms, performing gene GSEA, and conducting immune infiltration analysis to identify the key hub genes related to oxidative stress. Diagnostic potential of candidate biomarkers was quantitatively assessed through ROC curve modeling. Additionally, we constructed a miRNA - gene regulatory network for the identified diagnostic genes and predicted potential candidate drugs. In the final step, we validated the significant hub gene using independent external datasets, the hypoxia model of the HTR-8/SVneo cell line, and human placental tissue samples. Results At last, leptin (LEP) was identified as a core gene through screening and was found to be upregulated. The results of quantitative real-time polymerase chain reaction (qRT -PCR) and immunohistochemistry validation were consistent with those obtained from the datasets. KEGG analysis revealed that LEP was significantly enriched in "allograft rejection," "antigen processing," "ECM receptor interaction" and "graft versus host disease." GO analysis revealed that LEP was involved in biological processes such as "antigen processing and presentation," "peptide antigen assembly with MHC protein complex," "complex of collagen trimers," "MHC class II protein complex" and "mitochondrial protein containing complex." Moreover, immune cell analysis indicated that T follicular helper cells, plasmacytoid dendritic cells, neutrophils, and activated dendritic cells were positively correlated with LEP expression, whereas γδT cells, eosinophils, and central memory CD4+ T cells showed a negative correlation. These findings suggest that LEP influences the immune microenvironment of PE through its interaction with arious immune cells. In addition, 28 miRNAs and 15 drugs were predicted to target LEP. Finally, the overexpression of LEP was verified using independent external datasets, the hypoxia model of the HTR-8/SVneo cell line, and human placental tissue. Conclusion Through an integrated analytical framework employing WGCNA coupled with three distinct machine learning-driven phenotypic classification models, we discovered a pivotal regulatory gene. This gene has the potential to act as a novel diagnostic biomarker for PE. Moreover, it can be considered as a promising target for drug development related to PE. Notably, it shows a strong correlation with the immune microenvironment, suggesting its crucial role in the complex pathophysiological processes underlying PE.
Collapse
Affiliation(s)
- Tiantian Yu
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian, China
- Fujian Clinical Research Center for Maternal - Fetal Medicine, Fuzhou, Fujian, China
- National Key Obstetric Clinical Specialty Construction Institution of China, Fuzhou, Fujian, China
| | - Guiying Wang
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian, China
- Fujian Clinical Research Center for Maternal - Fetal Medicine, Fuzhou, Fujian, China
- National Key Obstetric Clinical Specialty Construction Institution of China, Fuzhou, Fujian, China
| | - Xia Xu
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian, China
- Fujian Clinical Research Center for Maternal - Fetal Medicine, Fuzhou, Fujian, China
- National Key Obstetric Clinical Specialty Construction Institution of China, Fuzhou, Fujian, China
| | - Jianying Yan
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian, China
- Fujian Clinical Research Center for Maternal - Fetal Medicine, Fuzhou, Fujian, China
- National Key Obstetric Clinical Specialty Construction Institution of China, Fuzhou, Fujian, China
| |
Collapse
|
2
|
Yue S, Meng J. Role of Decidual Natural Killer Cells in the Pathogenesis of Preeclampsia. Am J Reprod Immunol 2025; 93:e70033. [PMID: 39739937 DOI: 10.1111/aji.70033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/02/2025] Open
Abstract
Preeclampsia is one of the most severe obstetric complications, yet its pathogenesis remains unclear. Decidual natural killer (dNK) cells, the most abundant immune cells at the maternal-fetal interface, are closely associated with preeclampsia due to abnormalities in their quantity, phenotype, and function. This review summarizes the molecular mechanisms by which dNK cells regulate extravillous trophoblast (EVT) invasion, promote uterine spiral artery remodeling, and maintain immune tolerance. Furthermore, it explores how disruptions in these mechanisms and changes in the decidual microenvironment alter dNK cell properties, driving the progression of preeclampsia. Understanding the mechanisms of dNK cells and identifying potential therapeutic targets may provide new insights for clinical intervention.
Collapse
Affiliation(s)
- Shuang Yue
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jinlai Meng
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan, China
| |
Collapse
|
3
|
Chen S, Zhu H, Jounaidi Y. Comprehensive snapshots of natural killer cells functions, signaling, molecular mechanisms and clinical utilization. Signal Transduct Target Ther 2024; 9:302. [PMID: 39511139 PMCID: PMC11544004 DOI: 10.1038/s41392-024-02005-w] [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: 05/19/2024] [Revised: 08/25/2024] [Accepted: 09/17/2024] [Indexed: 11/15/2024] Open
Abstract
Natural killer (NK) cells, initially identified for their rapid virus-infected and leukemia cell killing and tumor destruction, are pivotal in immunity. They exhibit multifaceted roles in cancer, viral infections, autoimmunity, pregnancy, wound healing, and more. Derived from a common lymphoid progenitor, they lack CD3, B-cell, or T-cell receptors but wield high cytotoxicity via perforin and granzymes. NK cells orchestrate immune responses, secreting inflammatory IFNγ or immunosuppressive TGFβ and IL-10. CD56dim and CD56bright NK cells execute cytotoxicity, while CD56bright cells also regulate immunity. However, beyond the CD56 dichotomy, detailed phenotypic diversity reveals many functional subsets that may not be optimal for cancer immunotherapy. In this review, we provide comprehensive and detailed snapshots of NK cells' functions and states of activation and inhibitions in cancer, autoimmunity, angiogenesis, wound healing, pregnancy and fertility, aging, and senescence mediated by complex signaling and ligand-receptor interactions, including the impact of the environment. As the use of engineered NK cells for cancer immunotherapy accelerates, often in the footsteps of T-cell-derived engineering, we examine the interactions of NK cells with other immune effectors and relevant signaling and the limitations in the tumor microenvironment, intending to understand how to enhance their cytolytic activities specifically for cancer immunotherapy.
Collapse
Affiliation(s)
- Sumei Chen
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China.
| | - Haitao Zhu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Youssef Jounaidi
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
4
|
Jia W, Ma L, Yu X, Wang F, Yang Q, Wang X, Fan M, Gu Y, Meng R, Wang J, Li Y, Li R, Shao X, Wang YL. Human CD56 +CD39 + dNK cells support fetal survival through controlling trophoblastic cell fate: immune mechanisms of recurrent early pregnancy loss. Natl Sci Rev 2024; 11:nwae142. [PMID: 38966071 PMCID: PMC11223582 DOI: 10.1093/nsr/nwae142] [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: 09/11/2023] [Revised: 03/26/2024] [Accepted: 04/04/2024] [Indexed: 07/06/2024] Open
Abstract
Decidual natural killer (dNK) cells are the most abundant immune cells at the maternal-fetal interface during early pregnancy in both mice and humans, and emerging single-cell transcriptomic studies have uncovered various human dNK subsets that are disrupted in patients experiencing recurrent early pregnancy loss (RPL) at early gestational stage, suggesting a connection between abnormal proportions or characteristics of dNK subsets and RPL pathogenesis. However, the functional mechanisms underlying this association remain unclear. Here, we established a mouse model by adoptively transferring human dNK cells into pregnant NOG (NOD/Shi-scid/IL-2Rγnull) mice, where human dNK cells predominantly homed into the uteri of recipients. Using this model, we observed a strong correlation between the properties of human dNK cells and pregnancy outcome. The transfer of dNK cells from RPL patients (dNK-RPL) remarkably worsened early pregnancy loss and impaired placental trophoblast cell differentiation in the recipients. These adverse effects were effectively reversed by transferring CD56+CD39+ dNK cells. Mechanistic studies revealed that CD56+CD39+ dNK subset facilitates early differentiation of mouse trophoblast stem cells (mTSCs) towards both invasive and syncytial pathways through secreting macrophage colony-stimulating factor (M-CSF). Administration of recombinant M-CSF to NOG mice transferred with dNK-RPL efficiently rescued the exacerbated pregnancy outcomes and fetal/placental development. Collectively, this study established a novel humanized mouse model featuring functional human dNK cells homing into the uteri of recipients and uncovered the pivotal role of M-CSF in fetal-supporting function of CD56+CD39+ dNK cells during early pregnancy, highlighting that M-CSF may be a previously unappreciated therapeutic target for intervening RPL.
Collapse
Affiliation(s)
- Wentong Jia
- State Key Laboratory of Stem cell and Reproductive Biology, Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Liyang Ma
- State Key Laboratory of Stem cell and Reproductive Biology, Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xin Yu
- State Key Laboratory of Stem cell and Reproductive Biology, Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of the Chinese Academy of Sciences, Beijing 101408, China
| | - Feiyang Wang
- State Key Laboratory of Stem cell and Reproductive Biology, Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of the Chinese Academy of Sciences, Beijing 101408, China
| | - Qian Yang
- NHC Key Lab of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai 200237, China
| | - Xiaoye Wang
- National Clinical Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| | - Mengjie Fan
- National Clinical Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| | - Yan Gu
- Department of Family Planning, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Ran Meng
- Department of Prenatal Screening, Haidian Maternal and Child Health Hospital, Beijing 100080, China
| | - Jian Wang
- NHC Key Lab of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai 200237, China
| | - Yuxia Li
- State Key Laboratory of Stem cell and Reproductive Biology, Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Rong Li
- National Clinical Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| | - Xuan Shao
- State Key Laboratory of Stem cell and Reproductive Biology, Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of the Chinese Academy of Sciences, Beijing 101408, China
| | - Yan-Ling Wang
- State Key Laboratory of Stem cell and Reproductive Biology, Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of the Chinese Academy of Sciences, Beijing 101408, China
| |
Collapse
|
5
|
Yao Y, Ye Y, Chen J, Zhang M, Cai X, Zheng C. Maternal-fetal immunity and recurrent spontaneous abortion. Am J Reprod Immunol 2024; 91:e13859. [PMID: 38722063 DOI: 10.1111/aji.13859] [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: 12/21/2023] [Revised: 04/15/2024] [Accepted: 04/18/2024] [Indexed: 06/26/2024] Open
Abstract
Recurrent Spontaneous Abortion (RSA) is a common pregnancy complication, that has multifactorial causes, and currently, 40%-50% of cases remain unexplained, referred to as Unexplained RSA (URSA). Due to the elusive etiology and mechanisms, clinical management is exceedingly challenging. In recent years, with the progress in reproductive immunology, a growing body of evidence suggests a relationship between URSA and maternal-fetal immunology, offering hope for the development of tailored treatment strategies. This article provides an immunological perspective on the pathogenesis, diagnosis, and treatment of RSA. On one hand, it comprehensively reviews the immunological mechanisms underlying RSA, including abnormalities in maternal-fetal interface immune tolerance, maternal-fetal interface immune cell function, gut microbiota-mediated immune dysregulation, and vaginal microbiota-mediated immune anomalies. On the other hand, it presents the diagnosis and existing treatment modalities for RSA. This article offers a clear knowledge framework for understanding RSA from an immunological standpoint. In conclusion, while the "layers of the veil" regarding immunological factors in RSA are gradually being unveiled, our current research may only scratch the surface. In terms of immunological etiology, effective diagnostic tools for RSA are currently lacking, and the efficacy and safety of immunotherapies, primarily based on lymphocyte immunotherapy and intravenous immunoglobulin, remain contentious.
Collapse
Affiliation(s)
- Yao Yao
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China
| | - Yiqing Ye
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China
| | - Jia Chen
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China
| | - Meng Zhang
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China
| | - Xiaoyu Cai
- Department of Pharmacy, Hangzhou First People's Hospital, Hangzhou, China
| | - Caihong Zheng
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China
| |
Collapse
|
6
|
Wei X, Yang X. The novel role of activating receptor KIR2DS5 in preeclampsia. Int Immunopharmacol 2023; 125:111087. [PMID: 37864908 DOI: 10.1016/j.intimp.2023.111087] [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: 08/14/2023] [Revised: 10/02/2023] [Accepted: 10/15/2023] [Indexed: 10/23/2023]
Abstract
Preeclampsia (PE) is a serious complication of pregnancy. Decidual natural killer (dNK) cells were reported to participate in the remodeling of spiral arteries through producing a group of cytokines, including granulocyte-macrophage colony stimulating factor (GM-CSF). KIR2DS5 is an activating receptor of NK cells that specifically recognizes HLA-C2 on trophoblasts. Currently, there are no reports regarding the precise mechanism of KIR2DS5 in PE. This study included 30 PE patients and 30 healthy pregnant women. We found that the expressions of KIR2DS5 were significantly lower in PE deciduae compared to those of healthy pregnancies. By transfecting knockdown and overexpression lentivirus vectors of KIR2DS5 into dNK cells isolated from deciduae of early pregnancy, we altered the KIR2DS5 expression level in dNK cells. Then, these dNK cells and trophoblast cell lines were co-cultured as trophoblast-dNK cells. In the trophoblast-dNK cells, we examined the influence of KIR2DS5 on the biological manifestations of trophoblasts. As anticipated, overexpression of KIR2DS5 could facilitate cell proliferation, migration, and invasion. Furthermore, increased expression of KIR2DS5 inhibited cell apoptosis and enhanced the progression of cells from theG1 to theS stage. Further mechanistic study demonstrated a positive relationship between KIR2DS5 and GM-CSF in trophoblast-dNK cells. Accordingly, our observations indicated that a decrease in KIR2DS5 could reduce the expression of GM-CSF via the JAK2/STAT5 pathway, resulting in the failure of the activated signal to be transmitted to dNK cells and ultimately leading to the occurrence of PE. KIR2DS5 may be a new contributor for the prediction and diagnosis of PE.
Collapse
Affiliation(s)
- Xiaoqi Wei
- Department of Obstetrics, The First Hospital of China Medical University, Shenyang 110000, China
| | - Xiuhua Yang
- Department of Obstetrics, The First Hospital of China Medical University, Shenyang 110000, China.
| |
Collapse
|
7
|
Carannante V, Wiklund M, Önfelt B. In vitro models to study natural killer cell dynamics in the tumor microenvironment. Front Immunol 2023; 14:1135148. [PMID: 37457703 PMCID: PMC10338882 DOI: 10.3389/fimmu.2023.1135148] [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: 12/31/2022] [Accepted: 06/05/2023] [Indexed: 07/18/2023] Open
Abstract
Immunotherapy is revolutionizing cancer therapy. The rapid development of new immunotherapeutic strategies to treat solid tumors is posing new challenges for preclinical research, demanding novel in vitro methods to test treatments. Such methods should meet specific requirements, such as enabling the evaluation of immune cell responses like cytotoxicity or cytokine release, and infiltration into the tumor microenvironment using cancer models representative of the original disease. They should allow high-throughput and high-content analysis, to evaluate the efficacy of treatments and understand immune-evasion processes to facilitate development of new therapeutic targets. Ideally, they should be suitable for personalized immunotherapy testing, providing information for patient stratification. Consequently, the application of in vitro 3-dimensional (3D) cell culture models, such as tumor spheroids and organoids, is rapidly expanding in the immunotherapeutic field, coupled with the development of novel imaging-based techniques and -omic analysis. In this paper, we review the recent advances in the development of in vitro 3D platforms applied to natural killer (NK) cell-based cancer immunotherapy studies, highlighting the benefits and limitations of the current methods, and discuss new concepts and future directions of the field.
Collapse
Affiliation(s)
- Valentina Carannante
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Martin Wiklund
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Björn Önfelt
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
- Center for Infectious Medicine, Department of Medicine Huddinge, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
8
|
Bangi EF, Yousuf MH, Upadhyay S, Jain P, Jain R. Comprehensive Review of Hypertensive Disorders Related to Pregnancy. South Med J 2023; 116:482-489. [PMID: 37263611 DOI: 10.14423/smj.0000000000001571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Hypertensive disorder of pregnancy is a common complication during pregnancy that affects approximately 10% of pregnancies and is responsible for nearly 14% of maternal deaths worldwide. It affects the mother and the fetus simultaneously, sometimes putting the health of the mother and the fetus at odds with each other. It may present with only hypertension and proteinuria or with life-threatening complications in the mother such as eclampsia; stroke; acute pulmonary edema; acute renal failure; disseminated intravascular coagulation; placental abruption; hemolysis, elevated liver enzymes, and low platelet syndrome; pregnancy loss; and fetal growth restriction and prematurity resulting from the frequent need of delivering preterm in the fetus. In this review, we aimed to describe hypertensive disorders of pregnancy, mainly preeclampsia and chronic hypertension in pregnancy, by discussing the pathophysiology, the central role of abnormal placentation, the release of antiangiogenic factors in the circulation and immunological factors, the clinical outcome in the mother and the fetus, and the diagnostic criteria and principles of management of both the conditions. We also discuss possible screening methods and prevention of preeclampsia using low-dose aspirin and eclampsia prophylaxis.
Collapse
Affiliation(s)
- Eera Fatima Bangi
- From the Seth Gordhandas Sunderdas Medical College and King Edward Memorial Hospital, Mumbai, India
| | | | | | | | - Rohit Jain
- Department of Internal Medicine, Penn State Milton S. Hershey Medical Center, Hershey, Pennsylvania
| |
Collapse
|
9
|
Rao VA, Kurian NK, Rao KA. Cytokines, NK cells and regulatory T cell functions in normal pregnancy and reproductive failures. Am J Reprod Immunol 2023; 89:e13667. [PMID: 36480305 DOI: 10.1111/aji.13667] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 10/22/2022] [Accepted: 12/02/2022] [Indexed: 12/14/2022] Open
Abstract
PURPOSE OF THE REVIEW Pregnancy brings about an intricate assortment of dynamic changes, which causes proper connection of genetically discordant maternal and foetal tissues. Uterine NK cells are immune cells populating the endometrium and play a major role in implantation and also regulate placentation. This review mainly aims explore the role of uterine NK cells in implantation and how it is affecting in adverse pregnancy outcomes. RECENT FINDINGS Though the functions of uterine NK (uNK) cells are not clearly understood, NK cell activity plays a vital role during immunomodulation which is the main step in implantation and sustaining the early pregnancy. Cytokines, cell surface receptors of NK cells and hormones such as progesterone modulate the NK cell activity in turn affect the implantation of the embryo. Altered NK cell activity (number and functionality) would be an important attributing factor in adverse pregnancy outcomes. Furthermore, T regulatory cells and cytokines also modulate the immune responses in the decidua which in turn contributes to successful implantation of embryos. SUMMARY Immunological responses and interactions in the Foetus-maternal interface is crucial in the successful implantation of allogenic foetus resulting in a healthy pregnancy. NK cells, Treg cells and cytokines play a major role in successful implantation which remains an enigma. Comprehending pregnancy-induced immunological changes at the foetus-maternal interface will allow newer therapeutic strategies to improve pregnancy outcomes.
Collapse
Affiliation(s)
| | - Noble K Kurian
- University Institute of Biotechnology, Chandigarh University, Mohali, India
| | - Kamini A Rao
- Department, of Reproductive Medicine, Bangalore, India
| |
Collapse
|
10
|
Davenport BN, Jones HN, Wilson RL. Placental treatment with insulin-like growth factor 1 via nanoparticle differentially impacts vascular remodeling factors in guinea pig sub-placenta/decidua. Front Physiol 2023; 13:1055234. [PMID: 36685211 PMCID: PMC9845775 DOI: 10.3389/fphys.2022.1055234] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 12/13/2022] [Indexed: 01/06/2023] Open
Abstract
Clinically, fetal growth restriction (FGR) is only detectable in later gestation, despite pathophysiological establishment likely earlier in pregnancy. Additionally, there are no effective in utero treatment options for FGR. We have developed a nanoparticle to deliver human insulin-like 1 growth factor (hIGF-1) in a trophoblast-specific manner which results in increased expression of hIGF-1. IGF-1 signaling in the placenta regulates multiple developmental processes including trophoblast invasion and maternal vascular remodeling, both of which can be diminished in the FGR placenta. We aimed to determine the effects of short-term hIGF-1 nanoparticle treatment on sub-placenta/decidua trophoblast signaling mechanisms in FGR and under normal growth conditions. Using the guinea pig maternal nutrient restriction (MNR) model of FGR, ultrasound-guided, intra-placenta injections of hIGF-1 nanoparticle were performed at gestational day 30-33, and dams sacrificed 5 days later. Sub-placenta/decidua tissue was separated from placenta for further analyses. Western blot was used to analyze protein expression of ERK/AKT/mTOR signaling proteins (phospho-Erk (pERK), phospho-Akt (pAKT), raptor, rictor and deptor). qPCR was used to analyze gene expression of vascular/remodeling factors [vascular endothelial growth factor (Vegf), placenta growth factor (Pgf), platelet-derived growth factor (Pdgf)) and tight junction/adhesion proteins (claudin 5 (Cldn5), p-glycoprotein (Abcb1), occludin (Ocln) and tight junction protein 1 (Zo1)]. MNR reduced expression of pERK, PdgfB and Cldn5, and increased expression of Ocln and Zo1 in the sub-placenta/decidua. In MNR + hIGF1 nanoparticle sub-placenta/decidua, expression of PdgfB, Ocln and Zo1 was normalized, whilst pAkt, VegfB, Vegf receptor 1 and PdgfB receptor were increased compared to MNR. In contrast, hIGF-1 nanoparticle treatment of normal placentas reduced expression of pERK, raptor and increased expression of the mTOR inhibitor deptor. This was associated with reduced expression of VegfA, Plgf, and PdgfB. Here we have shown that the impact of hIGF-1 nanoparticle treatment is dependent on pregnancy environment. Under MNR/FGR, hIGF-1 nanoparticle treatment triggers increased expression of growth factors and normalization of EMT factors. However, under normal conditions, the response of the placenta is to decrease AKT/mTOR signaling and growth factor expression to achieve homeostasis.
Collapse
Affiliation(s)
- Baylea N. Davenport
- Center for Research in Perinatal Outcomes, University of Florida College of Medicine, Gainesville, FL, United States
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL, United States
| | - Helen N. Jones
- Center for Research in Perinatal Outcomes, University of Florida College of Medicine, Gainesville, FL, United States
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL, United States
| | - Rebecca L. Wilson
- Center for Research in Perinatal Outcomes, University of Florida College of Medicine, Gainesville, FL, United States
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL, United States
| |
Collapse
|
11
|
Shen MJ, Pan DC, Du L, Jiang GJ. Clinical Research into Treating Unexplained Recurrent Spontaneous Abortion during Early Pregnancy with the Qing Yi Tiao Mian Formula. Explore (NY) 2023; 19:52-57. [PMID: 35397998 DOI: 10.1016/j.explore.2022.03.009] [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: 11/02/2021] [Revised: 03/22/2022] [Accepted: 03/24/2022] [Indexed: 01/25/2023]
Abstract
OBJECTIVE The present study aims to analyze the clinical effect of the Qing Yi Tiao Mian (QYTM) formula on unexplained recurrent spontaneous abortion (URSA) during early pregnancy and the immune balance of T lymphocytes. METHODS With their consent, 45 patients with URSA in weeks 4-9 of pregnancy were separated into three groups, i.e., the conventional fetal protection (n = 15), prednisone treatment (n = 10), and QYTM formula treatment (n = 20) groups. These patients received treatment once they had been diagnosed with an intrauterine pregnancy. The conventional fetal protection group was given progesterone (20 ∼ 40 mg daily injection) for four weeks. The prednisone treatment group was given progesterone (20 ∼ 40 mg daily injection) + prednisone (5 mg/d) for four weeks. The QYTM formula treatment group was given progesterone (20 ∼ 40 mg daily injection) + QYTM formula (one dose per day) for four weeks. In addition, women who had previously had a normal pregnancy were enrolled as a control group (n = 18). The success rate of the pregnancy in the first trimester was observed in each group, and the proportion of T lymphocytes in the peripheral blood before and after treatment was recorded. RESULTS Among the 20 patients with URSA in the QYTM formula treatment group, 19 remained pregnant. Thus, the success rate during early pregnancy was 95%, which was significantly higher than the conventional fetal protection (53.33%) and prednisone treatment (70%) groups. The CD8+ T and natural killer (NK) cells population in the URSA groups was higher compared with the control group (P < 0.01). The QYTM formula treatment significantly decreased the ratio of CD8+ T lymphocytes (P < 0.01) and NK cells (P < 0.01). CONCLUSION The QYTM formula significantly decreased the spontaneous abortion rate in patients with URSA during early pregnancy. The mechanism may be closely related to the inhibition of the killer lymphocytes' proliferation by CD8+ T lymphocytes and NK cells.
Collapse
Affiliation(s)
- Ming-Jie Shen
- Department of Gynecology and Obstetrics, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ding-Chen Pan
- Department of Gynecology and Obstetrics, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Le Du
- Department of Gynecology and Obstetrics, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Guo-Jing Jiang
- Department of Gynecology and Obstetrics, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
12
|
Roy R, Singh SK, Misra S. Advancements in Cancer Immunotherapies. Vaccines (Basel) 2022; 11:vaccines11010059. [PMID: 36679904 PMCID: PMC9861770 DOI: 10.3390/vaccines11010059] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 12/28/2022] Open
Abstract
Recent work has suggested involvement of the immune system in biological therapies specifically targeting tumor microenvironment. Substantial advancement in the treatment of malignant tumors utilizing immune cells, most importantly T cells that play a key role in cell-mediated immunity, have led to success in clinical trials. Therefore, this article focuses on the therapeutic approaches and developmental strategies to treat cancer. This review emphasizes the immunomodulatory response, the involvement of key tumor-infiltrating cells, the mechanistic aspects, and prognostic biomarkers. We also cover recent advancements in therapeutic strategies.
Collapse
Affiliation(s)
- Ruchi Roy
- UICentre for Drug Discovery, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60607, USA
- Correspondence:
| | - Sunil Kumar Singh
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Sweta Misra
- UICentre for Drug Discovery, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60607, USA
| |
Collapse
|
13
|
Korenevsky AV, Gert TN, Berezkina ME, Sinyavin SA, Mikhailova VA, Markova KL, Simbirtsev AS, Selkov SA, Sokolov DI. Protein Fractions of Natural Killer Cell Lysates Affect the Phenotype, Proliferation and Migration of Endothelial Cells in vitro. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022070171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
14
|
Zhang Y, An C, Yu Y, Lin J, Jin L, Li C, Tan T, Yu Y, Fan Y. Epidermal growth factor induces a trophectoderm lineage transcriptome resembling that of human embryos during reconstruction of blastoids from extended pluripotent stem cells. Cell Prolif 2022; 55:e13317. [PMID: 35880490 PMCID: PMC9628219 DOI: 10.1111/cpr.13317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/23/2022] [Accepted: 06/27/2022] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVES This study aims to optimize the human extended pluripotent stem cell (EPSC) to trophectoderm (TE)-like cell induction with addition of EGF and improve the quality of the reconstructing blastoids. MATERIALS AND METHODS TE-like cells were differentiated from human EPSCs. RNA-seq data analysis was performed to compare with TE-like cells from multiple human pluripotent stem cells (hPSCs) and embryos. A small-scale compound selection was performed for optimizing the TE-like cell induction and the efficiency was characterized using TE-lineage markers expression by immunofluorescence stanning. Blastoids were generated by using the optimized TE-like cells and the undifferentiated human EPSCs through three-dimensional culture system. Single-cell RNA sequencing was performed to investigate the lineage segregation of the optimized blastoids to human blastocysts. RESULTS TE-like cells derived from human EPSCs exhibited similar transcriptome with TE cells from embryos. Additionally, TE-like cells from multiple naive hPSCs exhibited heterogeneous gene expression patterns and signalling pathways because of the incomplete silencing of naive-specific genes and loss of imprinting. Furthermore, with the addition of EGF, TE-like cells derived from human EPSCs enhanced the TE lineage-related signalling pathways and exhibited more similar transcriptome to human embryos. Through resembling with undifferentiated human EPSCs, we elevated the quality and efficiency of reconstructing blastoids and separated more lineage cells with precise temporal and spatial expression, especially the PE lineage. CONCLUSION Addition of EGF enhanced TE lineage differentiation and human blastoids reconstruction. The optimized blastoids could be used as a blastocyst model for simulating early embryonic development.
Collapse
Affiliation(s)
- Yingying Zhang
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education InstitutesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Chenrui An
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education InstitutesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Yanhong Yu
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education InstitutesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Jiajing Lin
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education InstitutesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Long Jin
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education InstitutesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Chaohui Li
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education InstitutesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Tao Tan
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingChina
| | - Yang Yu
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center for Reproductive Medicine, Department of Obstetrics and GynecologyPeking University Third HospitalBeijingChina
| | - Yong Fan
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education InstitutesThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| |
Collapse
|
15
|
Placental Syndromes-A New Paradigm in Perinatology. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19127392. [PMID: 35742640 PMCID: PMC9224387 DOI: 10.3390/ijerph19127392] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/08/2022] [Accepted: 06/13/2022] [Indexed: 12/10/2022]
Abstract
Placental syndromes include pregnancy loss, fetal growth restriction, preeclampsia, preterm delivery, premature rupture of membranes, placental abruption and intrauterine fetal demise. This paper discusses the common etiopathogenesis of those syndromes and the role of angiogenic biomarkers in their development. Pregnancy implantation, placental development and maternal adaptation are complex processes in which fetal and maternal cells interact. The syncytiotrophoblast, trophoblast, uterine natural killer cells and regulatory T cells interfere and interact in all the above-mentioned processes. The proper angioneogenesis and vasculogenesis of the placenta, as well as maternal circulatory adaptation, are dependent on angiogenic factor expression. Insufficient maternal immunotolerance, dysregulation in uterine natural killer or regulatory T cell function, syncytiotrophoblast and trophoblast ischemia and hypoxia or impaired balance in angiogenic factors are all related to the occurrence of placental syndromes. Differences in the time of impairment onset and its intensity and correlation with other dysfunctions result in the development of a specific syndrome. The clinical manifestations in the form of a combination of specific symptoms determine the diagnosis. However, they are just symptoms of an underlying complex trophoblast disorder.
Collapse
|
16
|
Xu X, Zhou Y, Fu B, Wei H. Uterine NK cell functions at maternal-fetal interface. Biol Reprod 2022; 107:327-338. [PMID: 35551350 DOI: 10.1093/biolre/ioac094] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 04/21/2022] [Accepted: 04/29/2022] [Indexed: 11/14/2022] Open
Abstract
During pregnancy, maternal decidual tissue interacts with fetal trophoblasts. They constitute the maternal-fetal interface responsible for supplying nutrition to the fetus. Uterine natural killer (uNK) cells are the most abundant immune cells at the maternal-fetal interface during early pregnancy and play critical roles throughout pregnancy. This review provides current knowledge about the functions of uNK cells. uNK cells have been shown to facilitate remodeling of the spiral artery, control the invasion of extravillous trophoblast (EVT) cells, contribute to the induction and maintenance of immune tolerance, protect against pathogen infection, and promote fetal development. Pregnancy-trained memory of uNK cells improves subsequent pregnancy outcomes. In addition, this review describes the distinct functions of three uNK cell subsets: CD27-CD11b-, CD27+ and CD27-CD11b+ uNK cells.
Collapse
Affiliation(s)
- Xiuxiu Xu
- Institute of Gerontology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, P.R. China.,Institute of Immunology, University of Science and Technology of China, Hefei, Anhui, 230001, P.R. China
| | - Yonggang Zhou
- Institute of Gerontology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, P.R. China.,Institute of Immunology, University of Science and Technology of China, Hefei, Anhui, 230001, P.R. China
| | - Binqing Fu
- Institute of Gerontology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, P.R. China.,Institute of Immunology, University of Science and Technology of China, Hefei, Anhui, 230001, P.R. China
| | - Haiming Wei
- Institute of Gerontology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, P.R. China.,Institute of Immunology, University of Science and Technology of China, Hefei, Anhui, 230001, P.R. China
| |
Collapse
|
17
|
Jin X, Mao L, Zhao W, Liu L, Li Y, Li D, Zhang Y, Du M. Decidualization-derived cAMP promotes decidual NK cells to be angiogenic phenotype. Am J Reprod Immunol 2022; 88:e13540. [PMID: 35348271 DOI: 10.1111/aji.13540] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 03/02/2022] [Accepted: 03/21/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND During early pregnancy, a large number of CD56bright natural killer cells (NKs) are accumulated in the decidua; unlike peripheral and cord blood NK cells (pNK and cNK), these decidual NK cells (dNK) display a great capacity to secrete a series of angiogenic/vascular factors, which are essential for placental development. However, the mechanism underlying the formation of dNK cells with an angiogenic phenotype remains unclear. METHODS First, we compared the difference between dNK and cNK/pNK cells in terms of the expression of CD56 and VEGF, and the regulation of the tube formation. The effect of cAMP on the differentiation of NK cells was evaluated by its analog and inhibitor stimulation. We further analyzed the differences in the phenotype of dNK cells and the expression of VEGF in dNK cells from normal pregnancies and miscarriages. RESULTS Different from cNK and pNK, dNK cells displayed high expression of CD56 and VEGF. And dNK cells showed a higher capacity of inducing tube formation of HUVEC by trophoblast. Meanwhile, we observed that cAMP-analogue increased the percentage of CD56bright NK population in cNK cells with up-regulated VEGF secretion and tube formation of HUVEC by trophoblast, which could be inhibited by pretreatment with VEGFR neutralizing antibody. Similar changes occurred when co-culturing cNK cells with DSCs but not ESCs. Interestingly, the inhibitor of cAMP signaling (Metadoxine, META) could significantly inhibit the upregulation of VEGF in cNK cells by DSCs. Furthermore, DSCs could secret much more cAMP than ESCs. Notably, decreased CD56bright NK population and VEGF secretion by dNK were related to pregnancy loss. CONCLUSIONS These findings suggest that dNK cells display an angiogenic phenotype that can be induced by decidualized cAMP signaling. Our study indicates the significance of decidualization-derived cAMP in regulating angiogenesis of decidual NKs and reveals complex crosstalk between different cell types in a critical period during early pregnancy.
Collapse
Affiliation(s)
- Xueling Jin
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Lie Mao
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Weijie Zhao
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Lu Liu
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Yanhong Li
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Dajin Li
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Ying Zhang
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Meirong Du
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China.,Department of Obstetrics and Gynecology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China.,State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau SAR, Macau, China
| |
Collapse
|
18
|
Du X, Zhu H, Jiao D, Nian Z, Zhang J, Zhou Y, Zheng X, Tong X, Wei H, Fu B. Human-Induced CD49a+ NK Cells Promote Fetal Growth. Front Immunol 2022; 13:821542. [PMID: 35185911 PMCID: PMC8854499 DOI: 10.3389/fimmu.2022.821542] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/19/2022] [Indexed: 01/27/2023] Open
Abstract
CD49a+ natural killer (NK) cells play a critical role in promoting fetal development and maintaining immune tolerance at the maternal-fetal interface during the early stages of pregnancy. However, given their residency in human tissue, thorough studies and clinical applications are difficult to perform. It is still unclear as to how functional human CD49a+ NK cells can be induced to benefit pregnancy outcomes. In this study, we established three no-feeder cell induction systems to induce human CD49a+ NK cells from umbilical cord blood hematopoietic stem cells (HSCs), bone marrow HSCs, and peripheral blood NK cells in vitro. These induced NK cells (iNKs) from three cell induction systems display high levels of CD49a, CD9, CD39, CD151 expression, low levels of CD16 expression, and no obvious cytotoxic capability. They are phenotypically and functionally similar to decidual NK cells. Furthermore, these iNKs display a high expression of growth-promoting factors and proangiogenic factors and can promote fetal growth and improve uterine artery blood flow in a murine pregnancy model in vivo. This research demonstrates the ability of human-induced CD49a+ NK cells to promote fetal growth via three cell induction systems, which could eventually be used to treat patients experiencing adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Xianghui Du
- The Department of Obstetrics and Gynecology, First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Huaiping Zhu
- The Section of Experimental Hematology, First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- *Correspondence: Huaiping Zhu, ; Haiming Wei, ; Binqing Fu,
| | - Defeng Jiao
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Zhigang Nian
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Jinghe Zhang
- The Department of Obstetrics and Gynecology, First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Yonggang Zhou
- The Department of Obstetrics and Gynecology, First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Xiaohu Zheng
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Xianhong Tong
- The Department of Obstetrics and Gynecology, First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Haiming Wei
- The Department of Obstetrics and Gynecology, First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
- *Correspondence: Huaiping Zhu, ; Haiming Wei, ; Binqing Fu,
| | - Binqing Fu
- The Department of Obstetrics and Gynecology, First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
- *Correspondence: Huaiping Zhu, ; Haiming Wei, ; Binqing Fu,
| |
Collapse
|
19
|
Imbalances in circulating angiogenic factors in the pathophysiology of preeclampsia and related disorders. Am J Obstet Gynecol 2022; 226:S1019-S1034. [PMID: 33096092 PMCID: PMC8884164 DOI: 10.1016/j.ajog.2020.10.022] [Citation(s) in RCA: 152] [Impact Index Per Article: 50.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/26/2020] [Accepted: 10/17/2020] [Indexed: 12/11/2022]
Abstract
Preeclampsia is a devastating medical complication of pregnancy that can lead to significant maternal and fetal morbidity and mortality. It is currently believed that there is abnormal placentation in as early as the first trimester in women destined to develop preeclampsia. Although the etiology of the abnormal placentation is being debated, numerous epidemiologic and experimental studies suggest that imbalances in circulating angiogenic factors released from the placenta are responsible for the maternal signs and symptoms of preeclampsia. In particular, circulating levels of soluble fms-like tyrosine kinase 1, an antiangiogenic factor, are markedly increased in women with preeclampsia, whereas free levels of its ligand, placental, growth factor are markedly diminished. Alterations in these angiogenic factors precede the onset of clinical signs of preeclampsia and correlate with disease severity. Recently, the availability of automated assays for the measurement of angiogenic biomarkers in the plasma, serum, and urine has helped investigators worldwide to demonstrate a key role for these factors in the clinical diagnosis and prediction of preeclampsia. Numerous studies have reported that circulating angiogenic biomarkers have a very high negative predictive value to rule out clinical disease among women with suspected preeclampsia. These blood-based biomarkers have provided a valuable tool to clinicians to accelerate the time to clinical diagnosis and minimize maternal adverse outcomes in women with preeclampsia. Angiogenic biomarkers have also been useful to elucidate the pathogenesis of related disorders of abnormal placentation such as intrauterine growth restriction, intrauterine fetal death, twin-to-twin transfusion syndrome, and fetal hydrops. In summary, the discovery and characterization of angiogenic proteins of placental origin have provided clinicians a noninvasive blood-based tool to monitor placental function and health and for early detection of disorders of placentation. Uncovering the mechanisms of altered angiogenic factors in preeclampsia and related disorders of placentation may provide insights into novel preventive and therapeutic options.
Collapse
|
20
|
Salazar MD, Wang WJ, Skariah A, He Q, Field K, Nixon M, Reed R, Dambaeva S, Beaman K, Gilman-Sachs A, Kwak-Kim J. Post-hoc evaluation of peripheral blood natural killer cell cytotoxicity in predicting the risk of recurrent pregnancy losses and repeated implantation failures. J Reprod Immunol 2022; 150:103487. [DOI: 10.1016/j.jri.2022.103487] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 12/04/2021] [Accepted: 01/16/2022] [Indexed: 12/24/2022]
|
21
|
Hofbauer Cells Spread Listeria monocytogenes among Placental Cells and Undergo Pro-Inflammatory Reprogramming while Retaining Production of Tolerogenic Factors. mBio 2021; 12:e0184921. [PMID: 34399615 PMCID: PMC8406333 DOI: 10.1128/mbio.01849-21] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pregnant women are highly susceptible to infection by the bacterial pathogen Listeria monocytogenes, leading to miscarriage, premature birth, and neonatal infection. L. monocytogenes is thought to breach the placental barrier by infecting trophoblasts at the maternal/fetal interface. However, the fate of L. monocytogenes within chorionic villi and how infection reaches the fetus are unsettled. Hofbauer cells (HBCs) are fetal placental macrophages and the only leukocytes residing in healthy chorionic villi, forming a last immune barrier protecting fetal blood from infection. Little is known about the HBCs’ antimicrobial responses to pathogens. Here, we studied L. monocytogenes interaction with human primary HBCs. Remarkably, despite their M2 anti-inflammatory phenotype at basal state, HBCs phagocytose and kill non-pathogenic bacteria like Listeria innocua and display low susceptibility to infection by L. monocytogenes. However, L. monocytogenes can exploit HBCs to spread to surrounding placental cells. Transcriptomic analyses by RNA sequencing revealed that HBCs undergo pro-inflammatory reprogramming upon L. monocytogenes infection, similarly to macrophages stimulated by the potent M1-polarizing agents lipopolysaccharide (LPS)/interferon gamma (IFN-γ). Infected HBCs also express pro-inflammatory chemokines known to promote placental infiltration by maternal leukocytes. However, HBCs maintain the expression of a collection of tolerogenic genes and secretion of tolerogenic cytokines, consistent with their tissue homeostatic role in prevention of fetal rejection. In conclusion, we propose a previously unrecognized model in which HBCs promote the spreading of L. monocytogenes among placental cells and transition to a pro-inflammatory state likely to favor innate immune responses, while maintaining the expression of tolerogenic factors known to prevent maternal anti-fetal adaptive immunity.
Collapse
|
22
|
Espino A, El Costa H, Tabiasco J, Al-Daccak R, Jabrane-Ferrat N. Innate Immune Response to Viral Infections at the Maternal-Fetal Interface in Human Pregnancy. Front Med (Lausanne) 2021; 8:674645. [PMID: 34368184 PMCID: PMC8339430 DOI: 10.3389/fmed.2021.674645] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 06/25/2021] [Indexed: 12/21/2022] Open
Abstract
The placenta, the first and largest organ to develop after conception, not only nurtures and promotes the development of the conceptus, but, it also functions as a barrier against invading pathogens. Early phases of pregnancy are associated with expansion of specific subsets of Natural Killer cells (dNK) and macrophages (dMφ) at the maternal uterine mucosa, the basal decidua. In concert with cells of fetal origin, dNK cells, and dMφ orchestrate all steps of placenta and fetus development, and provide the first line of defense to limit vertical transmission. However, some pathogens that infect the mother can overcome this protective barrier and jeopardize the fetus health. In this review, we will discuss how members of the classical TORCH family (Toxoplasma, Other, Rubella, Cytomegalovirus, and Herpes simplex virus) and some emerging viruses (Hepatitis E virus, Zika virus, and SARS-CoV2) can afford access to the placental fortress. We will also discuss how changes in the intrauterine environment as a consequence of maternal immune cell activation contribute to placental diseases and devastating pregnancy outcomes.
Collapse
Affiliation(s)
- Ana Espino
- Infinity, Université de Toulouse - CNRS - Inserm, CHU Purpan, Toulouse, France
| | - Hicham El Costa
- Infinity, Université de Toulouse - CNRS - Inserm, CHU Purpan, Toulouse, France
| | - Julie Tabiasco
- Infinity, Université de Toulouse - CNRS - Inserm, CHU Purpan, Toulouse, France
| | - Reem Al-Daccak
- Inserm UMRS976 - Université de Paris - Hôpital Saint-Louis, Paris, France
| | | |
Collapse
|
23
|
Wang Y, Hu C, Fang T, Jin Y, Wu R. Perspective on prenatal polychlorinated biphenyl exposure and the development of the progeny nervous system (Review). Int J Mol Med 2021; 48:150. [PMID: 34132363 PMCID: PMC8219518 DOI: 10.3892/ijmm.2021.4983] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 05/26/2021] [Indexed: 02/05/2023] Open
Abstract
The developmental origins of health and disease concept illustrates that exposure in early life to various factors may affect the offspring's long-term susceptibility to disease. During development, the nervous system is sensitive and vulnerable to the environmental insults. Polychlorinated biphenyls (PCBs), which are divided into dioxin-like (DL-PCBs) and non-dioxin-like PCBs (NDL-PCBs), are synthetic persistent environmental endocrine-disrupting chemicals. The toxicological mechanisms of DL-PCBs have been associated with the activation of the aryl hydrocarbon receptor and NDL-PCBs have been associated with ryanodine receptor-mediated calcium ion channels, which affect neuronal migration, promote dendritic growth and alter neuronal connectivity. In addition, PCB accumulation in the placenta destroys the fetal placental unit and affects endocrine function, particularly thyroid hormones and the dopaminergic system, leading to neuroendocrine disorders. However, epidemiological investigations have not achieved a consistent result in different study cohorts. The present review summarizes the epidemiological differences and possible mechanisms of the effects of intrauterine PCB exposure on neurological development.
Collapse
Affiliation(s)
- Yinfeng Wang
- Department of Gynecology and Obstetrics, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Changchang Hu
- Department of Gynecology and Obstetrics, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Tao Fang
- Department of Gynecology and Obstetrics, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Yang Jin
- Department of Gynecology and Obstetrics, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Ruijin Wu
- Department of Gynecology and Obstetrics, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| |
Collapse
|
24
|
Llobat L. Pluripotency and Growth Factors in Early Embryonic Development of Mammals: A Comparative Approach. Vet Sci 2021; 8:vetsci8050078. [PMID: 34064445 PMCID: PMC8147802 DOI: 10.3390/vetsci8050078] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 04/27/2021] [Accepted: 05/02/2021] [Indexed: 12/24/2022] Open
Abstract
The regulation of early events in mammalian embryonic development is a complex process. In the early stages, pluripotency, cellular differentiation, and growth should occur at specific times and these events are regulated by different genes that are expressed at specific times and locations. The genes related to pluripotency and cellular differentiation, and growth factors that determine successful embryonic development are different (or differentially expressed) among mammalian species. Some genes are fundamental for controlling pluripotency in some species but less fundamental in others, for example, Oct4 is particularly relevant in bovine early embryonic development, whereas Oct4 inhibition does not affect ovine early embryonic development. In addition, some mechanisms that regulate cellular differentiation do not seem to be clear or evolutionarily conserved. After cellular differentiation, growth factors are relevant in early development, and their effects also differ among species, for example, insulin-like growth factor improves the blastocyst development rate in some species but does not have the same effect in mice. Some growth factors influence genes related to pluripotency, and therefore, their role in early embryo development is not limited to cell growth but could also involve the earliest stages of development. In this review, we summarize the differences among mammalian species regarding the regulation of pluripotency, cellular differentiation, and growth factors in the early stages of embryonic development.
Collapse
Affiliation(s)
- Lola Llobat
- Research Group Microbiological Agents Associated with Animal Reproduction (PROVAGINBIO), Department of Animal Production and Health, Veterinary Public Health and Food Science and Technology (PASAPTA) Facultad de Veterinaria, Universidad Cardenal Herrera-CEU, CEU Universities, 46113 Valencia, Spain
| |
Collapse
|
25
|
Fonseca BM, Cunha SC, Gonçalves D, Mendes A, Braga J, Correia-da-Silva G, Teixeira NA. Decidual NK cell-derived conditioned medium from miscarriages affects endometrial stromal cell decidualisation: endocannabinoid anandamide and tumour necrosis factor-α crosstalk. Hum Reprod 2021; 35:265-274. [PMID: 31990346 DOI: 10.1093/humrep/dez260] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 11/04/2019] [Accepted: 11/11/2019] [Indexed: 12/15/2022] Open
Abstract
STUDY QUESTION What are the effects of endocannabinoid anandamide (AEA) in uterine natural killer (unK) cells from miscarriage decidua, regarding their cytokine profile and endometrial stromal cell (ESC) crosstalk? SUMMARY ANSWER uNK-conditioned media from miscarriage samples present high TNF-α levels which inhibit ESC decidualisation. WHAT IS KNOWN ALREADY AEA plasma levels are higher in women who have suffered a miscarriage. Moreover, AEA inhibits ESC proliferation and differentiation, although the levels and impact on the uNK cell cytokine profile at the feto-maternal interface remain elusive. STUDY DESIGN, SIZE, DURATION This laboratory-based study used human primary uNK cells which were isolated from first-trimester decidua (gestational age, 5-12 weeks) derived from 8 women with elective pregnancy termination and 18 women who suffered a miscarriage. PARTICIPANTS/MATERIALS, SETTING, METHODS The first-trimester placental tissues were assayed for AEA levels by UPLC-MS/MS and respective enzymatic profile by western blot. The uNK cells were isolated and maintained in culture. The expression of angiogenic markers in uNK cells was examined by quantitative PCR (qPCR). The uNK-conditioned medium was analysed for IFN-γ, TNF-α and IL-10 production by enzyme-linked immunosorbent assay, and the impact on ESC differentiation was assessed by measuring decidual markers Prl, Igfbp-1 and Fox01 mRNA expression using qPCR. MAIN RESULTS AND THE ROLE OF CHANCE AEA levels were higher in miscarriage decidua compared with decidua from elective terminations. The uNK cell-conditioned medium from the miscarriage samples exhibited high TNF-α levels and interfered with the decidualisation of ESCs. Exacerbated inflammation and elevated TNF-α levels at the feto-maternal interface may trigger AEA signalling pathways that, in turn, may impact decidualisation and the angiogenic ability of uNK cells. LARGE-SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION Primary uNK cell responses are based on a simple in vitro model. Thus, in complex microenvironments, such as the feto-maternal interface, the mechanisms may not be exactly the same. Also, the inflammatory events of miscarriage that, in this study, have happened prior to processing of the samples may cause different responses to that observed. In addition, the magnitude of the inflammatory response, required to trigger the AEA pathways that impact decidualisation and the uNK angiogenic ability in vivo, is still unclear. WIDER IMPLICATIONS OF THE FINDINGS The endocannabinoid AEA is a modulator of reproductive competence. AEA not only may contribute to neuroendocrine homeostasis but also can take part in uterine changes occurring during early pregnancy. STUDY FUNDING/COMPETING INTEREST(S) The work was supported by UID/MULTI/04378/2019 with funding from Fundação para a Ciência e a Tecnologia (FCT)/MCTES through national funds and PORTUGAL 2020 Partnership Agreement, NORTE-01-0145-FEDER-000024. S.C. Cunha acknowledges FCT for the IF/01616/2015 contract. There are no conflicts of interest.
Collapse
Affiliation(s)
- B M Fonseca
- UCIBIO, REQUIMTE, Laboratório de Bioquímica, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - S C Cunha
- LAQV, REQUIMTE, Laboratório de Bromatologia e Hidrologia, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - D Gonçalves
- Departamento da Mulher e da Medicina Reprodutiva, Serviço de Obstetrícia, Centro Materno-Infantil do Norte, Centro Hospitalar do Porto, Porto, Portugal
| | - A Mendes
- Departamento da Mulher e da Medicina Reprodutiva, Serviço de Obstetrícia, Centro Materno-Infantil do Norte, Centro Hospitalar do Porto, Porto, Portugal
| | - J Braga
- Departamento da Mulher e da Medicina Reprodutiva, Serviço de Obstetrícia, Centro Materno-Infantil do Norte, Centro Hospitalar do Porto, Porto, Portugal
| | - G Correia-da-Silva
- UCIBIO, REQUIMTE, Laboratório de Bioquímica, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - N A Teixeira
- UCIBIO, REQUIMTE, Laboratório de Bioquímica, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| |
Collapse
|
26
|
Intratumor Regulatory Noncytotoxic NK Cells in Patients with Hepatocellular Carcinoma. Cells 2021; 10:cells10030614. [PMID: 33802077 PMCID: PMC7999652 DOI: 10.3390/cells10030614] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 03/05/2021] [Accepted: 03/07/2021] [Indexed: 12/16/2022] Open
Abstract
Previous studies support the role of natural killer (NK) cells in controlling hepatocellular carcinoma (HCC) progression. However, ambiguity remains about the multiplicity and the role of different NK cell subsets, as a pro-oncogenic function has been suggested. We performed phenotypic and functional characterization of NK cells infiltrating HCC, with the corresponding nontumorous tissue and liver from patients undergoing liver resection for colorectal liver metastasis used as controls. We identified a reduced number of NK cells in tumors with higher frequency of CD56BRIGHTCD16- NK cells associated with higher expression of NKG2A, NKp44, and NKp30 and downregulation of NKG2D. Liver-resident (CXCR6+) NK cells were reduced in the tumors where T-bethiEomeslo expression was predominant. HCCs showed higher expression of CD49a with particular enrichment in CD49a+Eomes+ NK cells, a subset typically represented in the decidua and playing a proangiogenic function. Functional analysis showed reduced TNF-α production along with impaired cytotoxic capacity that was inversely related to CXCR6-, T-bethiEomeslo, and CD49a+Eomes+ NK cells. In conclusion, we identified a subset of NK cells infiltrating HCC, including non-liver-resident cells that coexpressed CD49a and Eomes and showed reduced cytotoxic potential. This NK cell subset likely plays a regulatory role in proangiogenic function.
Collapse
|
27
|
Molecular and immunological developments in placentas. Hum Immunol 2021; 82:317-324. [PMID: 33581928 DOI: 10.1016/j.humimm.2021.01.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/03/2021] [Accepted: 01/21/2021] [Indexed: 12/20/2022]
Abstract
Cytotrophoblasts differentiate in two directions during early placentation: syncytiotrophoblasts (STBs) and extravillous trophoblasts (EVTs). STBs face maternal immune cells in placentas, and EVTs, which invade the decidua and uterine myometrium, face the cells in the uterus. This situation, in which trophoblasts come into contact with maternal immune cells, is known as the maternal-fetal interface. Despite fetuses and fetus-derived trophoblast cells being of the semi-allogeneic conceptus, fetuses and placentas are not rejected by the maternal immune system because of maternal-fetal tolerance. The acquired tolerance develops during normal placentation, resulting in normal fetal development in humans. In this review, we introduce placental development from the viewpoint of molecular biology. In addition, we discuss how the disruption of placental development could lead to complications in pregnancy, such as hypertensive disorder of pregnancy, fetal growth restriction, or miscarriage.
Collapse
|
28
|
Dizaji Asl K, Velaei K, Rafat A, Tayefi Nasrabadi H, Movassaghpour AA, Mahdavi M, Nozad Charoudeh H. The role of KIR positive NK cells in diseases and its importance in clinical intervention. Int Immunopharmacol 2021; 92:107361. [PMID: 33429335 DOI: 10.1016/j.intimp.2020.107361] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/14/2020] [Accepted: 12/28/2020] [Indexed: 12/20/2022]
Abstract
Natural killer (NK) cells are essential for the elimination of the transformed and cancerous cells. Killer cell immunoglobulin-like receptors (KIRs) which expressed by T and NK cells, are key regulator of NK cell function. The KIR and their ligands, MHC class I (HLA-A, B and C) molecules, are highly polymorphic and their related genes are located on 19 q13.4 and 6 q21.3 chromosomes, respectively. It is clear that particular interaction between the KIRs and their related ligands can influence on the prevalence, progression and outcome of several diseases, like complications of pregnancy, viral infection, autoimmune diseases, and hematological malignancies. The mechanisms of immune signaling in particular NK cells involvement in causing pathological conditions are not completely understood yet. Therefore, better understanding of the molecular mechanism of KIR-MHC class I interaction could facilitate the treatment strategy of diseases. The present review focused on the main characteristics and functional details of various KIR and their combination with related ligands in diseases and also highlights ongoing efforts to manipulate the key checkpoints in NK cell-based immunotherapy.
Collapse
Affiliation(s)
- Khadijeh Dizaji Asl
- Stem Cell Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Kobra Velaei
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Rafat
- Stem Cell Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Tayefi Nasrabadi
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Akbar Movassaghpour
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Majid Mahdavi
- Department of Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | | |
Collapse
|
29
|
Lang F, Rajaxavier J, Singh Y, Brucker SY, Salker MS. The Enigmatic Role of Serum & Glucocorticoid Inducible Kinase 1 in the Endometrium. Front Cell Dev Biol 2020; 8:556543. [PMID: 33195190 PMCID: PMC7609842 DOI: 10.3389/fcell.2020.556543] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 09/24/2020] [Indexed: 11/13/2022] Open
Abstract
The serum- and glucocorticoid-inducible kinase 1 (SGK1) is subject to genetic up-regulation by diverse stimulators including glucocorticoids, mineralocorticoids, dehydration, ischemia, radiation and hyperosmotic shock. To become active, the expressed kinase requires phosphorylation, which is accomplished by PI3K/PDK1 and mTOR dependent signaling. SGK1 enhances the expression/activity of various transport proteins including Na+/K+-ATPase as well as ion-, glucose-, and amino acid- carriers in the plasma membrane. SGK1 can further up-regulate diverse ion channels, such as Na+-, Ca2+-, K+- and Cl- channels. SGK1 regulates expression/activity of a wide variety of transcription factors (such as FKHRL1/Foxo3a, β-catenin, NFκB and p53). SGK1 thus contributes to the regulation of transport, glycolysis, angiogenesis, cell survival, immune regulation, cell migration, tissue fibrosis and tissue calcification. In this review we summarized the current findings that SGK1 plays a crucial function in the regulation of endometrial function. Specifically, it plays a dual role in the regulation of endometrial receptivity necessary for implantation and, subsequently in pregnancy maintenance. Furthermore, fetal programming of blood pressure regulation requires maternal SGK1. Underlying mechanisms are, however, still ill-defined and there is a substantial need for additional information to fully understand the role of SGK1 in the orchestration of embryo implantation, embryo survival and fetal programming.
Collapse
Affiliation(s)
- Florian Lang
- Department of Physiology, Eberhard-Karls University, Tübingen, Germany
| | - Janet Rajaxavier
- Research Institute of Women’s Health, Eberhard-Karls University, Tübingen, Germany
| | - Yogesh Singh
- Research Institute of Women’s Health, Eberhard-Karls University, Tübingen, Germany
- Institute of Medical Genetics and Applied Genomics, Eberhard-Karls University, Tübingen, Germany
| | - Sara Y. Brucker
- Research Institute of Women’s Health, Eberhard-Karls University, Tübingen, Germany
| | - Madhuri S. Salker
- Research Institute of Women’s Health, Eberhard-Karls University, Tübingen, Germany
| |
Collapse
|
30
|
Guerrero B, Hassouneh F, Delgado E, Casado JG, Tarazona R. Natural killer cells in recurrent miscarriage: An overview. J Reprod Immunol 2020; 142:103209. [PMID: 32992208 DOI: 10.1016/j.jri.2020.103209] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 07/31/2020] [Accepted: 09/14/2020] [Indexed: 12/20/2022]
Abstract
Recurrent Miscarriage is an early pregnancy complication which affects about 1-3 % of child-bearing couples. The mechanisms involved in the occurrence of recurrent miscarriages are not clearly understood. In the last decade Natural Killer cells have been studied in peripheral blood and uterus in order to determine if there are specific characteristics of Natural Killer cells associated with miscarriage. Different authors have described an increased number of uterine and peripheral blood Natural Killer cells in women with recurrent miscarriages compared to control women. However, its relationship with miscarriage has not been confirmed. In patients with recurrent miscarriage a lack of inhibition of decidua Natural Killer cells can be observed, which leads to a more activated state characterized by higher levels of proinflammatory cytokines. In peripheral blood, it has been also reported a dysfunctional cytokine production by Natural Killer cells, with an increase of interferon-γ levels and a decrease of Interleukin-4. Significant progress has been made in the last decade in understanding the biology of Natural Killer cells, including the identification of new receptors that also contribute to the activation and regulation of Natural Killer cells. In this review, we summarize the current progress in the study of Natural Killer cells in recurrent miscarriage.
Collapse
Affiliation(s)
| | | | - Elena Delgado
- Clínica Norba, Ginecología y Reproducción, Cáceres, Spain
| | - Javier G Casado
- Stem Cell Therapy Unit, Jesús Usón Minimally Invasive Surgery Centre, Cáceres, Spain
| | | |
Collapse
|
31
|
Lin F, Yang C, Feng T, Yang S, Zhou R, Li H. The Maternal-Fetal Interface in Small-for-Gestational-Age Pregnancies Is Associated With a Reduced Quantity of Human Decidual NK Cells With Weaker Functional Ability. Front Cell Dev Biol 2020; 8:633. [PMID: 33015028 PMCID: PMC7509437 DOI: 10.3389/fcell.2020.00633] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 06/24/2020] [Indexed: 12/17/2022] Open
Abstract
Small for gestational age (SGA) refers to a birth weight that is less than the 10th percentile of the mean weight of infants at the same gestational age. This condition is associated with a variety of complications, and a high risk of cardiovascular and cerebrovascular diseases in adulthood. Decidual natural killer (dNK) cells at the maternal–fetal interface have received significant research attention in terms of normal pregnancy or miscarriage; however, data relating to SGA are limited. In this study, we aimed to investigate the characteristics and regulatory role of dNK cells at the maternal–fetal interface in SGA. Using immunofluorescence assays, we found that dNK cells maintained close contact with extra-villous trophoblasts, and the proportion of dNK cells in SGA decreased more than in appropriate for gestational age (AGA). Flow cytometry also showed that there was a significantly lower percentage of dNK cells in SGA (25.01 ± 2.43%) than in AGA (34.25 ± 2.30%) (p = 0.0103). The expression of the inhibitory receptor NKG2A on dNK cells and the secretion levels of both perforin and TGF-β1 from dNK cells were significantly higher in SGA than in AGA, while the cytotoxicity of dNK cells in SGA against K562 cells was attenuated. Compared to AGA, the functional ability of dNK cells in SGA showed significant functional impairment in promoting proliferation, migration, invasion, and tube formation in trophoblast cells or vascular endothelial cells. The abnormal function of dNK cells may affect fetal growth and development, and could therefore participate in the pathogenesis of SGA.
Collapse
Affiliation(s)
- Fang Lin
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Chuan Yang
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Ting Feng
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Shuo Yang
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Rong Zhou
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Hong Li
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
32
|
Huber WJ, Sauerbrun-Cutler MT, Krueger PM, Lambert-Messerlian G, Sharma S. Human chorionic gonadotropin-mediated modulation of pregnancy-compatible peripheral blood natural killer cells in frozen embryo transfer cycles. Am J Reprod Immunol 2020; 85:e13324. [PMID: 33245601 DOI: 10.1111/aji.13324] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/10/2020] [Accepted: 08/12/2020] [Indexed: 12/13/2022] Open
Abstract
PROBLEM To evaluate pregnancy-compatible phenotypic and functional changes in peripheral blood natural killer (pNK) cells during frozen embryo transfer (FET) cycles. METHOD OF STUDY Peripheral blood was collected from patients undergoing frozen embryo transfer cycles at three separate time points in the cycle. pNK cell phenotype was analyzed by flow cytometry. Impact of pregnancy status on pNK cell cytotoxicity was characterized by two methods: (1) a three-dimensional endovascular tube formation approach and (2) a NK cell-specific K562 cell kill assay. RESULTS A total of 35 patients were enrolled, 15 with clinical pregnancies and 20 with negative serum β-hCG levels. Overall percentage of CD45+ CD3- CD56+ pNK cell did not change during the FET cycle. Pregnancy resulted in an increase in CD45+ CD3- CD56+ pNK cell population on the day of serum β-hCG. pNK cells from non-pregnant patients caused significant tube disruption when compared to pregnant patients. Addition of serum from pregnant women reduced the tube disruption by pNK cells from non-pregnant patients. pNK cells from pregnant patients showed significantly lower cytotoxicity toward K562 cells in serum-free conditions. The addition of pregnancy serum decreased non-pregnant pNK cell cytotoxicity. Pregnancy status had no impact on VEGF-A and VEGF-C serum levels. Recombinant hCG added to non-pregnant serum resulted in a significant reduction in non-pregnant pNK cell-mediated K562 cell kill. CONCLUSION There was no difference in pNK cell populations based on timing of the FET cycle. However, pregnancy increased the percentage of CD45+ CD3- CD56+ pNK cells. Additionally, pNK cells from pregnant women have reduced cytotoxicity and this is possibly mediated by hCG.
Collapse
Affiliation(s)
- Warren J Huber
- Department of Obstetrics and Gynecology, Women & Infants Hospital, Alpert Medical School of Brown University, Providence, RI, USA
| | - May-Tal Sauerbrun-Cutler
- Department of Obstetrics and Gynecology, Women & Infants Hospital, Alpert Medical School of Brown University, Providence, RI, USA
| | - Paula M Krueger
- Department of Pediatrics, Women & Infants Hospital, Alpert Medical School of Brown University, Providence, RI, USA
| | - Geralyn Lambert-Messerlian
- Department of Obstetrics and Gynecology, Women & Infants Hospital, Alpert Medical School of Brown University, Providence, RI, USA.,Department of Pathology and Laboratory Medicine, Women & Infants Hospital, Alpert Medical School of Brown University, Providence, RI, USA
| | - Surendra Sharma
- Department of Pediatrics, Women & Infants Hospital, Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
33
|
Yang X, Yang E, Wang WJ, He Q, Jubiz G, Katukurundage D, Dambaeva S, Beaman K, Kwak-Kim J. Decreased HLA-C1 alleles in couples of KIR2DL2 positive women with recurrent pregnancy loss. J Reprod Immunol 2020; 142:103186. [PMID: 32846355 DOI: 10.1016/j.jri.2020.103186] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 08/03/2020] [Accepted: 08/08/2020] [Indexed: 12/11/2022]
Abstract
Specific killer cell immunoglobulin-like receptor (KIR) in women with recurrent pregnancy loss (RPL) and HLA ligands in couples invoke a susceptibility to RPL. However, the relationship between KIR2DL2 and its cognate ligand HLA-C1 has not been explored. In this prospective cohort study, 160 Caucasian women with RPL and 99 partners were included. KIR/HLA-C typing, NK assay, Th1/Th2 intracellular cytokine ratios, 25-(OH)-vitamin D level, and the presence of autoantibodies were analyzed. KIR2DL2 positive women (P = 0.023) and their partners (P = 0.017) had lower allele frequencies of HLA-C1 than those of KIR2DL2 negative women. KIR2DL2 positive women had significantly lower genotype frequency of HLA-C1C1 as compared to the North American Caucasian population controls (P < 0.05). In the partners of KIR2DL2 positive women, there was a substantially higher frequency of HLA-C2C2 than controls (P = 0.016). Besides, KIR2DL2 negative women had a higher prevalence of anti-ssDNA antibody as compared with that of KIR2DL2 positive women (P = 0.043). There were no differences in the distribution of HLA-C genotypes based on KIR2DL2, regardless of pregnancy outcome in women with RPL and their partners while on immunomodulation treatment. In conclusion, decreased ligands for inhibitory KIRs (inhKIR) could lead to insufficient inhibition of maternal uterine NK cells toward the trophoblast, thereby contributing to the pathogenesis of RPL. Specific KIR and HLA-C genotyping may predict the reproductive outcome of women with RPL.
Collapse
Affiliation(s)
- Xiuhua Yang
- Reproductive Medicine and Immunology, Department of Obstetrics and Gynecology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, Vernon Hills, 60061, IL, USA; Clinical Immunology Laboratory, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, 60064, IL, USA; Department of Obstetrics, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China
| | - Ellen Yang
- Reproductive Medicine and Immunology, Department of Obstetrics and Gynecology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, Vernon Hills, 60061, IL, USA; Clinical Immunology Laboratory, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, 60064, IL, USA
| | - Wen-Juan Wang
- Reproductive Medicine and Immunology, Department of Obstetrics and Gynecology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, Vernon Hills, 60061, IL, USA; Clinical Immunology Laboratory, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, 60064, IL, USA
| | - Qiaohua He
- Reproductive Medicine and Immunology, Department of Obstetrics and Gynecology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, Vernon Hills, 60061, IL, USA; Clinical Immunology Laboratory, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, 60064, IL, USA
| | - Giovanni Jubiz
- Reproductive Medicine and Immunology, Department of Obstetrics and Gynecology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, Vernon Hills, 60061, IL, USA; Clinical Immunology Laboratory, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, 60064, IL, USA
| | - Dimantha Katukurundage
- Clinical Immunology Laboratory, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, 60064, IL, USA
| | - Svetlana Dambaeva
- Clinical Immunology Laboratory, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, 60064, IL, USA
| | - Kenneth Beaman
- Clinical Immunology Laboratory, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, 60064, IL, USA
| | - Joanne Kwak-Kim
- Reproductive Medicine and Immunology, Department of Obstetrics and Gynecology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, Vernon Hills, 60061, IL, USA; Clinical Immunology Laboratory, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, 60064, IL, USA.
| |
Collapse
|
34
|
Munir H, Mazzaglia C, Shields JD. Stromal regulation of tumor-associated lymphatics. Adv Drug Deliv Rev 2020; 161-162:75-89. [PMID: 32783989 DOI: 10.1016/j.addr.2020.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 07/27/2020] [Accepted: 08/06/2020] [Indexed: 02/08/2023]
Abstract
Recent advances have identified a growing array of roles played by lymphatics in the tumor microenvironment, from providing a route of metastasis to immune modulation. The tumor microenvironment represents an exceptionally complex, dynamic niche comprised of a diverse mixture of cancer cells and normal host cells termed the stroma. This review discusses our current understanding of stromal elements and how they regulate lymphatic growth and functional properties in the tumor context.
Collapse
Affiliation(s)
- Hafsa Munir
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Box 197 Cambridge Biomedical Campus, Cambridge, CB2 0XZ
| | - Corrado Mazzaglia
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Box 197 Cambridge Biomedical Campus, Cambridge, CB2 0XZ
| | - Jacqueline D Shields
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Box 197 Cambridge Biomedical Campus, Cambridge, CB2 0XZ.
| |
Collapse
|
35
|
Guo F, Cui J. Anti-angiogenesis: Opening a new window for immunotherapy. Life Sci 2020; 258:118163. [PMID: 32738363 DOI: 10.1016/j.lfs.2020.118163] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 07/14/2020] [Accepted: 07/25/2020] [Indexed: 12/31/2022]
Abstract
The tumor microenvironment (TME) provides a guarantee for the survival and development of solid tumors. In recent years, treatment strategies for TME have set off a great upsurge in the field of cancer research. Tumor angiogenesis and tumor immune microenvironment are two important research branches of TME, and antiangiogenic therapy and immunotherapy have gradually become one important focus of cancer treatment research. More interestingly, increasing number of studies have indicated that there are complex regulatory interactions between the two treatment strategies, with multiple regulatory mechanisms involved. Based on these findings, clinical studies on the combination of immunotherapy and antiangiogenic therapy have also been carried out gradually. This combination strategy has shown good results in many types of tumors, but it also faces many challenges. The paper analysed the potential mechanism of the immunotherapy and antiangiogenic therapy combination, discussed the latest significant clinical trial progress and the existing challenges and problems, aiming to offer some available insights on the effective clinical application of this combination pattern.
Collapse
Affiliation(s)
- Feifei Guo
- Cancer Center, The First Hospital of Jilin University, Changchun 130021, China
| | - Jiuwei Cui
- Cancer Center, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
36
|
Bozorgmehr M, Gurung S, Darzi S, Nikoo S, Kazemnejad S, Zarnani AH, Gargett CE. Endometrial and Menstrual Blood Mesenchymal Stem/Stromal Cells: Biological Properties and Clinical Application. Front Cell Dev Biol 2020; 8:497. [PMID: 32742977 PMCID: PMC7364758 DOI: 10.3389/fcell.2020.00497] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/25/2020] [Indexed: 12/11/2022] Open
Abstract
A highly proliferative mesenchymal stem/stromal cell (MSC) population was recently discovered in the dynamic, cyclically regenerating human endometrium as clonogenic stromal cells that fulfilled the International Society for Cellular Therapy (ISCT) criteria. Specific surface markers enriching for clonogenic endometrial MSC (eMSC), CD140b and CD146 co-expression, and the single marker SUSD2, showed their perivascular identity in the endometrium, including the layer which sheds during menstruation. Indeed, cells with MSC properties have been identified in menstrual fluid and commonly termed menstrual blood stem/stromal cells (MenSC). MenSC are generally retrieved from menstrual fluid as plastic adherent cells, similar to bone marrow MSC (bmMSC). While eMSC and MenSC share several biological features with bmMSC, they also show some differences in immunophenotype, proliferation and differentiation capacities. Here we review the phenotype and functions of eMSC and MenSC, with a focus on recent studies. Similar to other MSC, eMSC and MenSC exert immunomodulatory and anti-inflammatory impacts on key cells of the innate and adaptive immune system. These include macrophages, T cells and NK cells, both in vitro and in small and large animal models. These properties suggest eMSC and MenSC as additional sources of MSC for cell therapies in regenerative medicine as well as immune-mediated disorders and inflammatory diseases. Their easy acquisition via an office-based biopsy or collected from menstrual effluent makes eMSC and MenSC attractive sources of MSC for clinical applications. In preparation for clinical translation, a serum-free culture protocol was established for eMSC which includes a small molecule TGFβ receptor inhibitor that prevents spontaneous differentiation, apoptosis, senescence, maintains the clonogenic SUSD2+ population and enhances their potency, suggesting potential for cell-therapies and regenerative medicine. However, standardization of MenSC isolation protocols and culture conditions are major issues requiring further research to maximize their potential for clinical application. Future research will also address crucial safety aspects of eMSC and MenSC to ensure these protocols produce cell products free from tumorigenicity and toxicity. Although a wealth of data on the biological properties of eMSC and MenSC has recently been published, it will be important to address their mechanism of action in preclinical models of human disease.
Collapse
Affiliation(s)
- Mahmood Bozorgmehr
- Reproductive Immunology Research Center, Avicenna Research Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Shanti Gurung
- Centre for Reproductive Health, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Saeedeh Darzi
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Shohreh Nikoo
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Somaieh Kazemnejad
- Nanobitechnology Research Center, Avicenna Research Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| | - Amir-Hassan Zarnani
- Reproductive Immunology Research Center, Avicenna Research Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Caroline E. Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
37
|
Abstract
Pregnancy is a natural process that poses an immunological challenge because non-self fetus must be accepted. During the pregnancy period, the fetus as 'allograft' inherits maternal and also paternal antigens. For successful and term pregnancy, the fetus is tolerated and nurtured enjoying immune privileges that minimize the risk of being rejected by maternal immune system. Multiple mechanisms contribute to tolerate the semi-allogeneic fetus. Here, we summarize the recent progresses on how the maternal immune system actively collaborates to maintain the immune balance and maternal-fetal tolerance.
Collapse
Affiliation(s)
- Xiaopeng Li
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.,Guangdong Engineering and Technology Research Center of Maternal-Fetal Medicine, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.,CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Jiayi Zhou
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.,Guangdong Engineering and Technology Research Center of Maternal-Fetal Medicine, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Min Fang
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.,CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,International College, University of the Chinese Academy of Sciences, Beijing, China
| | - Bolan Yu
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.,Guangdong Engineering and Technology Research Center of Maternal-Fetal Medicine, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
38
|
Murata H, Tanaka S, Tsuzuki-Nakao T, Kido T, Kakita-Kobayashi M, Kida N, Hisamatsu Y, Tsubokura H, Hashimoto Y, Kitada M, Okada H. The transcription factor HAND2 up-regulates transcription of the IL15 gene in human endometrial stromal cells. J Biol Chem 2020; 295:9596-9605. [PMID: 32444497 DOI: 10.1074/jbc.ra120.012753] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 05/18/2020] [Indexed: 12/26/2022] Open
Abstract
Cyclic changes of the human endometrium, such as proliferation, secretion, and decidualization, occur during regular menstrual cycles. Heart- and neural crest derivatives-expressed transcript 2 (HAND2) is a key transcription factor in progestin-induced decidualization of human endometrial stromal cells (ESCs). It has been suggested that HAND2 regulates interleukin 15 (IL15), a key immune factor required for the activation and survival of uterine natural killer (uNK) cells. Activated uNK cells can promote spiral artery remodeling and secrete cytokines to induce immunotolerance. To date, no studies have evaluated the transcription factors that regulate IL15 expression in human ESCs. In the present study, we examined whether HAND2 controls IL15 transcriptional regulation in human ESCs. Quantitative RT-PCR and histological analyses revealed that HAND2 and IL15 levels increase considerably in the secretory phase of human endometrium tissues. Results from ChIP-quantitative PCR suggested that HAND2 binds to a putative HAND2 motif, which we identified in the upstream region of the human IL15 gene through in silico analysis. Using a luciferase reporter assay, we found that the upstream region of the human IL15 gene up-regulates reporter gene activities in response to estradiol and a progestin representative (medroxyprogesterone) in ESCs. The upstream region of the human IL15 gene also exhibited increasing responsiveness to transfection with a HAND2 expression vector. Of note, deletion and substitution variants of the putative HAND2 motif in the upstream region of IL15 did not respond to HAND2 transfection. These findings confirm that HAND2 directly up-regulates human IL15 transcription in ESCs.
Collapse
Affiliation(s)
- Hiromi Murata
- Department of Obstetrics and Gynecology, Kansai Medical University, Osaka, Japan
| | - Susumu Tanaka
- Department of Anatomy, Kansai Medical University, Osaka, Japan
| | - Tomoko Tsuzuki-Nakao
- Department of Obstetrics and Gynecology, Kansai Medical University, Osaka, Japan
| | - Takeharu Kido
- Department of Obstetrics and Gynecology, Kansai Medical University, Osaka, Japan
| | | | - Naoko Kida
- Department of Obstetrics and Gynecology, Kansai Medical University, Osaka, Japan
| | - Yoji Hisamatsu
- Department of Obstetrics and Gynecology, Kansai Medical University, Osaka, Japan
| | - Hiroaki Tsubokura
- Department of Obstetrics and Gynecology, Kansai Medical University, Osaka, Japan
| | - Yoshiko Hashimoto
- Department of Obstetrics and Gynecology, Kansai Medical University, Osaka, Japan
| | - Masaaki Kitada
- Department of Anatomy, Kansai Medical University, Osaka, Japan
| | - Hidetaka Okada
- Department of Obstetrics and Gynecology, Kansai Medical University, Osaka, Japan
| |
Collapse
|
39
|
Du M, Wang W, Huang L, Guan X, Lin W, Yao J, Li L. Natural killer cells in the pathogenesis of preeclampsia: a double-edged sword. J Matern Fetal Neonatal Med 2020; 35:1028-1035. [PMID: 32188324 DOI: 10.1080/14767058.2020.1740675] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Objective: To investigate the relationship between natural killer (NK) cells, extravillous trophoblast cells (EVTs) and vessel remodeling in early human pregnancy, and the association between NK cells and preeclampsia (PE) in late human pregnancy.Methods: Human decidual tissues from women with normal pregnancies were collected and examined for the relationship of NK cells with uterine vessel remodeling using immunohistochemistry. Percentages of peripheral blood NK (pNK) and decidual NK (dNK) cells and the levels of intracellular interferon (IFN)-γ, perforin and granzyme B in normal pregnancies, late-onset and early-onset PE were analyzed using flow cytometry. Cytolytic functions of dNK cells from normal and PE pregnancies were examined. Effects of conditioned medium (CM) of dNK cells from normal and PE pregnancies on first trimester trophoblast invasion and migration were tested.Results: In early pregnancy samples (9-13 weeks of gestation), we noted moderate vessel remodeling with abundant perivascular NK cells but a limited number of surrounding EVTs. The numbers of both human pNK cells and dNK cells and intracellular interferon (IFN)-γ, perforin and granzyme B production were significantly higher in PE compared with normal pregnancies at the time of delivery for both early- and late-onset disease. dNK cells from PE pregnancies not only killed first trimester trophoblasts but also inhibited their invasion and migration when compared to normal controls.Conclusion: Our results suggest that NK cells, in conjunction with EVTs, may play an important role in controlling uterine SA remodeling at the early stages of vessel remodeling, but they contribute to the pathogenesis of PE in late pregnancy.
Collapse
Affiliation(s)
- Mingyue Du
- Department of Obstetrics and Gynecology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Weijia Wang
- Department of Obstetrics and Gynecology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Lina Huang
- Department of Obstetrics and Gynecology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Xiuting Guan
- Department of Obstetrics and Gynecology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Wen Lin
- Department of Obstetrics and Gynecology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Jingxin Yao
- Department of Obstetrics and Gynecology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Liping Li
- Department of Obstetrics and Gynecology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| |
Collapse
|
40
|
Abstract
Brazilian-born British biologist Dr. Peter Medawar played an integral role in developing the concepts of immunologic rejection and tolerance, which led to him receiving the Nobel Prize "for the discovery of acquired immunologic tolerance" and eventually made organ transplantation a reality. However, at the time of his early work in tolerance, a paradox to his theories was brought to his attention; how was pregnancy possible? Pregnancy resembles organ transplantation in that the fetus, possessing paternal antigens, is a semi-allogeneic graft that can survive without immunosuppression for 9 months. To answer this question, Medawar proposed three hypotheses of how a mother supports her fetus in utero, now known as "Medawar's Paradox." The mechanisms that govern fetomaternal tolerance are still incompletely understood but may provide critical insight into how to achieve immune tolerance in organ transplantation. Here, we review current understanding of the immune factors responsible for fetomaternal tolerance during pregnancy and discuss the potential implications for advances in transplantation science.
Collapse
Affiliation(s)
- Victoria Rendell
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Natalie M Bath
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Todd V Brennan
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
41
|
Frontier Progress in the Establishment of Trophoblast Stem Cell and the Identification of New Cell Subtypes at the Maternal-Fetal Interface. MATERNAL-FETAL MEDICINE 2019. [DOI: 10.1097/fm9.0000000000000023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
42
|
Lu H, Hu R. The role of immunity in the pathogenesis and development of pre‐eclampsia. Scand J Immunol 2019; 90:e12756. [PMID: 30739345 DOI: 10.1111/sji.12756] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 01/25/2019] [Accepted: 02/05/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Hui‐Qing Lu
- Hospital of Obstetrics and Gynecology Fudan University Shanghai China
| | - Rong Hu
- Hospital of Obstetrics and Gynecology Fudan University Shanghai China
| |
Collapse
|
43
|
Shokri MR, Bozorgmehr M, Ghanavatinejad A, Falak R, Aleahmad M, Kazemnejad S, Shokri F, Zarnani AH. Human menstrual blood-derived stromal/stem cells modulate functional features of natural killer cells. Sci Rep 2019; 9:10007. [PMID: 31292483 PMCID: PMC6620360 DOI: 10.1038/s41598-019-46316-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 06/25/2019] [Indexed: 12/13/2022] Open
Abstract
Although natural killer (NK) cells play a crucial role in the maintenance of a successful pregnancy, their cytotoxic activity should be tightly controlled. We hypothesized that endometrial mesenchymal stromal/stem cells (eMSCs) could potentially attenuate the functional features of NK cells. Herein, we assessed immunomodulatory effects of menstrual blood-derived stromal/stem cells (MenSCs), as a surrogate for eMSCs, on NK cells function. Our results showed that MenSCs induced proliferation of NK cells. However, IFN-γ/IL-1β pretreated MenSCs significantly inhibited NK cell proliferation. Of 41 growth factors tested, MenSCs produced lower levels of insulin-like growth factor binding proteins (IGFBPs) 1-4, VEGF-A, β-NGF, and M-CSF compared to bone marrow-derived mesenchymal stem cells (BMSCs). MenSCs displayed high activity of IDO upon IFN-γ treatment. The antiproliferative potential of IFN-γ/IL-1β-pretreated MenSCs was mediated through IL-6 and TGF-β. MenSCs impaired the cytotoxic activity of NK cells on K562 cells, consistent with the lower expression of perforin, granzymes A, and B. We also observed that in vitro decidualization of MenSCs in the presence of IFN-γ reduced the inhibitory effect of MenSCs on NK cell cytotoxicity against K562 target cells. Additionally, MenSCs were found to be prone to NK cell-mediated lysis in an MHC-independent manner. Our findings imply that dysregulation of NK cells in such pregnancy-related disorders as miscarriage may stem from dysfunctioning of eMSCs.
Collapse
Affiliation(s)
- Mohammad-Reza Shokri
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmood Bozorgmehr
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Reproductive Immunology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Alireza Ghanavatinejad
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Falak
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Mehdi Aleahmad
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Somaieh Kazemnejad
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Fazel Shokri
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir-Hassan Zarnani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
- Reproductive Immunology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran.
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
44
|
Jha SK, Rauniyar K, Chronowska E, Mattonet K, Maina EW, Koistinen H, Stenman UH, Alitalo K, Jeltsch M. KLK3/PSA and cathepsin D activate VEGF-C and VEGF-D. eLife 2019; 8:44478. [PMID: 31099754 PMCID: PMC6588350 DOI: 10.7554/elife.44478] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 05/16/2019] [Indexed: 11/13/2022] Open
Abstract
Vascular endothelial growth factor-C (VEGF-C) acts primarily on endothelial cells, but also on non-vascular targets, for example in the CNS and immune system. Here we describe a novel, unique VEGF-C form in the human reproductive system produced via cleavage by kallikrein-related peptidase 3 (KLK3), aka prostate-specific antigen (PSA). KLK3 activated VEGF-C specifically and efficiently through cleavage at a novel N-terminal site. We detected VEGF-C in seminal plasma, and sperm liquefaction occurred concurrently with VEGF-C activation, which was enhanced by collagen and calcium binding EGF domains 1 (CCBE1). After plasmin and ADAMTS3, KLK3 is the third protease shown to activate VEGF-C. Since differently activated VEGF-Cs are characterized by successively shorter N-terminal helices, we created an even shorter hypothetical form, which showed preferential binding to VEGFR-3. Using mass spectrometric analysis of the isolated VEGF-C-cleaving activity from human saliva, we identified cathepsin D as a protease that can activate VEGF-C as well as VEGF-D. The lymphatic system is composed of networks of vessels that drain fluids from the body’s tissues and filter it back into the blood. Growing these vessels depends on a factor known as VEGF-C, which is released in an inactive form and must be cut by enzymes before it can work. One enzyme that is known to activate the VEGF-C signal when the early embryo is developing is ADAMTS3. If this signal fails to switch on this can result in a condition known as lymphedema – whereby problems in the lymphatic system cause tissues to swell due to insufficient drainage. However, it is unknown whether the VEGF-C signal can be activated by enzymes other than ADAMTS3. To investigate this Jha, Rauniyar et al. tested a specific family of proteins commonly found in the human prostate, which have previously been predicted to act on VEGF-C. This revealed that the lymphatic vessel growth factor can also be activated by an enzyme found in seminal fluid called prostate specific antigen, or PSA for short. To see if enzymes in other bodily fluids could switch on VEGF-C, different components of human saliva were separated and tested to see which could cut inactive VEGF-C. This showed that VEGF-C could be converted to an active form by another enzyme called cathepsin D. Unexpectedly, Jha, Rauniyar et al. found that VEGF-C was also present in semen. For conception to occur PSA must liquify the semen following ejaculation. It was discovered that PSA activates VEGF-C just as the semen starts to liquify, suggesting that the lymphatic vessel growth factor might also play an important role in reproduction. In addition to VEGF-C, both PSA and cathepsin D were found to activate another growth factor called VEGF-D, which has an unknown role in the human body. VEGF-C helps the spread of tumors, and blocking the two enzymes that activate this growth factor may be a new therapeutic approach for cancer. However, more work is needed to validate which types of tumor, if any, use these enzymes to activate VEGF-C. In addition, understanding the relationship between PSA and VEGF-C could help improve our knowledge of human reproduction.
Collapse
Affiliation(s)
- Sawan Kumar Jha
- Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland.,Wihuri Research Institute, Helsinki, Finland
| | - Khushbu Rauniyar
- Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
| | - Ewa Chronowska
- Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland.,Jagiellonian University Medical College, Cracow, Poland
| | - Kenny Mattonet
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Eunice Wairimu Maina
- Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland
| | - Hannu Koistinen
- Department of Clinical Chemistry, University of Helsinki, Helsinki, Finland.,Helsinki University Hospital, Helsinki, Finland
| | - Ulf-Håkan Stenman
- Department of Clinical Chemistry, University of Helsinki, Helsinki, Finland.,Helsinki University Hospital, Helsinki, Finland
| | - Kari Alitalo
- Wihuri Research Institute, Helsinki, Finland.,Helsinki University Hospital, Helsinki, Finland.,Translational Cancer Medicine Research Program, University of Helsinki, Helsinki, Finland
| | - Michael Jeltsch
- Individualized Drug Therapy Research Program, University of Helsinki, Helsinki, Finland.,Wihuri Research Institute, Helsinki, Finland
| |
Collapse
|
45
|
Ander SE, Diamond MS, Coyne CB. Immune responses at the maternal-fetal interface. Sci Immunol 2019; 4:eaat6114. [PMID: 30635356 PMCID: PMC6744611 DOI: 10.1126/sciimmunol.aat6114] [Citation(s) in RCA: 394] [Impact Index Per Article: 65.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 11/29/2018] [Indexed: 12/13/2022]
Abstract
Pregnancy poses an immunological challenge because a genetically distinct (nonself) fetus must be supported within the pregnant female for the required gestational period. Placentation, or the establishment of the fetally derived placenta, is a common strategy used by eutherian mammals to protect the fetus and promote its growth. However, the substantial morphological differences of the placental architecture among species suggest that the process of placentation results from convergent evolution. Although there are considerable similarities in placental function across placental mammals, there are important differences that arise owing to species-specific immunological (and other biological) constraints. This Review focuses on the immunological similarities and differences that occur at the maternal-fetal interface in the context of human and mouse pregnancies. We discuss how the decidua and placenta of these different species form key immunological barriers that sustain maternal tolerance yet generate innate immune responses that prevent microbial infections.
Collapse
Affiliation(s)
- Stephanie E Ander
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
- Center for Microbial Pathogenesis, University of Pittsburgh Medical Center (UPMC) Children's Hospital of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carolyn B Coyne
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.
- Center for Microbial Pathogenesis, University of Pittsburgh Medical Center (UPMC) Children's Hospital of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
- R. K. Mellon Pediatric Research Institute, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA 15219, USA
| |
Collapse
|
46
|
Złotkowska A, Adamczyk S, Andronowska A. Presence of trophoblast in the uterine lumen affects VEGF-C expression in porcine endometrium. Theriogenology 2018; 125:216-223. [PMID: 30471615 DOI: 10.1016/j.theriogenology.2018.11.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 10/24/2018] [Accepted: 11/13/2018] [Indexed: 01/15/2023]
Abstract
Early pregnancy is associated with morphological and functional changes within the uterus, accompanied by angiogenesis, increased vascular permeability and activation of immune tolerance. Intensive angiogenesis leads to accelerated vascular leakage and accumulation of interstitial fluid in endometrium. To protect the trophoblast from the harmful effect of extracellular fluid, process known as lymphangiogenesis is crucial. These studies are focused on VEGF-C, factor responsible of lymphatic vessels creating, and its receptors: Flk1 (VEGFR2) and Flt4 (VEGFR3) during the time of implantation as well as the effect of trophoblast signals (IFNG and E2) on VEGF-C production. Endometrial samples were collected from mature gilts from days 8, 10, 12, 14 of estrous cycle and pregnancy. Real-Time PCR analysis revealed increased mRNA expression of VEGF-C on days 10, 12, 14 of pregnancy compared to corresponding days of estrous cycle. The highest VEGF-C mRNA expression was observed on 14 day of pregnancy (p < 0.05). Increased mRNA expression of Flk1 and Flt4 was noticed on day 14 of pregnancy in comparison to day 10. Enhanced Flk1 mRNA expression during 14 day of pregnancy was observed compared to corresponding day of estrous cycle (p < 0.05). No significant difference on the protein level was revealed. VEGF-C and its receptors were localized mainly in luminal and glandular epithelial cells, but their presence were confirmed also in endothelial cells of blood and lymphatic vessels and 14 d trophoblasts. In vitro studies revealed positive effect of IFNG on VEGF-C mRNA expression in stromal cells and protein content in medium after stromal cells culture (p < 0.05). Our studies demonstrated the presence of VEGF-C system in porcine endometrium and indicated its possible important role during the time of implantation.
Collapse
Affiliation(s)
- A Złotkowska
- Department of Hormonal Action Mechanisms, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - S Adamczyk
- Department of Hormonal Action Mechanisms, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - A Andronowska
- Department of Hormonal Action Mechanisms, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland.
| |
Collapse
|
47
|
Li D, Li J, Jia B, Wang Y, Zhang J, Liu G. Genome-wide identification of microRNAs in decidual natural killer cells from patients with unexplained recurrent spontaneous abortion. Am J Reprod Immunol 2018; 80:e13052. [PMID: 30339301 DOI: 10.1111/aji.13052] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 08/19/2018] [Accepted: 09/04/2018] [Indexed: 12/17/2022] Open
Abstract
PROBLEM This study revealed miRNA regulation and functional microarray-based profiles of miRNAs in the natural killer (NK) cells of the decidual tissue obtained from patients with unexplained recurrent spontaneous abortion (URSA). METHOD OF STUDY Patients with URSA were categorized based on the occurrence of at least two or more successive spontaneous abortions between 7th and 10th gestational week. Total RNA was isolated from the NK cells of the decidual tissue obtained from patients with induced abortion at about the 8th gestational week. The potential contribution of regulatory miRNAs to a genetic predisposition to URSA was characterized by comparison with healthy and fertile controls and bioinformatics analyses. RESULTS Analysis of the miRNA expression profiles identified 50 miRNAs that were differentially expressed, including one down-regulated and 49 up-regulated miRNAs in the URSA group. MiRNA-Gene-Network, miRNA-GO-Network and miRNA-Gene-TF-Network were constructed. The key miRNAs, genes, GOs and core TFs in the network were determined. CONCLUSION Our results suggest that a close relationship exists between the aberrant miRNAs and URSA. Furthermore, these findings support the notion that altered expression of miRNAs may contribute to the clinical diagnosis of URSA and the potential to develop novel strategies for therapeutic targets against URSA.
Collapse
Affiliation(s)
- Dandan Li
- Department of Obstetrics and Gynecology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jian Li
- Family Planning Department, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Bin Jia
- Department of Urology, the Third People's Provincial Hospital of Henan Province, Zhengzhou, Henan, China
| | - Yue Wang
- Department of Obstetrics and Gynecology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Juxin Zhang
- Department of Obstetrics and Gynecology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Guangzhi Liu
- Department of Obstetrics and Gynecology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
48
|
Yang C, Shen C, Feng T, Li H. Noncoding RNA in NK cells. J Leukoc Biol 2018; 105:63-71. [PMID: 30265761 DOI: 10.1002/jlb.1ru0518-197rr] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 09/05/2018] [Accepted: 09/09/2018] [Indexed: 12/14/2022] Open
Abstract
Noncoding RNAs (ncRNA) are important regulators that modulate cell proliferation, apoptosis, the cell cycle, and DNA methylation. NK cells mediate the immune response via the secretion of various cytokines and are important innate immune cells in the human immune system. Recent studies have found that ncRNA plays an important role in NK cell development and function. With recent advances in bioinformatics and next-generation sequencing, novel ncRNAs have been identified, allowing us to more fully appreciate its functions in NK cell biology. In this review, we summarize and discuss the latest studies on the functions and regulatory mechanisms of long noncoding RNA (lncRNA) and microRNA in NK cells from the viewpoint of epigenetic mechanisms to help us clearly understand ncRNA in NK cells.
Collapse
Affiliation(s)
- Chuan Yang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Chongyang Shen
- Basic Medicine College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ting Feng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Hong Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
49
|
Feng C, Shen JM, Lv PP, Jin M, Wang LQ, Rao JP, Feng L. Construction of implantation failure related lncRNA-mRNA network and identification of lncRNA biomarkers for predicting endometrial receptivity. Int J Biol Sci 2018; 14:1361-1377. [PMID: 30123082 PMCID: PMC6097487 DOI: 10.7150/ijbs.25081] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 07/02/2018] [Indexed: 12/12/2022] Open
Abstract
Insufficient endometrial receptivity is a major factor leading to implantation failure (IF), and the traditional way of morphological observation of endometrium cannot determine the condition of receptivity sufficiently. Considering that long-noncoding RNAs (lncRNAs) regulate endometrial receptivity and competing endogenous RNA (ceRNA) mechanism works in plenty of biological processes, ceRNA is likely to function in the pathology of IF. In the present study, we aim to construct an implantation failure related lncRNA-mRNA network (IFLMN), and to identify the key lncRNAs as the candidates for predicting endometrial receptivity. The global background network was constructed based on the presumed lncRNA-miRNA and miRNA-mRNA pairs obtained from lncRNASNP and miRTarBase. Differentially expressed genes (DEGs) of IF were calculated using the data of GSE26787, and then re-annotated as differentially expressed mRNAs (DEMs) and lncRNAs (DELs). IFLMN was constructed by hypergeometric test, including 255 lncRNA-mRNA pairs, 10 lncRNAs, and 212 mRNAs. Topological analysis determined the key lncRNAs with the highest centroid. Functional enrichment analyses were performed by unsupervised clustering, GO classification, KEGG pathway, and co-expression module analyses, achieving six key lncRNAs and their ceRNA sub-networks, which were involved in immunological activity, growth factor binding, vascular proliferation, apoptosis, and steroid biosynthesis in uterus and prepared endometrium for embryo implantation. Sixteen endometrial samples were collected during mid-luteal phase, including 8 recurrent implantation failure (RIF) or recurrent miscarriage (RM) women and 8 controls who conceived successfully. Quantitative real-time PCR was performed to compare the expression of the above six lncRNAs, which validated that the expression of all these lncRNAs was significantly elevated in endometrium of RIF/RM patients. Further studies are needed to investigate the underlying mechanism, and the lncRNAs may be developed into predictive biomarkers for endometrial receptivity.
Collapse
Affiliation(s)
- Chun Feng
- The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Jin-Ming Shen
- The First Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou, Zhejiang 310006, China
| | - Ping-Ping Lv
- The Women's Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
| | - Min Jin
- The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Li-Quan Wang
- The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Jin-Peng Rao
- The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Lei Feng
- The First Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou, Zhejiang 310006, China
| |
Collapse
|
50
|
Zhao X, Jiang Y, Wang L, Li Z, Li Q, Feng X. Advances in Understanding the Immune Imbalance between T-Lymphocyte Subsets and NK Cells in Recurrent Spontaneous Abortion. Geburtshilfe Frauenheilkd 2018; 78:677-683. [PMID: 30258242 PMCID: PMC6150770 DOI: 10.1055/a-0634-1813] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 04/16/2018] [Accepted: 05/24/2018] [Indexed: 01/07/2023] Open
Abstract
Recurrent spontaneous abortion is a global problem, and unexplained recurrent abortion triggered by immunological factors is an important focus of current research. Helper T lymphocytes (Th cells) and regulatory T lymphocytes (Treg cells) are central in human immune regulation and play a complex role in pregnancy. Natural killer cells (NK cells) exist in the endometrium and cooperate with T lymphocytes to create immune tolerance at the maternal-fetal interface, which is essential for successful pregnancy. This review has analyzed studies on Th17 cell, Treg cell and NK cell dysfunction and cellular imbalances which may contribute to unexplained recurrent spontaneous abortion to suggest a possible direction for future immunotherapies.
Collapse
Affiliation(s)
- Xiaoxuan Zhao
- Heilongjiang University of Chinese Medicine, Harbin 150040, Heilongjiang, China
| | - Yuepeng Jiang
- Heilongjiang University of Chinese Medicine, Harbin 150040, Heilongjiang, China
| | - Lin Wang
- Heilongjiang University of Chinese Medicine, Harbin 150040, Heilongjiang, China
| | - Zhihao Li
- Heilongjiang University of Chinese Medicine, Harbin 150040, Heilongjiang, China
| | - Qiang Li
- Heilongjiang University of Chinese Medicine, Harbin 150040, Heilongjiang, China
| | - Xiaoling Feng
- First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin 150040, Heilongjiang, China
| |
Collapse
|